101
|
Tahara N, Brush M, Kawakami Y. Cell migration during heart regeneration in zebrafish. Dev Dyn 2016; 245:774-87. [PMID: 27085002 PMCID: PMC5839122 DOI: 10.1002/dvdy.24411] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/17/2016] [Accepted: 04/12/2016] [Indexed: 12/27/2022] Open
Abstract
Zebrafish possess the remarkable ability to regenerate injured hearts as adults, which contrasts the very limited ability in mammals. Although very limited, mammalian hearts do in fact have measurable levels of cardiomyocyte regeneration. Therefore, elucidating mechanisms of zebrafish heart regeneration would provide information of naturally occurring regeneration to potentially apply to mammalian studies, in addition to addressing this biologically interesting phenomenon in itself. Studies over the past 13 years have identified processes and mechanisms of heart regeneration in zebrafish. After heart injury, pre-existing cardiomyocytes dedifferentiate, enter the cell cycle, and repair the injured myocardium. This process requires interaction with epicardial cells, endocardial cells, and vascular endothelial cells. Epicardial cells envelope the heart, while endocardial cells make up the inner lining of the heart. They provide paracrine signals to cardiomyocytes to regenerate the injured myocardium, which is vascularized during heart regeneration. In addition, accumulating results suggest that local migration of these major cardiac cell types have roles in heart regeneration. In this review, we summarize the characteristics of various heart injury methods used in the research community and regeneration of the major cardiac cell types. Then, we discuss local migration of these cardiac cell types and immune cells during heart regeneration. Developmental Dynamics 245:774-787, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Naoyuki Tahara
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Michael Brush
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
102
|
Saera-Vila A, Kish PE, Kahana A. Fgf regulates dedifferentiation during skeletal muscle regeneration in adult zebrafish. Cell Signal 2016; 28:1196-1204. [PMID: 27267062 DOI: 10.1016/j.cellsig.2016.06.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/22/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
Abstract
Fibroblast growth factors (Fgfs) regulate critical biological processes such as embryonic development, tissue homeostasis, wound healing, and tissue regeneration. In zebrafish, Fgf signaling plays an important role in the regeneration of the spinal cord, liver, heart, fin, and photoreceptors, although its exact mechanism of action is not fully understood. Utilizing an adult zebrafish extraocular muscle (EOM) regeneration model, we demonstrate that blocking Fgf receptor function using either a chemical inhibitor (SU5402) or a dominant-negative transgenic construct (dnFGFR1a:EGFP) impairs muscle regeneration. Adult zebrafish EOMs regenerate through a myocyte dedifferentiation process, which involves a muscle-to-mesenchyme transition and cell cycle reentry by differentiated myocytes. Blocking Fgf signaling reduced cell proliferation and active caspase 3 levels in the regenerating muscle with no detectable levels of apoptosis, supporting the hypothesis that Fgf signaling is involved in the early steps of dedifferentiation. Fgf signaling in regenerating myocytes involves the MAPK/ERK pathway: inhibition of MEK activity with U0126 mimicked the phenotype of the Fgf receptor inhibition on both muscle regeneration and cell proliferation, and activated ERK (p-ERK) was detected in injured muscles by immunofluorescence and western blot. Interestingly, following injury, ERK2 expression is specifically induced and activated by phosphorylation, suggesting a key role in muscle regeneration. We conclude that the critical early steps of myocyte dedifferentiation in EOM regeneration are dependent on Fgf signaling.
Collapse
Affiliation(s)
- Alfonso Saera-Vila
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Phillip E Kish
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Alon Kahana
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
103
|
Abstract
SIGNIFICANCE Hydrogen peroxide (H2O2) is not only a key mediator of oxidative stress but also one of the most important cellular second messengers. This small short-lived molecule is involved in the regulation of a wide range of different biological processes, including regulation of cellular signaling pathways. Studying the role of H2O2 in living systems would be challenging without modern approaches. A genetically encoded fluorescent biosensor, HyPer, is one of the most effective tools for this purpose. RECENT ADVANCES HyPer has been used by many investigators of redox signaling in various models of different scales: from cytoplasmic subcompartments and single cells to tissues of whole organisms. In many studies, the results obtained using HyPer have enabled a better understanding of the roles of H2O2 in these biological processes. However, much remains to be learned. CRITICAL ISSUES In this review, we focus on the uses of HyPer. We provide a general description of HyPer and its improved versions. Separate chapters are devoted to the results obtained by various groups who have used this biosensor for their experiments in living cells and organisms. FUTURE DIRECTIONS HyPer is an effective tool for H2O2 imaging in living systems as indicated by the increasing numbers of publications each year since its development. However, this biosensor requires further improvements. In particular, much brighter and more pH-stable versions of HyPer are necessary for imaging in mammalian tissues. Antioxid. Redox Signal. 24, 731-751.
Collapse
Affiliation(s)
- Dmitry S Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry , Moscow, Russia
| | | |
Collapse
|
104
|
Lin CY, Chiang CY, Tsai HJ. Zebrafish and Medaka: new model organisms for modern biomedical research. J Biomed Sci 2016; 23:19. [PMID: 26822757 PMCID: PMC4730764 DOI: 10.1186/s12929-016-0236-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 01/20/2016] [Indexed: 12/18/2022] Open
Abstract
Although they are primitive vertebrates, zebrafish (Danio rerio) and medaka (Oryzias latipes) have surpassed other animals as the most used model organisms based on their many advantages. Studies on gene expression patterns, regulatory cis-elements identification, and gene functions can be facilitated by using zebrafish embryos via a number of techniques, including transgenesis, in vivo transient assay, overexpression by injection of mRNAs, knockdown by injection of morpholino oligonucleotides, knockout and gene editing by CRISPR/Cas9 system and mutagenesis. In addition, transgenic lines of model fish harboring a tissue-specific reporter have become a powerful tool for the study of biological sciences, since it is possible to visualize the dynamic expression of a specific gene in the transparent embryos. In particular, some transgenic fish lines and mutants display defective phenotypes similar to those of human diseases. Therefore, a wide variety of fish model not only sheds light on the molecular mechanisms underlying disease pathogenesis in vivo but also provides a living platform for high-throughput screening of drug candidates. Interestingly, transgenic model fish lines can also be applied as biosensors to detect environmental pollutants, and even as pet fish to display beautiful fluorescent colors. Therefore, transgenic model fish possess a broad spectrum of applications in modern biomedical research, as exampled in the following review.
Collapse
Affiliation(s)
- Cheng-Yung Lin
- Graduate Institute of Biomedical Sciences, Mackay Medical College, No.46, Section 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 252, Taiwan
| | - Cheng-Yi Chiang
- Graduate Institute of Biomedical Sciences, Mackay Medical College, No.46, Section 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 252, Taiwan
| | - Huai-Jen Tsai
- Graduate Institute of Biomedical Sciences, Mackay Medical College, No.46, Section 3, Zhongzheng Rd., Sanzhi Dist., New Taipei City, 252, Taiwan.
| |
Collapse
|
105
|
Affiliation(s)
- Mo Li
- From the Gene Expression Laboratory, the Salk Institute for Biological Studies, La Jolla, CA (M.L., J.C.I.B.); and Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, Murcia, Spain (M.L.)
| | - Juan Carlos Izpisua Belmonte
- From the Gene Expression Laboratory, the Salk Institute for Biological Studies, La Jolla, CA (M.L., J.C.I.B.); and Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, Murcia, Spain (M.L.)
| |
Collapse
|
106
|
Jewhurst K, McLaughlin KA. Beyond the Mammalian Heart: Fish and Amphibians as a Model for Cardiac Repair and Regeneration. J Dev Biol 2015; 4:jdb4010001. [PMID: 29615574 PMCID: PMC5831815 DOI: 10.3390/jdb4010001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/04/2015] [Accepted: 12/17/2015] [Indexed: 12/12/2022] Open
Abstract
The epidemic of heart disease, the leading cause of death worldwide, is made worse by the fact that the adult mammalian heart is especially poor at repair. Damage to the mammal heart-such as that caused by myocardial infarction-leads to scarring, resulting in cardiac dysfunction and heart failure. In contrast, the hearts of fish and urodele amphibians are capable of complete regeneration of cardiac tissue from multiple types of damage, with full restoration of functionality. In the last decades, research has revealed a wealth of information on how these animals are able to perform this remarkable feat, and non-mammalian models of heart repair have become a burgeoning new source of data on the morphological, cellular, and molecular processes necessary to heal cardiac damage. In this review we present the major findings from recent research on the underlying mechanisms of fish and amphibian heart regeneration. We also discuss the tools and techniques that have been developed to answer these important questions.
Collapse
Affiliation(s)
- Kyle Jewhurst
- Department of Biology, Tufts University, Medford, MA 02155, USA.
| | | |
Collapse
|
107
|
Luxán G, D'Amato G, MacGrogan D, de la Pompa JL. Endocardial Notch Signaling in Cardiac Development and Disease. Circ Res 2015; 118:e1-e18. [PMID: 26635389 DOI: 10.1161/circresaha.115.305350] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/22/2015] [Indexed: 01/03/2023]
Abstract
The Notch signaling pathway is an ancient and highly conserved signaling pathway that controls cell fate specification and tissue patterning in the embryo and in the adult. Region-specific endocardial Notch activity regulates heart morphogenesis through the interaction with multiple myocardial-, epicardial-, and neural crest-derived signals. Mutations in NOTCH signaling elements cause congenital heart disease in humans and mice, demonstrating its essential role in cardiac development. Studies in model systems have provided mechanistic understanding of Notch function in cardiac development, congenital heart disease, and heart regeneration. Notch patterns the embryonic endocardium into prospective territories for valve and chamber formation, and later regulates the signaling processes leading to outflow tract and valve morphogenesis and ventricular trabeculae compaction. Alterations in NOTCH signaling in the endocardium result in congenital structural malformations that can lead to disease in the neonate and adult heart.
Collapse
Affiliation(s)
- Guillermo Luxán
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.)
| | - Gaetano D'Amato
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.)
| | - Donal MacGrogan
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.)
| | - José Luis de la Pompa
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.).
| |
Collapse
|
108
|
Seto SW, Kiat H, Lee SMY, Bensoussan A, Sun YT, Hoi MPM, Chang D. Zebrafish models of cardiovascular diseases and their applications in herbal medicine research. Eur J Pharmacol 2015; 768:77-86. [PMID: 26494630 DOI: 10.1016/j.ejphar.2015.10.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/02/2015] [Accepted: 10/16/2015] [Indexed: 01/12/2023]
Abstract
The zebrafish (Danio rerio) has recently become a powerful animal model for cardiovascular research and drug discovery due to its ease of maintenance, genetic manipulability and ability for high-throughput screening. Recent advances in imaging techniques and generation of transgenic zebrafish have greatly facilitated in vivo analysis of cellular events of cardiovascular development and pathogenesis. More importantly, recent studies have demonstrated the functional similarity of drug metabolism systems between zebrafish and humans, highlighting the clinical relevance of employing zebrafish in identifying lead compounds in Chinese herbal medicine with potential beneficial cardiovascular effects. This paper seeks to summarise the scope of zebrafish models employed in cardiovascular studies and the application of these research models in Chinese herbal medicine to date.
Collapse
Affiliation(s)
- Sai-Wang Seto
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Hosen Kiat
- Faculty of Medicine, University of New South Wales, NSW, Australia; School of Medicine, Western Sydney University, Locked Bag 1797, Penrith, NSW, Australia; Faculty of Medicine and Health Sciences, Macquarie University, NSW, Australia
| | - Simon M Y Lee
- State Key Laboratory Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Alan Bensoussan
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Yu-Ting Sun
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Maggie P M Hoi
- State Key Laboratory Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dennis Chang
- National Institute of Complementary Medicine, Western Sydney University, Campbelltown, NSW, Australia.
| |
Collapse
|
109
|
Diao J, Wang H, Chang N, Zhou XH, Zhu X, Wang J, Xiong JW. PEG–PLA nanoparticles facilitate siRNA knockdown in adult zebrafish heart. Dev Biol 2015; 406:196-202. [DOI: 10.1016/j.ydbio.2015.08.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 08/21/2015] [Accepted: 08/28/2015] [Indexed: 01/12/2023]
|
110
|
Liu HL, Zhu JG, Liu YQ, Fan ZG, Zhu C, Qian LM. Identification of the microRNA expression profile in the regenerative neonatal mouse heart by deep sequencing. Cell Biochem Biophys 2015; 70:635-42. [PMID: 24756729 DOI: 10.1007/s12013-014-9967-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that are involved in key biological processes, including development, differentiation, and regeneration. The global miRNA expression profile that regulates the regenerative potential of the neonatal mouse heart has not been reported. We performed deep sequencing to determine the genome-wide miRNA expression profile of the neonatal mouse heart at three key ages (1, 6, and 7 days). The miRNAs at least 1.4-fold differentially expressed between the three time points were selected for further analysis. Two miRNAs (mmu-miR-22-5p and mmu-miR-338-3p) were significantly upregulated, and nine miRNAs (mmu-miR-324-5p, mmu-miR-337-5p, mmu-miR-339-5p, mmu-miR-365-1-5p, mmu-miR-500-3p, mmu-miR-505-5p, mmu-miR-542-5p, mmu-miR-668-3p, and mmu-miR-92a-1-5p) were significantly downregulated in cardiac tissue of 7-day-old mice compared to 1- and 6-day-old mice. The expression patterns of five significantly different miRNAs were verified by quantitative real-time PCR. Furthermore, the potential targets of these putative miRNAs were suggested using miRNA target prediction tools. The candidate target genes are involved in the myocardial regenerative process, with a prominent role for the Notch signaling pathway. Our study provides a valuable resource for future investigation of the biological function of miRNAs in heart regeneration.
Collapse
Affiliation(s)
- H L Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | | | | | | | | | | |
Collapse
|
111
|
Regeneration, Plasticity, and Induced Molecular Programs in Adult Zebrafish Brain. BIOMED RESEARCH INTERNATIONAL 2015; 2015:769763. [PMID: 26417601 PMCID: PMC4568348 DOI: 10.1155/2015/769763] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 09/25/2014] [Accepted: 09/25/2014] [Indexed: 12/20/2022]
Abstract
Regenerative capacity of the brain is a variable trait within animals. Aquatic vertebrates such as zebrafish have widespread ability to renew their brains upon damage, while mammals have—if not none—very limited overall regenerative competence. Underlying cause of such a disparity is not fully evident; however, one of the reasons could be activation of peculiar molecular programs, which might have specific roles after injury or damage, by the organisms that regenerate. If this hypothesis is correct, then there must be genes and pathways that (a) are expressed only after injury or damage in tissues, (b) are biologically and functionally relevant to restoration of neural tissue, and (c) are not detected in regenerating organisms. Presence of such programs might circumvent the initial detrimental effects of the damage and subsequently set up the stage for tissue redevelopment to take place by modulating the plasticity of the neural stem/progenitor cells. Additionally, if transferable, those “molecular mechanisms of regeneration” could open up new avenues for regenerative therapies of humans in clinical settings. This review focuses on the recent studies addressing injury/damage-induced molecular programs in zebrafish brain, underscoring the possibility of the presence of genes that could be used as biomarkers of neural plasticity and regeneration.
Collapse
|
112
|
Bednarek D, González-Rosa JM, Guzmán-Martínez G, Gutiérrez-Gutiérrez Ó, Aguado T, Sánchez-Ferrer C, Marques IJ, Galardi-Castilla M, de Diego I, Gómez MJ, Cortés A, Zapata A, Jiménez-Borreguero LJ, Mercader N, Flores I. Telomerase Is Essential for Zebrafish Heart Regeneration. Cell Rep 2015; 12:1691-703. [PMID: 26321646 PMCID: PMC4589159 DOI: 10.1016/j.celrep.2015.07.064] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 05/27/2015] [Accepted: 07/29/2015] [Indexed: 11/05/2022] Open
Abstract
After myocardial infarction in humans, lost cardiomyocytes are replaced by an irreversible fibrotic scar. In contrast, zebrafish hearts efficiently regenerate after injury. Complete regeneration of the zebrafish heart is driven by the strong proliferation response of its cardiomyocytes to injury. Here we show that, after cardiac injury in zebrafish, telomerase becomes hyperactivated, and telomeres elongate transiently, preceding a peak of cardiomyocyte proliferation and full organ recovery. Using a telomerase-mutant zebrafish model, we found that telomerase loss drastically decreases cardiomyocyte proliferation and fibrotic tissue regression after cryoinjury and that cardiac function does not recover. The impaired cardiomyocyte proliferation response is accompanied by the absence of cardiomyocytes with long telomeres and an increased proportion of cardiomyocytes showing DNA damage and senescence characteristics. These findings demonstrate the importance of telomerase function in heart regeneration and highlight the potential of telomerase therapy as a means of stimulating cell proliferation upon myocardial infarction.
Collapse
Affiliation(s)
- Dorota Bednarek
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Juan Manuel González-Rosa
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Gabriela Guzmán-Martínez
- Cardiovascular Imaging in Humans, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Óscar Gutiérrez-Gutiérrez
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Tania Aguado
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Carlota Sánchez-Ferrer
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Inês João Marques
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - María Galardi-Castilla
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Irene de Diego
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Manuel José Gómez
- Bioinformatic Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Alfonso Cortés
- Electron Microscopy Center, Complutense University, Madrid 28040, Spain
| | - Agustín Zapata
- Electron Microscopy Center, Complutense University, Madrid 28040, Spain
| | - Luis Jesús Jiménez-Borreguero
- Cardiovascular Imaging in Humans, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Nadia Mercader
- Development of the Epicardium and Its Role during Regeneration Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| | - Ignacio Flores
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| |
Collapse
|
113
|
Hamada M, Goricki S, Byerly MS, Satoh N, Jeffery WR. Evolution of the chordate regeneration blastema: Differential gene expression and conserved role of notch signaling during siphon regeneration in the ascidian Ciona. Dev Biol 2015. [PMID: 26206613 DOI: 10.1016/j.ydbio.2015.07.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The regeneration of the oral siphon (OS) and other distal structures in the ascidian Ciona intestinalis occurs by epimorphosis involving the formation of a blastema of proliferating cells. Despite the longstanding use of Ciona as a model in molecular developmental biology, regeneration in this system has not been previously explored by molecular analysis. Here we have employed microarray analysis and quantitative real time RT-PCR to identify genes with differential expression profiles during OS regeneration. The majority of differentially expressed genes were downregulated during OS regeneration, suggesting roles in normal growth and homeostasis. However, a subset of differentially expressed genes was upregulated in the regenerating OS, suggesting functional roles during regeneration. Among the upregulated genes were key members of the Notch signaling pathway, including those encoding the delta and jagged ligands, two fringe modulators, and to a lesser extent the notch receptor. In situ hybridization showed a complementary pattern of delta1 and notch gene expression in the blastema of the regenerating OS. Chemical inhibition of the Notch signaling pathway reduced the levels of cell proliferation in the branchial sac, a stem cell niche that contributes progenitor cells to the regenerating OS, and in the OS regeneration blastema, where siphon muscle fibers eventually re-differentiate. Chemical inhibition also prevented the replacement of oral siphon pigment organs, sensory receptors rimming the entrance of the OS, and siphon muscle fibers, but had no effects on the formation of the wound epidermis. Since Notch signaling is involved in the maintenance of proliferative activity in both the Ciona and vertebrate regeneration blastema, the results suggest a conserved evolutionary role of this signaling pathway in chordate regeneration. The genes identified in this investigation provide the foundation for future molecular analysis of OS regeneration.
Collapse
Affiliation(s)
- Mayuko Hamada
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - Spela Goricki
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Mardi S Byerly
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Noriyuki Satoh
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna, Okinawa 904-0495, Japan
| | - William R Jeffery
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543, USA.
| |
Collapse
|
114
|
Abstract
The ability to repair damaged or lost tissues varies significantly among vertebrates. The regenerative ability of the heart is clinically very relevant, because adult teleost fish and amphibians can regenerate heart tissue, but we mammals cannot. Interestingly, heart regeneration is possible in neonatal mice, but this ability is lost within 7 days after birth. In zebrafish and neonatal mice, lost cardiomyocytes are regenerated via proliferation of spared, differentiated cardiomyocytes. While some cardiomyocyte turnover occurs in adult mammals, the cardiomyocyte production rate is too low in response to injury to regenerate the heart. Instead, mammalian hearts respond to injury by remodeling of spared tissue, which includes cardiomyocyte hypertrophy. Wnt/β-catenin signaling plays important roles during vertebrate heart development, and it is re-activated in response to cardiac injury. In this review, we discuss the known functions of this signaling pathway in injured hearts, its involvement in cardiac fibrosis and hypertrophy, and potential therapeutic approaches that might promote cardiac repair after injury by modifying Wnt/β-catenin signaling. Regulation of cardiac remodeling by this signaling pathway appears to vary depending on the injury model and the exact stages that have been studied. Thus, conflicting data have been published regarding a potential role of Wnt/β-catenin pathway in promotion of fibrosis and cardiomyocyte hypertrophy. In addition, the Wnt inhibitory secreted Frizzled-related proteins (sFrps) appear to have Wnt-dependent and Wnt-independent roles in the injured heart. Thus, while the exact functions of Wnt/β-catenin pathway activity in response to injury still need to be elucidated in the non-regenerating mammalian heart, but also in regenerating lower vertebrates, manipulation of the pathway is essential for creation of therapeutically useful cardiomyocytes from stem cells in culture. Hopefully, a detailed understanding of the in vivo role of Wnt/β-catenin signaling in injured mammalian and non-mammalian hearts will also contribute to the success of current efforts towards developing regenerative therapies.
Collapse
Affiliation(s)
- Gunes Ozhan
- Izmir Biomedicine and Genome Center (iBG-izmir), Dokuz Eylul University, Inciralti-Balcova, 35340 Izmir, Turkey ; Department of Medical Biology and Genetics, Dokuz Eylul University Medical School, Inciralti-Balcova, 35340 Izmir, Turkey
| | - Gilbert Weidinger
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
115
|
Wyatt C, Bartoszek EM, Yaksi E. Methods for studying the zebrafish brain: past, present and future. Eur J Neurosci 2015; 42:1746-63. [PMID: 25900095 DOI: 10.1111/ejn.12932] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 01/16/2023]
Abstract
The zebrafish (Danio rerio) is one of the most promising new model organisms. The increasing popularity of this amazing small vertebrate is evident from the exponentially growing numbers of research articles, funded projects and new discoveries associated with the use of zebrafish for studying development, brain function, human diseases and screening for new drugs. Thanks to the development of novel technologies, the range of zebrafish research is constantly expanding with new tools synergistically enhancing traditional techniques. In this review we will highlight the past and present techniques which have made, and continue to make, zebrafish an attractive model organism for various fields of biology, with a specific focus on neuroscience.
Collapse
Affiliation(s)
- Cameron Wyatt
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef, Leuven, Belgium.,VIB, Leuven, Belgium
| | - Ewelina M Bartoszek
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef, Leuven, Belgium.,VIB, Leuven, Belgium.,Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Emre Yaksi
- Neuro-Electronics Research Flanders, Imec Campus, Kapeldreef, Leuven, Belgium.,VIB, Leuven, Belgium.,KU Leuven, Leuven, Belgium.,Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
116
|
The Phosphorylation State of GSK3β Serine 9 Correlated to the Development of Valproic Acid-Associated Fetal Cardiac Teratogenicity, Fetal VPA Syndrome, Rescued by Folic Acid Administration. Cardiovasc Toxicol 2015; 16:34-45. [DOI: 10.1007/s12012-015-9316-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
117
|
Darehzereshki A, Rubin N, Gamba L, Kim J, Fraser J, Huang Y, Billings J, Mohammadzadeh R, Wood J, Warburton D, Kaartinen V, Lien CL. Differential regenerative capacity of neonatal mouse hearts after cryoinjury. Dev Biol 2015; 399:91-99. [PMID: 25555840 PMCID: PMC4339535 DOI: 10.1016/j.ydbio.2014.12.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 01/05/2023]
Abstract
Neonatal mouse hearts fully regenerate after ventricular resection similar to adult zebrafish. We established cryoinjury models to determine if different types and varying degrees of severity in cardiac injuries trigger different responses in neonatal mouse hearts. In contrast to ventricular resection, neonatal mouse hearts fail to regenerate and show severe impairment of cardiac function post transmural cryoinjury. However, neonatal hearts fully recover after non-transmural cryoinjury. Interestingly, cardiomyocyte proliferation does not significantly increase in neonatal mouse hearts after cryoinjuries. Epicardial activation and new coronary vessel formation occur after cryoinjury. The profibrotic marker PAI-1 is highly expressed after transmural but not non-transmural cryoinjuries, which may contribute to the differential scarring. Our results suggest that regenerative medicine strategies for heart injuries should vary depending on the nature of the injury.
Collapse
Affiliation(s)
- Ali Darehzereshki
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Nicole Rubin
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Laurent Gamba
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Jieun Kim
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - James Fraser
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Ying Huang
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Joshua Billings
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California
| | - Robabeh Mohammadzadeh
- Broad Center of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, CA
| | - John Wood
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David Warburton
- The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan
| | - Ching-Ling Lien
- Heart Institute, Los Angeles, California; The Saban Research Institute of Children׳s Hospital Los Angeles, Los Angeles, California; Department of Surgery, Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA.
| |
Collapse
|
118
|
Kikuchi K. Dedifferentiation, Transdifferentiation, and Proliferation: Mechanisms Underlying Cardiac Muscle Regeneration in Zebrafish. CURRENT PATHOBIOLOGY REPORTS 2015; 3:81-88. [PMID: 25722956 PMCID: PMC4333235 DOI: 10.1007/s40139-015-0063-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The adult mammalian heart is increasingly recognized as a regenerative organ with a measurable capacity to replenish cardiomyocytes throughout its lifetime, illuminating the possibility of stimulating endogenous regenerative capacity to treat heart diseases. Unlike mammals, certain vertebrates possess robust capacity for regenerating a damaged heart, providing a model to understand how regeneration could be augmented in injured human hearts. Facilitated by its rich history in the study of heart development, the teleost zebrafish Danio rerio has been established as a robust model to investigate the underlying mechanism of cardiac regeneration. This review discusses the current understanding of the endogenous mechanisms behind cardiac regeneration in zebrafish, with a particular focus on cardiomyocyte dedifferentiation, transdifferentiation, and proliferation.
Collapse
Affiliation(s)
- Kazu Kikuchi
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010 Australia
- St. Vincent’s Clinical School, University of New South Wales, Kensington, NSW 2052 Australia
| |
Collapse
|
119
|
Abstract
The latest discoveries and advanced knowledge in the fields of stem cell biology and developmental cardiology hold great promise for cardiac regenerative medicine, enabling researchers to design novel therapeutic tools and approaches to regenerate cardiac muscle for diseased hearts. However, progress in this arena has been hampered by a lack of reproducible and convincing evidence, which at best has yielded modest outcomes and is still far from clinical practice. To address current controversies and move cardiac regenerative therapeutics forward, it is crucial to gain a deeper understanding of the key cellular and molecular programs involved in human cardiogenesis and cardiac regeneration. In this review, we consider the fundamental principles that govern the "programming" and "reprogramming" of a human heart cell and discuss updated therapeutic strategies to regenerate a damaged heart.
Collapse
Affiliation(s)
- Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden Department of Medicine-Cardiology, Karolinska Institute, Stockholm, Sweden
| | - Federica Santoro
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Kenneth R Chien
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden Department of Medicine-Cardiology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
120
|
Gerbin KA, Murry CE. The winding road to regenerating the human heart. Cardiovasc Pathol 2015; 24:133-40. [PMID: 25795463 DOI: 10.1016/j.carpath.2015.02.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Regenerating the human heart is a challenge that has engaged researchers and clinicians around the globe for nearly a century. From the repair of the first septal defect in 1953, followed by the first successful heart transplant in 1967, and later to the first infusion of bone marrow-derived cells to the human myocardium in 2002, significant progress has been made in heart repair. However, chronic heart failure remains a leading pathological burden worldwide. Why has regenerating the human heart been such a challenge, and how close are we to achieving clinically relevant regeneration? Exciting progress has been made to establish cell transplantation techniques in recent years, and new preclinical studies in large animal models have shed light on the promises and challenges that lie ahead. In this review, we will discuss the history of cell therapy approaches and provide an overview of clinical trials using cell transplantation for heart regeneration. Focusing on the delivery of human stem cell-derived cardiomyocytes, current experimental strategies in the field will be discussed as well as their clinical translation potential. Although the human heart has not been regenerated yet, decades of experimental progress have guided us onto a promising path. SUMMARY Previous work in clinical cell therapy for heart repair using bone marrow mononuclear cells, mesenchymal stem cells, and cardiac-derived cells have overall demonstrated safety and modest efficacy. Recent advancements using human stem cell-derived cardiomyocytes have established them as a next generation cell type for moving forward, however certain challenges must be overcome for this technique to be successful in the clinics.
Collapse
Affiliation(s)
- Kaytlyn A Gerbin
- Department of Bioengineering, Center for Cardiovascular Biology and the Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Charles E Murry
- Department of Bioengineering, Center for Cardiovascular Biology and the Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Pathology, Center for Cardiovascular Biology and the Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA; Department of Medicine/Cardiology, Center for Cardiovascular Biology and the Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
121
|
Use of echocardiography reveals reestablishment of ventricular pumping efficiency and partial ventricular wall motion recovery upon ventricular cryoinjury in the zebrafish. PLoS One 2014; 9:e115604. [PMID: 25532015 PMCID: PMC4274112 DOI: 10.1371/journal.pone.0115604] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/28/2014] [Indexed: 11/27/2022] Open
Abstract
Aims While zebrafish embryos are amenable to in vivo imaging, allowing the study of morphogenetic processes during development, intravital imaging of adults is hampered by their small size and loss of transparency. The use of adult zebrafish as a vertebrate model of cardiac disease and regeneration is increasing at high speed. It is therefore of great importance to establish appropriate and robust methods to measure cardiac function parameters. Methods and Results Here we describe the use of 2D-echocardiography to study the fractional volume shortening and segmental wall motion of the ventricle. Our data show that 2D-echocardiography can be used to evaluate cardiac injury and also to study recovery of cardiac function. Interestingly, our results show that while global systolic function recovered following cardiac cryoinjury, ventricular wall motion was only partially restored. Conclusion Cryoinjury leads to long-lasting impairment of cardiac contraction, partially mimicking the consequences of myocardial infarction in humans. Functional assessment of heart regeneration by echocardiography allows a deeper understanding of the mechanisms of cardiac regeneration and has the advantage of being easily transferable to other cardiovascular zebrafish disease models.
Collapse
|
122
|
Aguirre A, Montserrat N, Zacchigna S, Nivet E, Hishida T, Krause MN, Kurian L, Ocampo A, Vazquez-Ferrer E, Rodriguez-Esteban C, Kumar S, Moresco JJ, Yates JR, Campistol JM, Sancho-Martinez I, Giacca M, Izpisua Belmonte JC. In vivo activation of a conserved microRNA program induces mammalian heart regeneration. Cell Stem Cell 2014; 15:589-604. [PMID: 25517466 DOI: 10.1016/j.stem.2014.10.003] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 08/19/2014] [Accepted: 10/08/2014] [Indexed: 01/14/2023]
Abstract
Heart failure is a leading cause of mortality and morbidity in the developed world, partly because mammals lack the ability to regenerate heart tissue. Whether this is due to evolutionary loss of regenerative mechanisms present in other organisms or to an inability to activate such mechanisms is currently unclear. Here we decipher mechanisms underlying heart regeneration in adult zebrafish and show that the molecular regulators of this response are conserved in mammals. We identified miR-99/100 and Let-7a/c and their protein targets smarca5 and fntb as critical regulators of cardiomyocyte dedifferentiation and heart regeneration in zebrafish. Although human and murine adult cardiomyocytes fail to elicit an endogenous regenerative response after myocardial infarction, we show that in vivo manipulation of this molecular machinery in mice results in cardiomyocyte dedifferentiation and improved heart functionality after injury. These data provide a proof of concept for identifying and activating conserved molecular programs to regenerate the damaged heart.
Collapse
Affiliation(s)
- Aitor Aguirre
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nuria Montserrat
- Center of Regenerative Medicine of Barcelona (CMRB), 08003 Barcelona, Spain
| | - Serena Zacchigna
- International Center for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | - Emmanuel Nivet
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tomoaki Hishida
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Marie N Krause
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Leo Kurian
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Alejandro Ocampo
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Eric Vazquez-Ferrer
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Sachin Kumar
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - James J Moresco
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, SR-11, La Jolla, CA 92037, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, SR-11, La Jolla, CA 92037, USA
| | - Josep M Campistol
- Renal Division, Hospital Clinic, University of Barcelona, IDIBAPS, 08036 Barcelona, Spain
| | - Ignacio Sancho-Martinez
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mauro Giacca
- International Center for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
| | | |
Collapse
|
123
|
Hadjiargyrou M, O'Keefe RJ. The convergence of fracture repair and stem cells: interplay of genes, aging, environmental factors and disease. J Bone Miner Res 2014; 29:2307-22. [PMID: 25264148 PMCID: PMC4455538 DOI: 10.1002/jbmr.2373] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 08/11/2014] [Accepted: 09/10/2014] [Indexed: 01/07/2023]
Abstract
The complexity of fracture repair makes it an ideal process for studying the interplay between the molecular, cellular, tissue, and organ level events involved in tissue regeneration. Additionally, as fracture repair recapitulates many of the processes that occur during embryonic development, investigations of fracture repair provide insights regarding skeletal embryogenesis. Specifically, inflammation, signaling, gene expression, cellular proliferation and differentiation, osteogenesis, chondrogenesis, angiogenesis, and remodeling represent the complex array of interdependent biological events that occur during fracture repair. Here we review studies of bone regeneration in genetically modified mouse models, during aging, following environmental exposure, and in the setting of disease that provide insights regarding the role of multipotent cells and their regulation during fracture repair. Complementary animal models and ongoing scientific discoveries define an increasing number of molecular and cellular targets to reduce the morbidity and complications associated with fracture repair. Last, some new and exciting areas of stem cell research such as the contribution of mitochondria function, limb regeneration signaling, and microRNA (miRNA) posttranscriptional regulation are all likely to further contribute to our understanding of fracture repair as an active branch of regenerative medicine.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | | |
Collapse
|
124
|
Keightley MC, Wang CH, Pazhakh V, Lieschke GJ. Delineating the roles of neutrophils and macrophages in zebrafish regeneration models. Int J Biochem Cell Biol 2014; 56:92-106. [DOI: 10.1016/j.biocel.2014.07.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/18/2014] [Accepted: 07/14/2014] [Indexed: 12/24/2022]
|
125
|
Felician G, Collesi C, Lusic M, Martinelli V, Ferro MD, Zentilin L, Zacchigna S, Giacca M. Epigenetic Modification at Notch Responsive Promoters Blunts Efficacy of Inducing Notch Pathway Reactivation After Myocardial Infarction. Circ Res 2014; 115:636-49. [DOI: 10.1161/circresaha.115.304517] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Giulia Felician
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| | - Chiara Collesi
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| | - Marina Lusic
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| | - Valentina Martinelli
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| | - Matteo Dal Ferro
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| | - Lorena Zentilin
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| | - Serena Zacchigna
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| | - Mauro Giacca
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy (G.F., C.C., M.L., V.M., M.D.F., L.Z., S.Z., M.G.); and Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy (C.C., M.D.F., M.G.); and Center for Translational Cardiology, Azienda Ospedaliero-Universitaria “Ospedali Riuniti di Trieste”, Trieste, Italy (C.C., M.D.F., M.G.)
| |
Collapse
|
126
|
Itou J, Akiyama R, Pehoski S, Yu X, Kawakami H, Kawakami Y. Regenerative responses after mild heart injuries for cardiomyocyte proliferation in zebrafish. Dev Dyn 2014; 243:1477-86. [PMID: 25074230 DOI: 10.1002/dvdy.24171] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The zebrafish heart regenerates after various severe injuries. Common processes of heart regeneration are cardiomyocyte proliferation, activation of epicardial tissue, and neovascularization. In order to further characterize heart regeneration processes, we introduced milder injuries and compared responses to those induced by ventricular apex resection, a widely used injury method. We used scratching of the ventricular surface and puncturing of the ventricle with a fine tungsten needle as injury-inducing techniques. RESULTS Scratching the ventricular surface induced subtle cardiomyocyte proliferation and responses of the epicardium. Endothelial cell accumulation was limited to the surface of the heart. Ventricular puncture induced cardiomyocyte proliferation, endocardial and epicardial activation, and neo-vascularization, similar to the resection method. However, the degree of the responses was milder, correlating with milder injury. Sham operation induced epicardial aldh1a2 expression but not tbx18 and WT1. CONCLUSIONS Puncturing the ventricle induces responses equivalent to resection at milder degrees in a shorter time frame and can be used as a simple injury model. Scratching the ventricle did not induce heart regeneration and can be used for studying wound responses to epicardium.
Collapse
Affiliation(s)
- Junji Itou
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | | |
Collapse
|
127
|
Hydrogen peroxide primes heart regeneration with a derepression mechanism. Cell Res 2014; 24:1091-107. [PMID: 25124925 DOI: 10.1038/cr.2014.108] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/06/2014] [Accepted: 06/09/2014] [Indexed: 12/28/2022] Open
Abstract
While the adult human heart has very limited regenerative potential, the adult zebrafish heart can fully regenerate after 20% ventricular resection. Although previous reports suggest that developmental signaling pathways such as FGF and PDGF are reused in adult heart regeneration, the underlying intracellular mechanisms remain largely unknown. Here we show that H2O2 acts as a novel epicardial and myocardial signal to prime the heart for regeneration in adult zebrafish. Live imaging of intact hearts revealed highly localized H2O2 (~30 μM) production in the epicardium and adjacent compact myocardium at the resection site. Decreasing H2O2 formation with the Duox inhibitors diphenyleneiodonium (DPI) or apocynin, or scavenging H2O2 by catalase overexpression markedly impaired cardiac regeneration while exogenous H2O2 rescued the inhibitory effects of DPI on cardiac regeneration, indicating that H2O2 is an essential and sufficient signal in this process. Mechanistically, elevated H2O2 destabilized the redox-sensitive phosphatase Dusp6 and hence increased the phosphorylation of Erk1/2. The Dusp6 inhibitor BCI achieved similar pro-regenerative effects while transgenic overexpression of dusp6 impaired cardiac regeneration. H2O2 plays a dual role in recruiting immune cells and promoting heart regeneration through two relatively independent pathways. We conclude that H2O2 potentially generated from Duox/Nox2 promotes heart regeneration in zebrafish by unleashing MAP kinase signaling through a derepression mechanism involving Dusp6.
Collapse
|
128
|
Kikuchi K. Advances in understanding the mechanism of zebrafish heart regeneration. Stem Cell Res 2014; 13:542-55. [PMID: 25127427 DOI: 10.1016/j.scr.2014.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/11/2014] [Accepted: 07/13/2014] [Indexed: 01/14/2023] Open
Abstract
The adult mammalian heart was once believed to be a post-mitotic organ without any capacity for regeneration, but recent findings have challenged this dogma. A modified view assigns the mammalian heart a measurable capacity for regeneration throughout its lifetime, with the implication that endogenous regenerative capacity can be therapeutically stimulated in the injury setting. Although extremely limited in adult mammals, the natural capacity for organ regeneration is a conserved trait in certain vertebrates. Urodele amphibians and teleosts are well-known examples of such animals that can efficiently regenerate various organs including the heart as adults. By understanding how these animals regenerate a damaged heart, one might obtain valuable insights into how regeneration can be augmented in injured human hearts. Among the regenerative vertebrate models, the teleost zebrafish, Danio rerio, is arguably the best characterized with respect to cardiac regenerative responses. Knowledge is still limited, but a decade of research in this model has led to results that may help to understand how cardiac regeneration is naturally stimulated and maintained. This review surveys recent advances in the field and discusses current understanding of the endogenous mechanisms of cardiac regeneration in zebrafish.
Collapse
Affiliation(s)
- Kazu Kikuchi
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Kensington, NSW 2052, Australia.
| |
Collapse
|
129
|
Schindler YL, Garske KM, Wang J, Firulli BA, Firulli AB, Poss KD, Yelon D. Hand2 elevates cardiomyocyte production during zebrafish heart development and regeneration. Development 2014; 141:3112-22. [PMID: 25038045 DOI: 10.1242/dev.106336] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Embryonic heart formation requires the production of an appropriate number of cardiomyocytes; likewise, cardiac regeneration following injury relies upon the recovery of lost cardiomyocytes. The basic helix-loop-helix (bHLH) transcription factor Hand2 has been implicated in promoting cardiomyocyte formation. It is unclear, however, whether Hand2 plays an instructive or permissive role during this process. Here, we find that overexpression of hand2 in the early zebrafish embryo is able to enhance cardiomyocyte production, resulting in an enlarged heart with a striking increase in the size of the outflow tract. Our evidence indicates that these increases are dependent on the interactions of Hand2 in multimeric complexes and are independent of direct DNA binding by Hand2. Proliferation assays reveal that hand2 can impact cardiomyocyte production by promoting division of late-differentiating cardiac progenitors within the second heart field. Additionally, our data suggest that hand2 can influence cardiomyocyte production by altering the patterning of the anterior lateral plate mesoderm, potentially favoring formation of the first heart field at the expense of hematopoietic and vascular lineages. The potency of hand2 during embryonic cardiogenesis suggested that hand2 could also impact cardiac regeneration in adult zebrafish; indeed, we find that overexpression of hand2 can augment the regenerative proliferation of cardiomyocytes in response to injury. Together, our studies demonstrate that hand2 can drive cardiomyocyte production in multiple contexts and through multiple mechanisms. These results contribute to our understanding of the potential origins of congenital heart disease and inform future strategies in regenerative medicine.
Collapse
Affiliation(s)
- Yocheved L Schindler
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA 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
| | - Kristina M Garske
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jinhu Wang
- Department of Cell Biology, Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Beth A Firulli
- Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Departments of Pediatrics and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anthony B Firulli
- Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Departments of Pediatrics and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kenneth D Poss
- Department of Cell Biology, Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA 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
|
130
|
Ito K, Morioka M, Kimura S, Tasaki M, Inohaya K, Kudo A. Differential reparative phenotypes between zebrafish and medaka after cardiac injury. Dev Dyn 2014; 243:1106-15. [PMID: 24947076 DOI: 10.1002/dvdy.24154] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 06/11/2014] [Accepted: 06/11/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Zebrafish have the ability for heart regeneration. However, another teleost animal model, the medaka, had not yet been investigated for this capacity. RESULTS Compared with zebrafish, the medaka heart responded differently to an injury: An excessive fibrotic response occurred in the medaka heart, and existing cardiomyocytes or cardiac progenitor cells remained dormant, resulting in no numerical difference between the uncut and injured heart with respect to the number of EdU-incorporated cardiomyocytes. The results obtained from the analysis of the medaka raldh2-GFP transgenic line showed a lack of raldh2 expression in the endocardium. Regarding periostin expression, the localization of medaka periostin-b, a marker of fibrillogenesis, in the medaka heart remained at the wound site at 30 dpa; whereas zebrafish periostin-b was no longer localized at the wound but was detected in the epicardium at that time. CONCLUSIONS Compared with zebrafish heart regeneration, the medaka heart phenotypes suggest the possibility that the medaka could hardly regenerate its heart tissue or that these phenotypes for heart regeneration showed a delay.
Collapse
Affiliation(s)
- Kohei Ito
- Department of Biological Information, Tokyo Institute of Technology, Yokohama, Japan
| | | | | | | | | | | |
Collapse
|
131
|
The modulation of cardiac progenitor cell function by hydrogel-dependent Notch1 activation. Biomaterials 2014; 35:8103-12. [PMID: 24974008 DOI: 10.1016/j.biomaterials.2014.05.082] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 05/28/2014] [Indexed: 11/24/2022]
Abstract
Myocardial infarction is the leading cause of death worldwide and phase I clinical trials utilizing cardiac progenitor cells (CPCs) have shown promising outcomes. Notch1 signaling plays a critical role in cardiac development and in the survival, cardiogenic lineage commitment, and differentiation of cardiac stem/progenitor cells. In this study, we functionalized self-assembling peptide (SAP) hydrogels with a peptide mimic of the Notch1 ligand Jagged1 (RJ) to evaluate the therapeutic benefit of CPC delivery in the hydrogels in a rat model of myocardial infarction. The behavior of CPCs cultured in the 3D hydrogels in vitro including gene expression, proliferation, and growth factor production was evaluated. Interestingly, we observed Notch1 activation to be dependent on hydrogel polymer density/stiffness with synergistic increase in presence of RJ. Our results show that RJ mediated Notch1 activation depending on hydrogel concentration differentially regulated cardiogenic gene expression, proliferation, and growth factor production in CPCs in vitro. In rats subjected to experimental myocardial infarction, improvement in acute retention and cardiac function was observed following cell therapy in RJ hydrogels compared to unmodified or scrambled peptide containing hydrogels. This study demonstrates the potential therapeutic benefit of functionalizing SAP hydrogels with RJ for CPC based cardiac repair.
Collapse
|
132
|
Grivas J, Haag M, Johnson A, Manalo T, Roell J, Das TL, Brown E, Burns AR, Lafontant PJ. Cardiac repair and regenerative potential in the goldfish (Carassius auratus) heart. Comp Biochem Physiol C Toxicol Pharmacol 2014; 163:14-23. [PMID: 24548889 PMCID: PMC4032620 DOI: 10.1016/j.cbpc.2014.02.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/29/2014] [Accepted: 02/10/2014] [Indexed: 12/20/2022]
Abstract
The remarkable ability of the heart to regenerate has been demonstrated in the zebrafish and giant danio, two fish members of the cyprinid family. Here we use light and electron microscopy to examine the repair response in the heart of another cyprinid, the goldfish (Carassius auratus), following cautery injury to a small portion of its ventricular myocardium. We observed a robust inflammatory response in the first two weeks consisting primarily of infiltrating macrophages, heterophils, and melanomacrophages. These inflammatory cells were identified in the lumen of the spongy heart, within the site of the wound, and attached to endocardial cells adjacent to the site of injury. Marked accumulation of collagen fibers and increased connective tissue were also observed during the first and second weeks in a transition zone between healthy and injured myocardium as well as in adjacent sub-epicardial regions. The accumulation of collagen and connective tissue however did not persist. The presence of capillaries was also noted in the injured area during repair. The replacement of the cauterized region of the ventricle by myocardial tissue was achieved in 6weeks. The presence of ethynyl deoxyuridine-positive cardiac myocytes and partially differentiated cardiac myocytes during repair suggest effective cardiac myocyte driven regeneration mechanisms also operate in the injured goldfish heart, and are similar to those observed in zebrafish and giant danio. Our data suggest the ability for cardiac regeneration may be widely conserved among cyprinids.
Collapse
Affiliation(s)
- Jamie Grivas
- DePauw University, Department of Biology, Greencastle, IN, USA
| | - Maria Haag
- DePauw University, Department of Biology, Greencastle, IN, USA
| | | | - Trina Manalo
- DePauw University, Department of Biology, Greencastle, IN, USA
| | - Julia Roell
- DePauw University, Department of Biology, Greencastle, IN, USA
| | - Tanmoy L. Das
- DePauw University, Department of Biology, Greencastle, IN, USA
| | - Evelyn Brown
- College of Optometry, University of Houston, Houston, TX, USA
| | - Alan R. Burns
- College of Optometry, University of Houston, Houston, TX, USA
| | - Pascal J. Lafontant
- DePauw University, Department of Biology, Greencastle, IN, USA
- Corresponding Author: Pascal J. Lafontant 1 E Hanna St, Olin 258 DePauw University Greencastle, IN 46135 Ph: (765) 721-0515 Fax: (765) 648-4766
| |
Collapse
|
133
|
The epicardium signals the way towards heart regeneration. Stem Cell Res 2014; 13:683-92. [PMID: 24933704 PMCID: PMC4241487 DOI: 10.1016/j.scr.2014.04.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/12/2014] [Accepted: 04/18/2014] [Indexed: 11/23/2022] Open
Abstract
From historical studies of developing chick hearts to recent advances in regenerative injury models, the epicardium has arisen as a key player in heart genesis and repair. The epicardium provides paracrine signals to nurture growth of the developing heart from mid-gestation, and epicardium-derived cells act as progenitors of numerous cardiac cell types. Interference with either process is terminal for heart development and embryogenesis. In adulthood, the dormant epicardium reinstates an embryonic gene programme in response to injury. Furthermore, injury-induced epicardial signalling is essential for heart regeneration in zebrafish. Given these critical roles in development, injury response and heart regeneration, the application of epicardial signals following adult heart injury could offer therapeutic strategies for the treatment of ischaemic heart disease and heart failure. The epicardium is a dynamic signalling centre during heart development and injury. Heart repair in lower vertebrates highlights the importance of epicardial signalling. Epicardial signals may be targeted to regenerate adult mammalian hearts.
Collapse
|
134
|
Xiong JW, Chang NN. Recent advances in heart regeneration. ACTA ACUST UNITED AC 2014; 99:160-9. [PMID: 24078494 DOI: 10.1002/bdrc.21039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 07/27/2013] [Accepted: 07/27/2013] [Indexed: 12/25/2022]
Abstract
Although cardiac stem cells (CSCs) and tissue engineering are very promising for cardiac regenerative medicine, studies with model organisms for heart regeneration will provide alternative therapeutic targets and opportunities. Here, we present a review on heart regeneration, with a particular focus on the most recent work in mouse and zebrafish. We attempt to summarize the recent progresses and bottlenecks of CSCs and tissue engineering for heart regeneration; and emphasize what we have learned from mouse and zebrafish regenerative models on discovering crucial genetic and epigenetic factors for stimulating heart regeneration; and speculate the potential application of these regenerative factors for heart failure. A brief perspective highlights several important and promising research directions in this exciting field.
Collapse
Affiliation(s)
- Jing-Wei Xiong
- are from Institute of Molecular Medicine, Peking University, Beijing, 100871, China and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| | | |
Collapse
|
135
|
Liu M, Zhu JG, Yu ZB, Song GX, Shen YH, Liu YQ, Zhu C, Qian LM. Identification of differentially expressed genes involved in transient regeneration of the neonatal C57BL/6J mouse heart by digital gene expression profiling. Mol Med Rep 2014; 9:2111-6. [PMID: 24699800 PMCID: PMC4055482 DOI: 10.3892/mmr.2014.2109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 03/12/2014] [Indexed: 01/01/2023] Open
Abstract
Accumulating evidence has revealed that the mammalian heart possesses a measurable capacity for renewal. Neonatal mice retain a regenerative capacity over a short time-frame (≤6 days), but this capacity is lost by 7 days of age. In the present study, differential gene expression profiling of mouse cardiac tissue was performed to further elucidate the mechanisms underlying this process. The global gene expression patterns of the neonatal C57BL/6J mouse heart were examined at three key time-points (1, 6 and 7 days old) using digital gene expression analysis. In the distribution of total clean tags, high-expression tags (>100 copies) were found to be predominant, whereas low expression tags (<5 copies) occupied the majority of distinct tag distributions. In total, 306 differentially expressed genes (DEGs) were detected in cardiac tissue, with the expression levels of 115 genes upregulated and those of 191 genes downregulated in 7-day-old mice compared with expression levels in 1- and 6-day-old mice, respectively. The expression levels of five DEGs were confirmed using quantitative polymerase chain reaction. Gene ontology analysis revealed a large proportion of DEGs distributed throughout the cell, and these DEGs were associated with binding as well as catalytic, hydrolase, transferase and molecular transducer activities. Furthermore, these genes were involved in cellular, metabolic and developmental processes, as well as biological regulation and signaling pathways. Pathway analysis identified the oxidative phosphorylation pathway to be the process most significantly putatively affected by the differential expression of these genes. These data provide the basis for future analysis of the gene expression patterns that regulate the molecular mechanism of cardiac regeneration.
Collapse
Affiliation(s)
- Ming Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jin-Gai Zhu
- Department of Pediatrics, Department of ICU, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Zhang-Bin Yu
- Department of Pediatrics, Department of ICU, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Gui-Xian Song
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ya-Hui Shen
- State Key Laboratory of Reproductive Medicine, Department of ICU, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Yao-Qiu Liu
- State Key Laboratory of Reproductive Medicine, Department of ICU, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Chun Zhu
- Department of Pediatrics, Department of ICU, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Ling-Mei Qian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
136
|
Crompton JG, Clever D, Vizcardo R, Rao M, Restifo NP. Reprogramming antitumor immunity. Trends Immunol 2014; 35:178-85. [PMID: 24661777 PMCID: PMC4373650 DOI: 10.1016/j.it.2014.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/10/2014] [Accepted: 02/13/2014] [Indexed: 01/01/2023]
Abstract
Regenerative medicine holds great promise in replacing tissues and organs lost to degenerative disease and injury. Application of the principles of cellular reprogramming for the treatment of cancer, however, is not well established. Here, we present an overview of cellular reprogramming techniques used in regenerative medicine, and within this context, envision how the scope of regenerative medicine may be expanded to treat metastatic cancer by revitalizing an exhausted and senescent immune system.
Collapse
Affiliation(s)
- Joseph G Crompton
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 2QH, UK; Department of Surgery, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - David Clever
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 2QH, UK
| | - Raul Vizcardo
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mahendra Rao
- Center for Regenerative Medicine, National Institutes of Health, NIH, Bethesda, MD 20892, USA
| | - Nicholas P Restifo
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Regenerative Medicine, National Institutes of Health, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
137
|
Zacchigna S, Giacca M. Extra- and intracellular factors regulating cardiomyocyte proliferation in postnatal life. Cardiovasc Res 2014; 102:312-20. [PMID: 24623280 DOI: 10.1093/cvr/cvu057] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
One of the striking differences that distinguish the adult from the embryonic heart in mammals and set it apart from the heart in urodeles and teleosts is the incapacity of cardiomyocytes to respond to damage by proliferation. While the molecular reasons underlying these characteristics still await elucidation, mounting evidence collected over the last several years indicates that cardiomyocyte proliferation can be modulated by different extracellular molecules. The exogenous administration of selected growth factors is capable of inducing neonatal and, in some instances, also adult cardiomyocyte proliferation. Other diffusible factors can regulate the proliferation and cardiac commitment of endogenous or implanted stem cells. While the individual role of these factors in the paracrine control of normal heart homeostasis still needs to be defined, this information is relevant for the development of novel therapeutic strategies for cardiac regeneration. In addition, recent evidence indicates that postnatal cardiomyocyte proliferation is controlled by genetically defined pathways, such as the Hippo pathway, and can be modulated by perturbing the endogenous cardiomyocyte microRNA network; the identification of the cytokines that activate these molecular circuits holds great potential for clinical translation.
Collapse
Affiliation(s)
- Serena Zacchigna
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology , Padriciano, 99, Trieste 34149, Italy
| | | |
Collapse
|
138
|
Abstract
OPINION STATEMENT Myocardial infarction is the most common cause of cardiac injury in humans and results in acute loss of large numbers of myocardial cells. Unfortunately, the mammalian heart is unable to replenish the cells that are lost following a myocardial infarction and an eventual progression to heart failure can often occur as a result. Regenerative medicine based approaches are actively being developed; however, a complete blueprint on how mammalian hearts can regenerate is still missing. Knowledge gained from studying animal models, such as zebrafish, newt, and neonatal mice, that can naturally regenerate their hearts after injury have provided an understanding of the molecular mechanisms involved in heart repair and regeneration. This research offers novel strategies to overcome the limited regenerative response observed in human patients.
Collapse
|
139
|
Notch signaling regulates cardiomyocyte proliferation during zebrafish heart regeneration. Proc Natl Acad Sci U S A 2014; 111:1403-8. [PMID: 24474765 DOI: 10.1073/pnas.1311705111] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The human heart's failure to replace ischemia-damaged myocardium with regenerated muscle contributes significantly to the worldwide morbidity and mortality associated with coronary artery disease. Remarkably, certain vertebrate species, including the zebrafish, achieve complete regeneration of amputated or injured myocardium through the proliferation of spared cardiomyocytes. Nonetheless, the genetic and cellular determinants of natural cardiac regeneration remain incompletely characterized. Here, we report that cardiac regeneration in zebrafish relies on Notch signaling. Following amputation of the zebrafish ventricular apex, Notch receptor expression becomes activated specifically in the endocardium and epicardium, but not the myocardium. Using a dominant negative approach, we discovered that suppression of Notch signaling profoundly impairs cardiac regeneration and induces scar formation at the amputation site. We ruled out defects in endocardial activation, epicardial activation, and dedifferentiation of compact myocardial cells as causative for the regenerative failure. Furthermore, coronary endothelial tubes, which we lineage traced from preexisting endothelium in wild-type hearts, formed in the wound despite the myocardial regenerative failure. Quantification of myocardial proliferation in Notch-suppressed hearts revealed a significant decrease in cycling cardiomyocytes, an observation consistent with a noncell autonomous requirement for Notch signaling in cardiomyocyte proliferation. Unexpectedly, hyperactivation of Notch signaling also suppressed cardiomyocyte proliferation and heart regeneration. Taken together, our data uncover the exquisite sensitivity of regenerative cardiomyocyte proliferation to perturbations in Notch signaling.
Collapse
|
140
|
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: 3.3] [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
|
141
|
Kimura W, Sadek HA. The cardiac hypoxic niche: emerging role of hypoxic microenvironment in cardiac progenitors. Cardiovasc Diagn Ther 2013; 2:278-89. [PMID: 24282728 DOI: 10.3978/j.issn.2223-3652.2012.12.02] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 12/10/2012] [Indexed: 12/11/2022]
Abstract
Resident stem cells persist throughout the entire lifetime of an organism where they replenishing damaged cells. Numerous types of resident stem cells are housed in a low-oxygen tension (hypoxic) microenvironment, or niches, which seem to be critical for survival and maintenance of stem cells. Recently our group has identified the adult mammalian epicardium and subepicardium as a hypoxic niche for cardiac progenitor cells. Similar to hematopoietic stem cells (LT-HSCs), progenitor cells in the hypoxic epicardial niche utilize cytoplasmic glycolysis instead of mitochondrial oxidative phosphorylation, where hypoxia inducible factor 1α (Hif-1α) maintains them in glycolytic undifferentiated state. In this review we summarize the relationship between hypoxic signaling and stem cell function, and discuss potential roles of several cardiac stem/progenitor cells in cardiac homeostasis and regeneration.
Collapse
Affiliation(s)
- Wataru Kimura
- Department of Internal Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
142
|
Poon KL, Brand T. The zebrafish model system in cardiovascular research: A tiny fish with mighty prospects. Glob Cardiol Sci Pract 2013; 2013:9-28. [PMID: 24688998 PMCID: PMC3963735 DOI: 10.5339/gcsp.2013.4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 01/29/2013] [Indexed: 12/26/2022] Open
Affiliation(s)
- Kar Lai Poon
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Hill End Road, Harefield, Middlesex, UB9 6JH, United Kingdom
| | - Thomas Brand
- Harefield Heart Science Centre, National Heart and Lung Institute, Imperial College London, Hill End Road, Harefield, Middlesex, UB9 6JH, United Kingdom
| |
Collapse
|
143
|
Affiliation(s)
- Aitor Aguirre
- From Salk Institute for Biological Studies, La Jolla, CA
| | | | | |
Collapse
|
144
|
Blum N, Begemann G. The roles of endogenous retinoid signaling in organ and appendage regeneration. Cell Mol Life Sci 2013; 70:3907-27. [PMID: 23479131 PMCID: PMC11113817 DOI: 10.1007/s00018-013-1303-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 01/29/2013] [Accepted: 02/14/2013] [Indexed: 12/20/2022]
Abstract
The ability to regenerate injured or lost body parts has been an age-old ambition of medical science. In contrast to humans, teleost fish and urodele amphibians can regrow almost any part of the body with seeming effortlessness. Retinoic acid is a molecule that has long been associated with these impressive regenerative capacities. The discovery 30 years ago that addition of retinoic acid to regenerating amphibian limbs causes "super-regeneration" initiated investigations into the presumptive roles of retinoic acid in regeneration of appendages and other organs. However, the evidence favoring or dismissing a role for endogenous retinoids in regeneration processes remained sparse and ambiguous. Now, the availability of genetic tools to manipulate and visualize the retinoic acid signaling pathway has opened up new routes to dissect its roles in regeneration. Here, we review the current understanding on endogenous functions of retinoic acid in regeneration and discuss key questions to be addressed in future research.
Collapse
Affiliation(s)
- Nicola Blum
- Developmental Biology, University of Bayreuth, 95440 Bayreuth, Germany
| | - Gerrit Begemann
- Developmental Biology, University of Bayreuth, 95440 Bayreuth, Germany
| |
Collapse
|
145
|
Lafontant PJ, Behzad AR, Brown E, Landry P, Hu N, Burns AR. Cardiac myocyte diversity and a fibroblast network in the junctional region of the zebrafish heart revealed by transmission and serial block-face scanning electron microscopy. PLoS One 2013; 8:e72388. [PMID: 24058412 PMCID: PMC3751930 DOI: 10.1371/journal.pone.0072388] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 07/09/2013] [Indexed: 01/08/2023] Open
Abstract
The zebrafish has emerged as an important model of heart development and regeneration. While the structural characteristics of the developing and adult zebrafish ventricle have been previously studied, little attention has been paid to the nature of the interface between the compact and spongy myocardium. Here we describe how these two distinct layers are structurally and functionally integrated. We demonstrate by transmission electron microscopy that this interface is complex and composed primarily of a junctional region occupied by collagen, as well as a population of fibroblasts that form a highly complex network. We also describe a continuum of uniquely flattened transitional cardiac myocytes that form a circumferential plate upon which the radially-oriented luminal trabeculae are anchored. In addition, we have uncovered within the transitional ring a subpopulation of markedly electron dense cardiac myocytes. At discrete intervals the transitional cardiac myocytes form contact bridges across the junctional space that are stabilized through localized desmosomes and fascia adherentes junctions with adjacent compact cardiac myocytes. Finally using serial block-face scanning electron microscopy, segmentation and volume reconstruction, we confirm the three-dimensional nature of the junctional region as well as the presence of the sheet-like fibroblast network. These ultrastructural studies demonstrate the previously unrecognized complexity with which the compact and spongy layers are structurally integrated, and provide a new basis for understanding development and regeneration in the zebrafish heart.
Collapse
Affiliation(s)
- Pascal J. Lafontant
- Department of Biology, DePauw University, Greencastle, Indiana, United States of America
- * E-mail:
| | - Ali R. Behzad
- Imaging and Characterization Core Lab, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Evelyn Brown
- College of Optometry, University of Houston, Houston, Texas, United States of America
| | - Paul Landry
- College of Optometry, University of Houston, Houston, Texas, United States of America
| | - Norman Hu
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Alan R. Burns
- College of Optometry, University of Houston, Houston, Texas, United States of America
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
146
|
Sun L, Yang H, Chen M, Ma D, Lin C. RNA-Seq reveals dynamic changes of gene expression in key stages of intestine regeneration in the sea cucumber Apostichopus japonicus. [corrected]. PLoS One 2013; 8:e69441. [PMID: 23936330 PMCID: PMC3735544 DOI: 10.1371/journal.pone.0069441] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 06/14/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Sea cucumbers (Holothuroidea; Echinodermata) have the capacity to regenerate lost tissues and organs. Although the histological and cytological aspects of intestine regeneration have been extensively studied, little is known of the genetic mechanisms involved. There has, however, been a renewed effort to develop a database of Expressed Sequence Tags (ESTs) in Apostichopus japonicus, an economically-important species that occurs in China. This is important for studies on genetic breeding, molecular markers and special physiological phenomena. We have also constructed a library of ESTs obtained from the regenerative body wall and intestine of A. japonicus. The database has increased to ~30000 ESTs. RESULTS We used RNA-Seq to determine gene expression profiles associated with intestinal regeneration in A. japonicus at 3, 7, 14 and 21 days post evisceration (dpe). This was compared to profiles obtained from a normally-functioning intestine. Approximately 5 million (M) reads were sequenced in every library. Over 2400 up-regulated genes (>10%) and over 1000 down-regulated genes (~5%) were observed at 3 and 7dpe (log2Ratio ≥ 1, FDR ≤ 0.001). Specific "Go terms" revealed that the DEGs (Differentially Expressed Genes) performed an important function at every regeneration stage. Besides some expected pathways (for example, Ribosome and Spliceosome pathway term), the "Notch signaling pathway," the "ECM-receptor interaction" and the "Cytokine-cytokine receptor interaction" were significantly enriched. We also investigated the expression profiles of developmental genes, ECM-associated genes and Cytoskeletal genes. Twenty of the most important differentially expressed genes (DEGs) were verified by Real-time PCR, which resulted in a trend concordance of almost 100% between the two techniques. CONCLUSION Our studies demonstrated dynamic changes in global gene expression during intestine regeneration and presented a series of candidate genes and enriched pathways that contribute to intestine regeneration in sea cucumbers. This provides a foundation for future studies on the genetics/molecular mechanisms associated with intestine regeneration.
Collapse
Affiliation(s)
- Lina Sun
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China
| | - Hongsheng Yang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China
| | - Muyan Chen
- Ocean University of China, Qingdao, PR China
| | - Deyou Ma
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Chenggang Lin
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
147
|
Faucherre A, Kissa K, Nargeot J, Mangoni ME, Jopling C. Piezo1 plays a role in erythrocyte volume homeostasis. Haematologica 2013; 99:70-5. [PMID: 23872304 DOI: 10.3324/haematol.2013.086090] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Mechanosensitivity is an inherent property of virtually all cell types, allowing them to sense and respond to physical environmental stimuli. Stretch-activated ion channels represent a class of mechanosensitive proteins which allow cells to respond rapidly to changes in membrane tension; however their identity has remained elusive. The piezo genes have recently been identified as a family of stretch-activated mechanosensitive ion channels. We set out to determine the role of piezo1 during zebrafish development. Here we report that morpholino-mediated knockdown of piezo1 impairs erythrocyte survival without affecting hematopoiesis or differentiation. Our results demonstrate that piezo1 is involved in erythrocyte volume homeostasis, disruption of which results in swelling/lysis of red blood cells and consequent anemia.
Collapse
|
148
|
Grottkau BE, Lin Y. Osteogenesis of Adipose-Derived Stem Cells. Bone Res 2013; 1:133-45. [PMID: 26273498 DOI: 10.4248/br201302003] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 04/12/2013] [Indexed: 01/01/2023] Open
Abstract
Current treatment options for skeletal repair, including immobilization, rigid fixation, alloplastic materials and bone grafts, have significant limitations. Bone tissue engineering offers a promising method for the repair of bone deficieny caused by fractures, bone loss and tumors. The use of adipose derived stem cells (ASCs) has received attention because of the self-renewal ability, high proliferative capacity and potential of osteogenic differentiation in vitro and in vivo studies of bone regeneration. Although cell therapies using ASCs are widely promising in various clinical fields, no large human clinical trials exist for bone tissue engineering. The aim of this review is to introduce how they are harvested, examine the characterization of ASCs, to review the mechanisms of osteogenic differentiation, to analyze the effect of mechanical and chemical stimuli on ASC osteodifferentiation, to summarize the current knowledge about usage of ASC in vivo studies and clinical trials, and finally to conclude with a general summary of the field and comments on its future direction.
Collapse
Affiliation(s)
- Brian E Grottkau
- Department of Orthopaedic Surgery, MassGeneral Hospital for Children and the Pediatric Orthopaedic Laboratory for Tissue Engineering and Regenerative Medicine, Harvard Medical School , Boston, Massachusetts, USA
| | - Yunfeng Lin
- Department of Orthopaedic Surgery, MassGeneral Hospital for Children and the Pediatric Orthopaedic Laboratory for Tissue Engineering and Regenerative Medicine, Harvard Medical School , Boston, Massachusetts, USA
| |
Collapse
|
149
|
Huang Y, Harrison MR, Osorio A, Kim J, Baugh A, Duan C, Sucov HM, Lien CL. Igf Signaling is Required for Cardiomyocyte Proliferation during Zebrafish Heart Development and Regeneration. PLoS One 2013; 8:e67266. [PMID: 23840646 PMCID: PMC3694143 DOI: 10.1371/journal.pone.0067266] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 05/14/2013] [Indexed: 12/26/2022] Open
Abstract
Unlike its mammalian counterpart, the adult zebrafish heart is able to fully regenerate after severe injury. One of the most important events during the regeneration process is cardiomyocyte proliferation, which results in the replacement of lost myocardium. Growth factors that induce cardiomyocyte proliferation during zebrafish heart regeneration remain to be identified. Signaling pathways important for heart development might be reutilized during heart regeneration. IGF2 was recently shown to be important for cardiomyocyte proliferation and heart growth during mid-gestation heart development in mice, although its role in heart regeneration is unknown. We found that expression of igf2b was upregulated during zebrafish heart regeneration. Following resection of the ventricle apex, igf2b expression was detected in the wound, endocardium and epicardium at a time that coincides with cardiomyocyte proliferation. Transgenic zebrafish embryos expressing a dominant negative form of Igf1 receptor (dn-Igf1r) had fewer cardiomyocytes and impaired heart development, as did embryos treated with an Igf1r inhibitor. Moreover, inhibition of Igf1r signaling blocked cardiomyocyte proliferation during heart development and regeneration. We found that Igf signaling is required for a subpopulation of cardiomyocytes marked by gata4:EGFP to contribute to the regenerating area. Our findings suggest that Igf signaling is important for heart development and myocardial regeneration in zebrafish.
Collapse
Affiliation(s)
- Ying Huang
- Heart Institute, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Craniofacial Biology Graduate Program, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Michael R. Harrison
- Heart Institute, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Arthela Osorio
- Heart Institute, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Jieun Kim
- Heart Institute, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Aaron Baugh
- Heart Institute, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Cunming Duan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Henry M. Sucov
- Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, California, United States of America
- Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, United States of America
| | - Ching-Ling Lien
- Heart Institute, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Program of Developmental Biology and Regenerative Medicine, The Saban Research Institute of Children’s Hospital Los Angeles, Los Angeles, California, United States of America
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, United States of America
- Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, United States of America
- * E-mail:
| |
Collapse
|
150
|
In vivo cardiac reprogramming contributes to zebrafish heart regeneration. Nature 2013; 498:497-501. [PMID: 23783515 PMCID: PMC4090927 DOI: 10.1038/nature12322] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 05/23/2013] [Indexed: 12/20/2022]
Abstract
Despite current treatment regimens, heart failure remains the leading cause of morbidity and mortality in the developed world due to the limited capacity of adult mammalian ventricular cardiomyocytes to divide and replace ventricular myocardium lost from ischemia-induced infarct1,2. As a result, there is great interest to identify potential cellular sources and strategies to generate new ventricular myocardium3. Past studies have shown that lower vertebrate and early postnatal mammalian ventricular cardiomyocytes can proliferate to help regenerate injured ventricles4–6; however, recent studies have suggested that additional endogenous cellular sources may contribute to this overall ventricular regeneration3. Here, we have developed in the zebrafish a combination of fluorescent reporter transgenes, genetic fate-mapping strategies, and a ventricle-specific genetic ablation system to discover that differentiated atrial cardiomyocytes can transdifferentiate into ventricular cardiomyocytes to contribute to zebrafish cardiac ventricular regeneration. Using in vivo time-lapse and confocal imaging, we monitored the dynamic cellular events during atrial-to-ventricular cardiomyocyte transdifferentiation to define intermediate cardiac reprogramming stages. Importantly, we observed that Notch signaling becomes activated in the atrial endocardium following ventricular ablation, and discovered that inhibiting Notch signaling blocked the atrial-to-ventricular transdifferentiation and cardiac regeneration. Overall, these studies not only provide evidence for the plasticity of cardiac lineages during myocardial injury, but more importantly reveal an abundant new potential cardiac resident cellular source for cardiac ventricular regeneration.
Collapse
|