51
|
Savage J, Conley AJ, Blais A, Skerjanc IS. SOX15 and SOX7 Differentially Regulate the Myogenic Program in P19 Cells. Stem Cells 2009; 27:1231-43. [DOI: 10.1002/stem.57] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
52
|
Riazi AM, Takeuchi JK, Hornberger LK, Zaidi SH, Amini F, Coles J, Bruneau BG, Van Arsdell GS. NKX2-5 regulates the expression of beta-catenin and GATA4 in ventricular myocytes. PLoS One 2009; 4:e5698. [PMID: 19479054 PMCID: PMC2684637 DOI: 10.1371/journal.pone.0005698] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 04/30/2009] [Indexed: 11/27/2022] Open
Abstract
Background The molecular pathway that controls cardiogenesis is temporally and spatially regulated by master transcriptional regulators such as NKX2-5, Isl1, MEF2C, GATA4, and β-catenin. The interplay between these factors and their downstream targets are not completely understood. Here, we studied regulation of β-catenin and GATA4 by NKX2-5 in human fetal cardiac myocytes. Methodology/Principal Findings Using antisense inhibition we disrupted the expression of NKX2-5 and studied changes in expression of cardiac-associated genes. Down-regulation of NKX2-5 resulted in increased β-catenin while GATA4 was decreased. We demonstrated that this regulation was conferred by binding of NKX2-5 to specific elements (NKEs) in the promoter region of the β-catenin and GATA4 genes. Using promoter-luciferase reporter assay combined with mutational analysis of the NKEs we demonstrated that the identified NKX2-5 binding sites were essential for the suppression of β-catenin, and upregulation of GATA4 by NKX2-5. Conclusions This study suggests that NKX2-5 modulates the β-catenin and GATA4 transcriptional activities in developing human cardiac myocytes.
Collapse
Affiliation(s)
- Ali M Riazi
- Labatt Family Heart Centre, Hospital for Sick Children, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
53
|
|
54
|
Vincentz JW, Barnes RM, Firulli BA, Conway SJ, Firulli AB. Cooperative interaction of Nkx2.5 and Mef2c transcription factors during heart development. Dev Dyn 2009; 237:3809-19. [PMID: 19035347 DOI: 10.1002/dvdy.21803] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The interactions of diverse transcription factors mediate the molecular programs that regulate mammalian heart development. Among these, Nkx2.5 and the Mef2c regulate common downstream targets and exhibit striking phenotypic similarities when disrupted, suggesting a potential interaction during heart development. Co-immunoprecipitation and mammalian two-hybrid experiments revealed a direct molecular interaction between Nkx2.5 and Mef2c. Assessment of mRNA expression verified spatiotemporal cardiac coexpression. Finally, genetic interaction studies employing histological and molecular analyses showed that, although Nkx2.5(-/-) and Mef2c(-/-) individual mutants both have identifiable ventricles, Nkx2.5(-/-);Mef2c(-/-) double mutants do not, and that mutant cardiomyocytes express only atrial and second heart field markers. Molecular marker and cell death and proliferation analyses provide evidence that ventricular hypoplasia is the result of defective ventricular cell differentiation. Collectively, these data support a hypothesis where physical, functional, and genetic interactions between Nkx2.5 and Mef2c are necessary for ventricle formation.
Collapse
Affiliation(s)
- Joshua W Vincentz
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research Division of Pediatrics Cardiology, Department of Anatomy, Indiana Medical School, Indianapolis, Indiana 46202-5225, USA
| | | | | | | | | |
Collapse
|
55
|
Mayorga M, Finan A, Penn M. Pre-transplantation specification of stem cells to cardiac lineage for regeneration of cardiac tissue. Stem Cell Rev Rep 2009; 5:51-60. [PMID: 19184567 DOI: 10.1007/s12015-009-9050-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Accepted: 01/08/2009] [Indexed: 02/07/2023]
Abstract
Myocardial infarction (MI) is a lead cause of mortality in the Western world. Treatment of acute MI is focused on restoration of antegrade flow which inhibits further tissue loss, but does not restore function to damaged tissue. Chronic therapy for injured myocardial tissue involves medical therapy that attempts to minimize pathologic remodeling of the heart. End stage therapy for chronic heart failure (CHF) involves inotropic therapy to increase surviving cardiac myocyte function or mechanical augmentation of cardiac performance. Not until the point of heart transplantation, a limited resource at best, does therapy focus on the fundamental problem of needing to replace injured tissue with new contractile tissue. In this setting, the potential for stem cell therapy has garnered significant interest for its potential to regenerate or create new contractile cardiac tissue. While to date adult stem cell therapy in clinical trials has suggested potential benefit, there is waning belief that the approaches used to date lead to regeneration of cardiac tissue. As the literature has better defined the pathways involved in cardiac differentiation, preclinical studies have suggested that stem cell pretreatment to direct stem cell differentiation prior to stem cell transplantation may be a more efficacious strategy for inducing cardiac regeneration. Here we review the available literature on pre-transplantation conditioning of stem cells in an attempt to better understand stem cell behavior and their readiness in cell-based therapy for myocardial regeneration.
Collapse
Affiliation(s)
- Maritza Mayorga
- Skirball Laboratory for Cardiovascular Cellular Therapeutics, Center for Cardiovascular Cell Therapy, Departments of Cardiovascular Medicine and Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
56
|
Chen K, Wu L, Wang ZZ. Extrinsic regulation of cardiomyocyte differentiation of embryonic stem cells. J Cell Biochem 2008; 104:119-28. [PMID: 17979183 DOI: 10.1002/jcb.21604] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease is one of leading causes of death throughout the U.S. and the world. The damage of cardiomyocytes resulting from ischemic injury is irreversible and leads to the development of progressive heart failure, which is characterized by the loss of functional cardiomyocytes. Because cardiomyocytes are unable to regenerate in the adult heart, cell-based therapy of transplantation provides a potential alternative approach to replace damaged myocardial tissue and restore cardiac function. A major roadblock toward this goal is the lack of donor cells; therefore, it is urgent to identify the cardiovascular cells that are necessary for achieving cardiac muscle regeneration. Pluripotent embryonic stem (ES) cells have enormous potential as a source of therapeutic tissues, including cardiovascular cells; however, the regulatory elements mediating ES cell differentiation to cardiomyocytes are largely unknown. In this review, we will focus on extrinsic factors that play a role in regulating different stages of cardiomyocyte differentiation of ES cells.
Collapse
Affiliation(s)
- Kang Chen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | | | | |
Collapse
|
57
|
Fathi F, Murasawa S, Hasegawa S, Asahara T, Kermani AJ, Mowla SJ. Cardiac differentiation of P19CL6 cells by oxytocin. Int J Cardiol 2008; 134:75-81. [PMID: 18538425 DOI: 10.1016/j.ijcard.2008.01.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 08/31/2007] [Accepted: 01/14/2008] [Indexed: 11/29/2022]
Abstract
BACKGROUND It has been reported that P19 embryonal carcinoma (EC) cells differentiate into beating cardiomyocytes under the action of oxytocin (OT). It has been suggested that dimethylsulfoxide (DMSO) acts via the oxytocin/oxytocin receptor pathway because an oxytocin receptor antagonist not only blocks oxytocin-induced cardiomyocyte differentiation, but also blocks DMSO-induced differentiation. In this study, the differentiation ability of OT was tested using P19CL6 cells. METHODS P19CL6 cells were cultured as a confluent monolayer and aggregated cells. OT was then added to culture media as an inducing agent. The cells treated with 1% DMSO were used as a positive control group. Differentiated cells were evaluated morphologically and immunocytochemically, as well as by RT-PCR. In addition, a stable line of green fluorescent protein (GFP)-expressing P19CL6 cells were differentiated into beating cardiomyocytes by OT. RESULTS Aggregated P19CL6 cells could be differentiated into cardiomyocytes, whereas monolayer cells could not differentiate and express specific cardiac muscle marker genes. In the control group, both aggregates and monolayer cells could be differentiated into cardiomyocytes by DMSO. In addition, GFP-expressing P19CL6 cells differentiated efficiently into beating cardiomyocytes when treated with OT. The results of all evaluations confirmed that the differentiated cells were cardiomyocytes. CONCLUSIONS We concluded that embryoid body formation (cell aggregation) is necessary for the differentiation of P19CL6 cells into cardiomyocytes when using OT as an inducer agent. Furthermore, because of the high rate of differentiation efficiency, GFP-expressing cardiomyocytes derived from P19CL6 cells have the potential to be used for regenerative therapies in experimental models.
Collapse
Affiliation(s)
- Fardin Fathi
- KDRC, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | | | | | | | | | | |
Collapse
|
58
|
Toenjes M, Schueler M, Hammer S, Pape UJ, Fischer JJ, Berger F, Vingron M, Sperling S. Prediction of cardiac transcription networks based on molecular data and complex clinical phenotypes. MOLECULAR BIOSYSTEMS 2008; 4:589-98. [PMID: 18493657 DOI: 10.1039/b800207j] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We present an integrative approach combining sophisticated techniques to construct cardiac gene regulatory networks based on correlated gene expression and optimized prediction of transcription factor binding sites. We analyze transcription levels of a comprehensive set of 42 genes in biopsies derived from hearts of a cohort of 190 patients as well as healthy individuals. To precisely describe the variety of heart malformations observed in the patients, we delineate a detailed phenotype ontology that allows description of observed clinical characteristics as well as the definition of informative meta-phenotypes. Based on the expression data obtained by real-time PCR we identify specific disease associated transcription profiles by applying linear models. Furthermore, genes that show highly correlated expression patterns are depicted. By predicting binding sites on promoter settings optimized using a cardiac specific chromatin immunoprecipitation data set, we reveal regulatory dependencies. Several of the found interactions have been previously described in literature, demonstrating that the approach is a versatile tool to predict regulatory networks.
Collapse
Affiliation(s)
- Martje Toenjes
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Ihnestr. 73, 14195 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
59
|
Liu XN, Yin Q, Zhang H, Zhang H, Zhu SJ, Wei YJ, Hu SS. Tissue extracts from infarcted myocardium of rats in promoting the differentiation of bone marrow stromal cells into cardiomyocyte-like cells. BIOMEDICAL AND ENVIRONMENTAL SCIENCES : BES 2008; 21:110-117. [PMID: 18548849 DOI: 10.1016/s0895-3988(08)60015-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
OBJECTIVE To investigate whether cardiac tissue extracts from rats could mimic the cardiac microenvironment and act as a natural inducer in promoting the differentiation of bone marrow stromal cells (BMSCs) into cardiomyocytes. METHODS Three kinds of tissue extract or cell lysate [infarcted myocardial tissue extract (IMTE), normal myocardial tissue extract (NMTE) and cultured neonatal myocardial lysate (NML)] were employed to induce BMSCs into cardiomyocyte-like cells. The cells were harvested at each time point for reverse transcription-polymerase chain reaction (RT-PCR) detection, immunocytochemical analysis, and transmission electron microscopy. RESULTS After a 7-day induction, BMSCs were enlarged and polygonal in morphology. Myofilaments, striated sarcomeres, Z-lines, and more mitochondia were observed under transmission electron microscope. Elevated expression levels of cardiac-specific genes and proteins were also confirmed by RT-PCR and immunocytochemistry. Moreover, IMTE showed a greater capacity of differentiating BMSCs into cardiomyocyte-like cells. CONCLUSIONS Cardiac tissue extracts, especially IMTE, can effectively differentiate BMSCs into cardiomyocyte-like cells.
Collapse
Affiliation(s)
- Xiao-Ning Liu
- Key Laboratory of Cardiovascular Regenerative Medicine, Ministry of Health, and Department of Cardiovascular Surgery, Cardiovascular Institute of Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100037, China
| | | | | | | | | | | | | |
Collapse
|
60
|
Nagel S, Meyer C, Quentmeier H, Kaufmann M, Drexler HG, MacLeod RAF. MEF2C is activated by multiple mechanisms in a subset of T-acute lymphoblastic leukemia cell lines. Leukemia 2007; 22:600-7. [PMID: 18079734 DOI: 10.1038/sj.leu.2405067] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In T-cell acute lymphoblastic leukemia (T-ALL) the cardiac homeobox gene NKX2-5 (at 5q35) is variously deregulated by regulatory elements coordinating with BCL11B (at 14q32.2), or the T-cell receptor gene TRD (at 14q11.2), respectively. NKX2-5 is normally expressed in developing spleen and heart, regulating fundamental processes, including differentiation and survival. In this study we investigated whether NKX2-5 expression in T-ALL cell lines reactivates these embryonal pathways contributing to leukemogenesis. Among 18 known targets analyzed, we identified three genes regulated by NKX2-5 in T-ALL cells, including myocyte enhancer factor 2C (MEF2C). Knockdown and overexpression assays confirmed MEF2C activation by NKX2-5 at both the RNA and protein levels. Direct interactions between NKX2-5 and GATA3 as indicated by co-immunoprecipitation data may contribute to MEF2C regulation. In T-ALL cell lines LOUCY and RPMI-8402 MEF2C expression was correlated with a 5q14 deletion, encompassing noncoding proximal gene regions. Fusion constructs with green fluorescent protein permitted subcellular detection of MEF2C protein in nuclear speckles interpretable as repression complexes. MEF2C consistently inhibits expression of NR4A1/NUR77, which regulates apoptosis via BCL2 transformation. Taken together, our data identify distinct mechanisms underlying ectopic MEF2C expression in T-ALL, either as a downstream target of NKX2-5, or via chromosomal aberrations deleting proximal gene regions.
Collapse
Affiliation(s)
- S Nagel
- Human and Animal Cell Cultures, DSMZ, Braunschweig, Germany.
| | | | | | | | | | | |
Collapse
|
61
|
Abstract
Cell-based therapy is emerging as an innovative approach for the treatment of degenerative diseases, and stem cells appear to be an ideal source of cells for this. In cardiology, in particular, human embryonic stem cell (hESC)-derived cardiomyocytes theoretically fulfill most, if not all, of the properties of an ideal donor cell, but several critical obstacles need to be overcome. Many research projects are focusing on set-up strategies for directing hESC differentiation toward the cardiac lineage. It is one of the main difficulties in the search to provide a valuable source of cells to effect regeneration of myocardial tissue in patients with severe heart failure. To date, there are no easy and efficient protocols for the induction of hESC differentiation toward the cardiac lineage. Discovering new molecules or tools capable of doing this is imperative.
Collapse
|
62
|
Ren X, Li Y, Ma X, Zheng L, Xu Y, Wang J. Activation of p38/MEF2C pathway by all-trans retinoic acid in cardiac myoblasts. Life Sci 2007; 81:89-96. [PMID: 17568621 DOI: 10.1016/j.lfs.2007.04.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 04/07/2007] [Accepted: 04/17/2007] [Indexed: 01/31/2023]
Abstract
Myocyte enhancer factor 2C (MEF2C) is a transcription factor particularly expressed in cardiac muscle. While the effects of all-trans retinoic acid (atRA) on embryonic heart are well described, the mechanism of atRA action on MEF2C activity in cardiomyocytes is less known. The aim of the present study was to investigate whether and how atRA regulates MEF2C activity in H9c2 rat ventricular cells. Here, our results, obtained from Western blot and protein kinase assays, showed that the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and MEF2C was induced by atRA in H9c2 myocardial cells. And the result from luciferase assays showed that the transactivation activity of MEF2C was upregulated by p38. Furthermore, using confocal microscopy and immunoprecipitation, we found that atRA hastened p38 translocation into nuclei to interact with MEF2C, and SB202190 inhibited nuclear translocation of p38. These results suggest that atRA may mediate p38/MEF2C signaling pathway during heart development.
Collapse
Affiliation(s)
- Xia Ren
- Laboratory of Development Molecular Biology, Department of Nutrition and Food Hygiene, School of Public Health, Peking University Health Science Center, Beijing 100083, PR China
| | | | | | | | | | | |
Collapse
|
63
|
Uchida S, Fuke S, Tsukahara T. Upregulations of Gata4 and oxytocin receptor are important in cardiomyocyte differentiation processes of P19CL6 cells. J Cell Biochem 2007; 100:629-41. [PMID: 16960874 DOI: 10.1002/jcb.21094] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Oxytocin induces P19 cells to differentiate into cardiomyocytes possibly through the oxytocin/oxytocin receptor system. We added oxytocin to the growth medium of P19CL6, a subline of P19, but they did not differentiate into cardiomyocytes as indicated by RT-PCR and Western blotting results. During the cardiac commitment time of P19CL6 cells, the mRNA expression levels of the oxytocin receptor were upregulated by the addition of oxytocin as well as DMSO, but an upregulation of Gata4 expression levels was only observed for the cells induced by DMSO. The in silico analysis of the upstream sequence of the oxytocin receptor predicted putative binding sites for Gata4 and Nkx2.5. These results suggest that upregulations of the oxytocin receptor and Gata4 are important for cardiomyocyte differentiation processes.
Collapse
Affiliation(s)
- Shizuka Uchida
- School of Materials Science, Japan Advanced Institute of Science and Technology, Ishikawa, Japan
| | | | | |
Collapse
|
64
|
Hosseinkhani M, Hosseinkhani H, Khademhosseini A, Bolland F, Kobayashi H, Gonzalez SP. Bone morphogenetic protein-4 enhances cardiomyocyte differentiation of cynomolgus monkey ESCs in knockout serum replacement medium. Stem Cells 2006; 25:571-80. [PMID: 17138962 DOI: 10.1634/stemcells.2006-0225] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Despite extensive research in the differentiation of rodent ESCs into cardiomyocytes, there have been few studies of this process in primates. In this study, we examined the role of bone morphogenic protein-4 (BMP-4) to induce cardiomyocyte differentiation of cynomolgus monkey ESCs. To study the role of BMP-4, EBs were formed and cultured in Knockout Serum Replacement (KSR) medium containing BMP-4 for 8 days and subsequently seeded in gelatin-coated dishes for 20 days. It was found that ESCs differentiated into cardiomyocytes upon stimulation with BMP-4 in KSR medium, which resulted in a large fraction of beating EBs ( approximately 16%) and the upregulation of cardiac-specific proteins in a dose and time-dependent manner. In contrast, the addition of BMP-4 in FBS-containing medium resulted in a lower fraction of beating EBs ( approximately 6%). BMP-4 acted principally between mesendodermal and mesoderm progenitors and subsequently enhanced their expression. Ultrastructural observation revealed that beating EBs contained mature cardiomyocytes with sarcomeric structures. In addition, immunostaining, reverse transcription-polymerase chain reaction, and Western blotting for cardiac markers confirmed the increased differentiation of cardiomyocytes in these cultures. Moreover, electrophysiological studies demonstrated that the differentiated cardiomyocytes were electrically activated. These findings may be useful in developing effective culture conditions to differentiate cynomolgus monkey ESCs into cardiomyocytes for studying developmental biology and for regenerative medicine.
Collapse
Affiliation(s)
- Mohsen Hosseinkhani
- International Center for Young Scientists, National Institute for Materials Science, Tsukuba, Ibaraki 305-0044, Japan.
| | | | | | | | | | | |
Collapse
|
65
|
Karamboulas C, Swedani A, Ward C, Al-Madhoun AS, Wilton S, Boisvenue S, Ridgeway AG, Skerjanc IS. HDAC activity regulates entry of mesoderm cells into the cardiac muscle lineage. J Cell Sci 2006; 119:4305-14. [PMID: 17038545 DOI: 10.1242/jcs.03185] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Class II histone deacetylases (HDAC4, HDAC5, HDAC7 and HDAC9) have been shown to interact with myocyte enhancer factors 2 (MEF2s) and play an important role in the repression of cardiac hypertrophy. We examined the role of HDACs during the differentiation of P19 embryonic carcinoma stem cells into cardiomyoctyes. Treatment of aggregated P19 cells with the HDAC inhibitor trichostatin A induced the entry of mesodermal cells into the cardiac muscle lineage, shown by the upregulation of transcripts Nkx2-5, MEF2C, GATA4 and cardiac α-actin. Furthermore, the overexpression of HDAC4 inhibited cardiomyogenesis, shown by the downregulation of cardiac muscle gene expression. Class II HDAC activity is inhibited through phosphorylation by Ca2+/calmodulin-dependent kinase (CaMK). Expression of an activated CaMKIV in P19 cells upregulated the expression of Nkx2-5, GATA4 and MEF2C, enhanced cardiac muscle development, and activated a MEF2-responsive promoter. Moreover, inhibition of CaMK signaling downregulated GATA4 expression. Finally, P19 cells constitutively expressing a dominant-negative form of MEF2C, capable of binding class II HDACs, underwent cardiomyogenesis more efficiently than control cells, implying the relief of an inhibitor. Our results suggest that HDAC activity regulates the specification of mesoderm cells into cardiomyoblasts by inhibiting the expression of GATA4 and Nkx2-5 in a stem cell model system.
Collapse
Affiliation(s)
- Christina Karamboulas
- Department of Biochemistry, Medical Sciences Building, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Angello JC, Kaestner S, Welikson RE, Buskin JN, Hauschka SD. BMP induction of cardiogenesis in P19 cells requires prior cell-cell interaction(s). Dev Dyn 2006; 235:2122-33. [PMID: 16773658 PMCID: PMC2572146 DOI: 10.1002/dvdy.20863] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mouse P19 embryonal carcinoma cells undergo cardiogenesis in response to high density and DMSO. We have derived a clonal subline that undergoes cardiogenesis in response to high density, but without requiring exposure to DMSO. The new subline retains the capacity to differentiate into skeletal muscle and neuronal cells in response to DMSO and retinoic acid. However, upon aggregation, these Oct 4-positive cells, termed P19-SI because they "self-induce" cardiac muscle, exhibit increased mRNAs encoding the mesodermal factor Brachyury, cardiac transcription factors Nkx 2.5 and GATA 4, the transcriptional repressor Msx-1, and cytokines Wnt 3a, Noggin, and BMP 4. Exposure of aggregated P19-SI cells to BMP 4, a known inducer of cardiogenesis, accelerates cardiogenesis, as determined by rhythmic beating and myosin staining. However, cardiogenesis is severely inhibited when P19-SI cells are aggregated in the presence of BMP 4. These results demonstrate that cell-cell interaction is required before P19-SI cells can undergo a cardiogenic response to BMP 4. A concurrent increase in the expression of Msx-1 suggests one possible process underlying the inhibition of cardiogenesis. The phenotype of P19-SI cells offers an opportunity to explore new aspects of cardiac induction.
Collapse
Affiliation(s)
- John C Angello
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
67
|
Abstract
Cardiac hypertrophy is a compensatory mechanism of the heart to maintain cardiac output under stresses that compromise cardiac function. Mechanical stretch and neurohumoral factors induce changes in intracellular signaling pathways resulting in increased protein synthesis and activation of specific genes promoting cardiac growth, eventually leading to left ventricular remodeling and cardiac dysfunction. The remodeling process results from alterations in cardiac myocytes as well as the extracellular matrix.
Collapse
Affiliation(s)
- Risto Kerkela
- Jefferson Medical College, 1025 Walnut Street, College Bldg, Room 316, Philadelphia, PA 19107, USA
| | | |
Collapse
|
68
|
Karamboulas C, Dakubo GD, Liu J, De Repentigny Y, Yutzey K, Wallace VA, Kothary R, Skerjanc IS. Disruption of MEF2 activity in cardiomyoblasts inhibits cardiomyogenesis. J Cell Sci 2006; 119:4315-21. [PMID: 17003108 DOI: 10.1242/jcs.03186] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myocyte enhancer factors (MEF2s) bind to muscle-specific promoters and activate transcription. Drosophila Mef2 is essential for Drosophila heart development, however, neither MEF2C nor MEF2B are essential for the early stages of murine cardiomyogenesis. Although Mef2c-null mice were defective in the later stages of heart morphogenesis, differentiation of cardiomyocytes still occurred. Since there are four isoforms of MEF2 factors (MEF2A, MEF2B, MEF2C and MEF2D), the ability of cells to differentiate may have been confounded by genetic redundancy. To eliminate this variable, the effect of a dominant-negative MEF2 mutant (MEF2C/EnR) during cardiomyogenesis was examined in transgenic mice and P19 cells. Targeting the expression of MEF2C/EnR to cardiomyoblasts using an Nkx2-5 enhancer in the P19 system resulted in the loss of both cardiomyocyte development and the expression of GATA4, BMP4, Nkx2-5 and MEF2C. In transiently transgenic mice, MEF2C/EnR expression resulted in embryos that lacked heart structures and exhibited defective differentiation. Our results show that MEF2C, or genes containing MEF2 DNA-binding sites, is required for the efficient differentiation of cardiomyoblasts into cardiomyocytes, suggesting conservation in the role of MEF2 from Drosophila to mammals.
Collapse
Affiliation(s)
- Christina Karamboulas
- Department of Biochemistry, Medical Sciences Building, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Gude N, Muraski J, Rubio M, Kajstura J, Schaefer E, Anversa P, Sussman MA. Akt promotes increased cardiomyocyte cycling and expansion of the cardiac progenitor cell population. Circ Res 2006; 99:381-8. [PMID: 16840722 DOI: 10.1161/01.res.0000236754.21499.1c] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activation of Akt is associated with enhanced cell cycling and cellular proliferation in nonmyocytes, but this effect of nuclear Akt accumulation has not been explored in the context of the myocardium. Cardiac-specific expression of nuclear-targeted Akt (Akt/nuc) in transgenics prolongs postnatal cell cycling as evidenced by increased numbers of Ki67+ cardiomyocytes at 2 to 3 weeks after birth. Similarly, nuclear-targeting of Akt promotes expansion of the presumptive cardiac progenitor cell population as assessed by immunolabeling for c-kit in combination with myocyte-specific markers Nkx 2.5 or MEF 2C. Increases in pro-proliferative cytokines, including tumor-necrosis superfamily 8, interleukin-17e, and hepatocyte growth factor, were found in nuclear-targeted Akt myocardial samples. Concurrent signaling mediated by paracrine factors downstream of Akt/nuc expression may be responsible for phenotypic effects of nuclear-targeted Akt in the myocardium, including enhanced cell proliferation and expansion of the stem cell population.
Collapse
Affiliation(s)
- Natalie Gude
- San Diego State University Heart Institute and Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | | | | | | | | | | | | |
Collapse
|
70
|
Ding B, Liu CJ, Huang Y, Hickey RP, Yu J, Kong W, Lengyel P. p204 Is Required for the Differentiation of P19 Murine Embryonal Carcinoma Cells to Beating Cardiac Myocytes. J Biol Chem 2006; 281:14882-92. [PMID: 16556595 DOI: 10.1074/jbc.m511747200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Among 10 adult mouse tissues tested, the p204 protein levels were highest in heart and skeletal muscle. We described previously that the MyoD-inducible p204 protein is required for the differentiation of cultured murine C2C12 skeletal muscle myoblasts to myotubes. Here we report that p204 was also required for the differentiation of cultured P19 murine embryonal carcinoma stem cells to beating cardiac myocytes. As shown by others, this process can be triggered by dimethyl sulfoxide (DMSO). We established that DMSO induced the formation of 204RNA and p204. Ectopic p204 could partially substitute for DMSO in inducing differentiation, whereas ectopic 204 antisense RNA inhibited the differentiation. Experiments with reporter constructs, including regulatory regions from the Ifi204 gene (encoding p204) in P19 cells and in cultured newborn rat cardiac myocytes, as well as chromatin coimmunoprecipitations with transcription factors, revealed that p204 expression was synergistically transactivated by the cardiac Gata4, Nkx2.5, and Tbx5 transcription factors. Furthermore, ectopic p204 triggered the expression of Gata4 and Nkx2.5 in P19 cells. p204 contains a nuclear export signal and was partially translocated to the cytoplasm during the differentiation. p204 from which the nuclear export signal was deleted was not translocated, and it did not induce differentiation. The various mechanisms by which p204 promoted the differentiation are reported in the accompanying article (Ding, B., Liu, C., Huang, Y., Yu, J., Kong, W., and Lengyel, P. (2006) J. Biol. Chem. 281, 14893-14906).
Collapse
Affiliation(s)
- Bo Ding
- Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar Street, New Haven, CT 06520-8024, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Schild L, Bukowska A, Gardemann A, Polczyk P, Keilhoff G, Täger M, Dudley SC, Klein HU, Goette A, Lendeckel U. Rapid pacing of embryoid bodies impairs mitochondrial ATP synthesis by a calcium-dependent mechanism--a model of in vitro differentiated cardiomyocytes to study molecular effects of tachycardia. Biochim Biophys Acta Mol Basis Dis 2006; 1762:608-15. [PMID: 16644187 PMCID: PMC3153943 DOI: 10.1016/j.bbadis.2006.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 03/17/2006] [Accepted: 03/20/2006] [Indexed: 11/26/2022]
Abstract
Tachycardia may cause substantial molecular and ultrastructural alterations in cardiac tissue. The underlying pathophysiology has not been fully explored. The purpose of this study was (I) to validate a three-dimensional in vitro pacing model, (II) to examine the effect of rapid pacing on mitochondrial function in intact cells, and (III) to evaluate the involvement of L-type-channel-mediated calcium influx in alterations of mitochondria in cardiomyocytes during rapid pacing. In vitro differentiated cardiomyocytes from P19 cells that formed embryoid bodies were paced for 24 h with 0.6 and 2.0 Hz. Pacing at 2.0 Hz increased mRNA expression and phosphorylation of ERK1/2 and caused cellular hypertrophy, indicated by increased protein/DNA ratio, and oxidative stress measured as loss of cellular thiols. Rapid pacing additionally provoked structural alterations of mitochondria. All these changes are known to occur in vivo during atrial fibrillation. The structural alterations of mitochondria were accompanied by limitation of ATP production as evidenced by decreased endogenous respiration in combination with decreased ATP levels in intact cells. Inhibition of calcium inward current with verapamil protected against hypertrophic response and oxidative stress. Verapamil ameliorated morphological changes and dysfunction of mitochondria. In conclusion, rapid pacing-dependent changes in calcium inward current via L-type channels mediate both oxidative stress and mitochondrial dysfunction. The in vitro pacing model presented here reflects changes occurring during tachycardia and, thus, allows functional analyses of the signaling pathways involved.
Collapse
Affiliation(s)
- Lorenz Schild
- Institute of Clinical Chemistry, Department of Pathobiochemistry, Otto-von-Guericke University Magdeburg, Germany
| | - Alicja Bukowska
- Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Germany
| | - Andreas Gardemann
- Institute of Clinical Chemistry, Department of Pathobiochemistry, Otto-von-Guericke University Magdeburg, Germany
| | - Pamela Polczyk
- Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Germany
- Division of Cardiology, University Hospital Magdeburg, Leipzigerstr. 44 39120 Magdeburg, Germany
| | - Gerburg Keilhoff
- Institute of Medical Neurobiology, Otto-von-Guericke University Magdeburg, Germany
| | | | - Samuel C. Dudley
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Helmut U. Klein
- Division of Cardiology, University Hospital Magdeburg, Leipzigerstr. 44 39120 Magdeburg, Germany
| | - Andreas Goette
- Division of Cardiology, University Hospital Magdeburg, Leipzigerstr. 44 39120 Magdeburg, Germany
- Corresponding author. Tel.: +49 391 6713203; fax: +49 391 673202. (A. Goette)
| | - Uwe Lendeckel
- Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Germany
| |
Collapse
|
72
|
Ding B, Liu CJ, Huang Y, Yu J, Kong W, Lengyel P. p204 protein overcomes the inhibition of the differentiation of P19 murine embryonal carcinoma cells to beating cardiac myocytes by Id proteins. J Biol Chem 2006; 281:14893-906. [PMID: 16556596 DOI: 10.1074/jbc.m511748200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We reported in the accompanying article (Ding, B., Liu, C., Huang, Y., Hickey, R. P., Yu, J., Kong, W., and Lengyel, P. (2006) J. Biol. Chem. 281, 14882-14892) that (i) the p204 protein is required for the differentiation of murine P19 embryonal carcinoma stem cells to beating cardiac myocytes, and (ii) the expression of p204 in the differentiating P19 cells is synergistically transactivated by the cardiac transcription factors Gata4, Nkx2.5, and Tbx5. Here we report that endogenous or ectopic inhibitor of differentiation (Id) proteins inhibited the differentiation of P19 cells to myocytes. This was in consequence of the binding of Id1, Id2, or Id3 protein to the Gata4 and Nkx2.5 proteins and the resulting inhibitions (i) of the binding of these transcription factors to each other and to DNA and (ii) of their synergistic transactivation of the expression of various genes, including atrial natriuretic factor and Ifi204 (encoding p204). p204 overcame this inhibition by Id proteins in consequence of (i) binding and sequestering Id proteins, (ii) accelerating their ubiquitination and degradation by proteasomes, and (iii) decreasing the level of Id proteins in the nucleus by increasing their translocation from the nucleus to the cytoplasm. Points (ii) and (iii) depended on the presence of the nuclear export signal in p204. In the course of the differentiation, Gata4, Nkx2.5, and p204 were components of a positive feedback loop. This loop arose in consequence of it that p204 overcame the inhibition of the synergistic activity of Gata4 and Nkx2.5 by the Id proteins.
Collapse
Affiliation(s)
- Bo Ding
- Department of Molecular Biophysics and Biochemistry, Yale University, 333 Cedar Street, New Haven, CT 06520-8024, USA
| | | | | | | | | | | |
Collapse
|
73
|
Lev S, Kehat I, Gepstein L. Differentiation Pathways in Human Embryonic Stem Cell-Derived Cardiomyocytes. Ann N Y Acad Sci 2006; 1047:50-65. [PMID: 16093484 DOI: 10.1196/annals.1341.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Human embryonic stem (hES) cells are pluripotent cell lines derived from the inner cell mass of the blastocyst-stage embryo. These unique cell lines can be propagated in the undifferentiated state in culture, while retaining the capacity to differentiate into derivatives of all three germ layers, including cardiomyocytes. The derivation of the hES cell lines presents a powerful tool to explore the early events of cardiac progenitor cell specification and differentiation, and it also provides a novel cell source for the emerging field of cardiovascular regenerative medicine. A spontaneous differentiation system of these stem cells to cardiomyocytes was established and the generated myocytes displayed molecular, structural, and functional properties of early-stage heart cells. In order to follow the in vitro differentiation process, the temporal expression of signaling molecules and transcription factors governing early cardiac differentiation was examined throughout the process. A characteristic pattern was noted recapitulating the normal in vivo cardiac differentiation scheme observed in other model systems. This review discusses the known pathways involved in cardiac specification and the possible factors that may be used to enhance cardiac differentiation of hES cells, as well as the steps required to fully harness the enormous potential of these unique cells.
Collapse
Affiliation(s)
- Sophie Lev
- The B. Rappaport Institute in the Medical Sciences, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | | | | |
Collapse
|
74
|
Bettiol E, Clement S, Krause KH, Jaconi ME. Embryonic and adult stem cell-derived cardiomyocytes: lessons from in vitro models. Rev Physiol Biochem Pharmacol 2006; 157:1-30. [PMID: 17236648 DOI: 10.1007/112_0508] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For years, research has focused on how to treat heart failure by sustaining the overloaded remaining cardiomyocytes. Recently, the concept of cell replacement therapy as a treatment of heart diseases has opened a new area of investigation. In vitro-generated cardiomyocytes could be injected into the heart to rescue the function of a damaged myocardium. Embryonic and/or adult stem cells could provide cardiac cells for this purpose. Knowledge of fundamental cardiac differentiation mechanisms unraveled by studies on animal models has been improved using in vitro models of cardiogenesis such as mouse embryonal carcinoma cells, mouse embryonic stem cells and, recently, human embryonic stem cells. On the other hand, studies suggesting the existence of cardiac stem cells and the potential of adult stem cells from bone marrow or skeletal muscle to differentiate toward unexpected phenotypes raise hope and questions about their potential use for cardiac cell therapy. In this review, we compare the specificities of embryonic vs adult stem cell populations regarding their cardiac differentiation potential, and we give an overview of what in vitro models have taught us about cardiogenesis.
Collapse
Affiliation(s)
- E Bettiol
- University of Geneva, Department of Pathology and Immunology, Faculty of Medicine, Switzerland
| | | | | | | |
Collapse
|
75
|
Balza RO, Misra RP. Role of the serum response factor in regulating contractile apparatus gene expression and sarcomeric integrity in cardiomyocytes. J Biol Chem 2005; 281:6498-510. [PMID: 16368687 DOI: 10.1074/jbc.m509487200] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The serum response factor (SRF) is a transcriptional regulator required for mesodermal development, including heart formation and function. Previous studies have described the role of SRF in controlling expression of structural genes involved in conferring the myogenic phenotype. Recent studies by us and others have demonstrated embryonic lethal cardiovascular phenotypes in SRF-null animals, but have not directly addressed the mechanistic role of SRF in controlling broad regulatory programs in cardiac cells. In this study, we used a loss-of-function approach to delineate the role of SRF in cardiomyocyte gene expression and function. In SRF-null neonatal cardiomyocytes, we observed severe defects in the contractile apparatus, including Z-disc and stress fiber formation, as well as mislocalization and/or attenuation of sarcomeric proteins. Consistent with this, gene array and reverse transcription-PCR analyses showed down-regulation of genes encoding key cardiac transcriptional regulatory factors and proteins required for the maintenance of sarcomeric structure, function, and regulation. Chromatin immunoprecipitation analysis revealed that at least a subset of these proteins are likely regulated directly by SRF. The results presented here indicate that SRF is an essential coordinator of cardiomyocyte function due to its ability to regulate expression of numerous genes (some previously identified and at least 28 targets newly identified in this study) that are involved in multiple and disparate levels of sarcomeric function and assembly.
Collapse
Affiliation(s)
- Robert O Balza
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | |
Collapse
|
76
|
Singla DK, Sobel BE. Enhancement by growth factors of cardiac myocyte differentiation from embryonic stem cells: A promising foundation for cardiac regeneration. Biochem Biophys Res Commun 2005; 335:637-42. [PMID: 16004962 DOI: 10.1016/j.bbrc.2005.06.105] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Accepted: 06/20/2005] [Indexed: 11/22/2022]
Abstract
Cell transplantation is a promising, still novel, potentially therapeutic approach for the treatment of heart diseases. Clinical applications require generation of large number of donor cells. Embryonic stem (ES) cells are capable of self-renewal apparently in an unlimited fashion, in vitro. Theoretically, they can differentiate into any cell type required for cell transplantation, including cardiac myocytes. Diverse growth factors have been implicated in programming diverse cellular processes, including development of the embryonic heart, ES cell self-renewal, and cardiac myocyte differentiation from ES cells. This review addresses the current understanding of the role of growth factors in the differentiation of cardiac myocytes from ES-embryoid body cell systems in vitro as well as cardiac regeneration in vivo.
Collapse
Affiliation(s)
- Dinender K Singla
- Department of Medicine, Vascular Biology Unit, and Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446, USA.
| | | |
Collapse
|
77
|
Kumar D, Kamp TJ, LeWinter MM. Embryonic stem cells: differentiation into cardiomyocytes and potential for heart repair and regeneration. Coron Artery Dis 2005; 16:111-6. [PMID: 15735404 DOI: 10.1097/00019501-200503000-00006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Many forms of heart disease are associated with the loss of cardiomyocytes both via apoptosis or necrosis, and despite the recent identification of resident cardiac stem cells, the native capacity for renewal and repair is inadequate. Cell transplantation strategies have emerged as a potential therapeutic approach for repairing injured myocardium. Many different cell types including embryonic stem cells have been transplanted in myocardial infarction (MI) models with resulting improvement in myocardial function. Here, we review the current state of knowledge with regard to the potential of embryonic stem (ES) cells to differentiate into cardiomyocytes in the embryonic stem cell derived-embryoid body (EB) in vitro system as well as for myocardial regeneration following myocardial infarction.
Collapse
Affiliation(s)
- Dinender Kumar
- Department of Medicine, University of Vermont, Burlington, VT, 05446, USA.
| | | | | |
Collapse
|
78
|
Liu H, Harris TM, Kim HH, Childs G. Cardiac myocyte differentiation: the Nkx2.5 and Cripto target genes in P19 clone 6 cells. Funct Integr Genomics 2005; 5:218-39. [PMID: 15806425 DOI: 10.1007/s10142-005-0140-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Revised: 01/07/2005] [Accepted: 01/12/2005] [Indexed: 01/29/2023]
Abstract
Genetic evidence has implicated several genes as being critical for the development of cardiomyocytes. Whereas a few of the targets of these genes and the pathways they constitute are known the majority of targets and the interrelationships of the pathways involved still remains largely unknown. The power of high-throughput analytical techniques like microarrays and real-time RT-PCR combined with the ability to selectively silence specific mRNA in model tissue culture systems can begin to fill in these gaps and increase our understanding of the molecular mechanisms of cell commitment and terminal differentiation. We have used microarray analysis and siRNA directed against the cardiac-specific transcription factor Nkx2.5 and one of its targets Cripto in P19 clone 6 (P19Cl6) cells to identify potential targets for these genes. We demonstrate Nkx2.5 affects genes that have been shown to be controlled by the canonical Wnt or TGFbeta/BMP signaling pathways. We also show that Cripto can regulate the critical stem cell gene Nanog and two Oct 4-regulated genes: Dppa2 and 4. Cripto also affects the formation of nitric oxide, a small signaling molecule that has been reported to be important for growth and development of cardiac and smooth muscle. It affects the nitric oxide system by regulating genes that control the levels of nitric oxide synthase mRNA concentration as well as the activation and bioavailability of the protein.
Collapse
Affiliation(s)
- Hailing Liu
- Department of Molecular Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
79
|
Abstract
Sonic Hedgehog (Shh) is a critical signaling factor for a variety of developmental pathways during embryogenesis, including the specification of left-right asymmetry in the heart. Mice that lack Hedgehog signaling show a delay in the induction of cardiomyogenesis, as indicated by a delayed expression of Nkx2-5. To further examine a role for Shh in cardiomyogenesis, clonal populations of P19 cells that stably express Shh, termed P19(Shh) cells, were isolated. In monolayer P19(Shh) cultures the Shh pathway was functional as shown by the up-regulation of Ptc1 and Gli1 expression, but no cardiac muscle markers were activated. However, Shh expression induced cardiomyogenesis following cellular aggregation, resulting in the expression of factors expressed in cardiac muscle including GATA-4, MEF2C, and Nkx2-5. Furthermore, aggregated P19 cell lines expressing Gli2 or Meox1 also up-regulated the expression of cardiac muscle factors, leading to cardiomyogenesis. Meox1 up-regulated the expression of Gli1 and Gli2 and, thus, can modify the Shh signaling pathway. Finally, Shh, Gli2, and Meox1 all up-regulated BMP-4 expression, implying that activation of the Hedgehog pathway can regulate bone morphogenetic protein signals. Taken together, we propose a model in which Shh, functioning via Gli1/2, can specify mesodermal cells into the cardiac muscle lineage.
Collapse
Affiliation(s)
- Peter J Gianakopoulos
- Department of Biochemistry, Medical Sciences Building, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | |
Collapse
|
80
|
Riazi AM, Lee H, Hsu C, Van Arsdell G. CSX/Nkx2.5 modulates differentiation of skeletal myoblasts and promotes differentiation into neuronal cells in vitro. J Biol Chem 2005; 280:10716-20. [PMID: 15653675 DOI: 10.1074/jbc.m500028200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CSX/Nkx2.5 transcription factor plays a pivotal role in cardiac development; however, its role in development and differentiation of other organs has not been investigated. In this study, we used C2C12 myoblasts and human fetal primary myoblasts to investigate the function of Nkx2.5 in skeletal myogenesis. The expression levels of Nkx2.5 decreased as C2C12 myoblasts elongated and fused to form myotubes. The expression of human NKX2.5 in C2C12 myoblasts inhibited myocyte differentiation and myotube formation, and up-regulated Gata4 and Tbx5 expression. The expression of NKX2.5 in terminally differentiated C2C12 myotubes resulted in a change in morphology and breakdown into smaller myotubes. Furthermore, overexpression of NKX2.5 in C2C12 cells and primary cultures of human fetal myoblasts led to differentiation of myoblasts into neuron-like cells and expression of neuronal markers. This study sheds light on the previously unknown non-cardiac functions of Nkx2.5 transcription factor.
Collapse
Affiliation(s)
- Ali M Riazi
- Division of Cardiovascular Surgery Research Rm. 7017, McMaster Bldg., The Hospital for Sick Children, 555 University Ave., Toronto, M5G 1X8 Ontario, Canada.
| | | | | | | |
Collapse
|
81
|
Vanpoucke G, Goossens S, De Craene B, Gilbert B, van Roy F, Berx G. GATA-4 and MEF2C transcription factors control the tissue-specific expression of the alphaT-catenin gene CTNNA3. Nucleic Acids Res 2004; 32:4155-65. [PMID: 15302915 PMCID: PMC514362 DOI: 10.1093/nar/gkh727] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AlphaT-catenin is a recently identified member of the alpha-catenin family of cell-cell adhesion molecules. Its expression is restricted mainly to cardiomyocytes, although it is also expressed in skeletal muscle, testis and brain. Like other alpha-catenins, alphaT-catenin provides an indispensable link between a cadherin-based adhesion complex and the actin cytoskeleton, resulting in strong cell-cell adhesion. We show here that the tissue-specificity of alphaT-catenin expression is controlled by its promoter region. By in silico analysis, we found that the alphaT-catenin promoter contains several binding sites for cardiac and muscle-specific transcription factors. By co-transfection studies in P19 embryonal carcinoma cells, we demonstrated that MEF2C and GATA-4 each have an activating effect on the alphaT-catenin promoter. Transfections with wild-type and mutant promoter constructs in cardiac HL-1 cells indicated that one GATA box is absolutely required for high alphaT-catenin promoter activity in these cells. Furthermore, we showed that the GATA-4 transcription factor specifically binds and activates the alphaT-catenin promoter in vivo in cardiac HL-1 cells. In vivo promoter analysis in transgenic mice revealed that the isolated alphaT-catenin promoter region could direct the tissue-specific expression of a LacZ reporter gene in concordance with endogenous alphaT-catenin expression.
Collapse
Affiliation(s)
- Griet Vanpoucke
- Unit of Molecular and Cellular Oncology, Department for Molecular Biomedical Research, VIB-Ghent University, B-9052 Ghent-Zwijnaarde, Belgium
| | | | | | | | | | | |
Collapse
|
82
|
Petropoulos H, Gianakopoulos PJ, Ridgeway AG, Skerjanc IS. Disruption of Meox or Gli activity ablates skeletal myogenesis in P19 cells. J Biol Chem 2004; 279:23874-81. [PMID: 15039437 DOI: 10.1074/jbc.m312612200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gli2 and Meox1 are transcription factors that are expressed in the developing somite and play roles in the commitment of cells to the skeletal muscle lineage. To further define their roles in regulating myogenesis, the function of wild type and dominant-negative forms of Gli2 and Meox1 were examined in the context of differentiating P19 stem cells. We found that Gli2 overexpression up-regulated transcript levels of Meox1 and, conversely, Meox1 overexpression resulted in the upregulation of Gli2 transcripts. Furthermore, dominant-negative forms of either Meox1 or Gli2 disrupted the ability of P19 cells to commit to the muscle lineage and to properly express either Gli2 or Meox1, respectively. Finally, Pax3 transcripts were induced by Gli2 overexpression and lost in the presence of either mutants Meox1 or Gli2. Taken together, these results support the existence of a regulatory loop between Gli2, Meox1, and Pax3 that is essential for specification of mesodermal cells into the muscle lineage.
Collapse
MESH Headings
- Animals
- Binding Sites
- Blotting, Northern
- Cell Differentiation
- Cell Line
- Cell Line, Tumor
- Cell Lineage
- Cells, Cultured
- DNA, Complementary/metabolism
- DNA-Binding Proteins/metabolism
- Down-Regulation
- Genes, Dominant
- Homeodomain Proteins/genetics
- Homeodomain Proteins/physiology
- Kruppel-Like Transcription Factors
- Mesoderm/metabolism
- Mice
- Mice, Inbred C3H
- Microscopy, Fluorescence
- Models, Biological
- Muscle Proteins/metabolism
- Muscle, Skeletal/embryology
- Muscle, Skeletal/metabolism
- Muscles/metabolism
- Mutation
- Myogenic Regulatory Factor 5
- Myogenin/metabolism
- PAX3 Transcription Factor
- Paired Box Transcription Factors
- Phenotype
- Plasmids/metabolism
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Time Factors
- Trans-Activators
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/physiology
- Transcription, Genetic
- Transfection
- Up-Regulation
- Zinc Finger Protein Gli2
Collapse
Affiliation(s)
- Helen Petropoulos
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | | | |
Collapse
|
83
|
Matsuura K, Nagai T, Nishigaki N, Oyama T, Nishi J, Wada H, Sano M, Toko H, Akazawa H, Sato T, Nakaya H, Kasanuki H, Komuro I. Adult cardiac Sca-1-positive cells differentiate into beating cardiomyocytes. J Biol Chem 2003; 279:11384-91. [PMID: 14702342 DOI: 10.1074/jbc.m310822200] [Citation(s) in RCA: 451] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although somatic stem cells have been reported to exist in various adult organs, there have been few reports concerning stem cells in the heart. We here demonstrate that Sca-1-positive (Sca-1+) cells in adult hearts have some of the features of stem cells. Sca-1+ cells were isolated from adult murine hearts by a magnetic cell sorting system and cultured on gelatin-coated dishes. A fraction of Sca-1+ cells stuck to the culture dish and proliferated slowly. When treated with oxytocin, Sca-1+ cells expressed genes of cardiac transcription factors and contractile proteins and showed sarcomeric structure and spontaneous beating. Isoproterenol treatment increased the beating rate, which was accompanied by the intracellular Ca(2+) transients. The cardiac Sca-1+ cells expressed oxytocin receptor mRNA, and the expression was up-regulated after oxytocin treatment. Some of the Sca-1+ cells expressed alkaline phosphatase after osteogenic induction and were stained with Oil-Red O after adipogenic induction. These results suggest that Sca-1+ cells in the adult murine heart have potential as stem cells and may contribute to the regeneration of injured hearts.
Collapse
Affiliation(s)
- Katsuhisa Matsuura
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Fijnvandraat AC, Lekanne Deprez RH, Christoffels VM, Ruijter JM, Moorman AFM. TBX5 overexpression stimulates differentiation of chamber myocardium in P19C16 embryonic carcinoma cells. J Muscle Res Cell Motil 2003; 24:211-8. [PMID: 14609032 DOI: 10.1023/a:1026063409656] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In vitro differentiation of pluripotent embryonic cells is becoming a model system to study factors and genes involved in early developmental processes including cardiogenesis. An additional application involves the development of donor cells for treatment of diseases among which cardiac infarction. For this purpose differentiated cells should meet the functional characteristics of chamber myocardium, a requirement not convincingly reached as yet. The T-box transcription factor Tbx5 has been demonstrated to be crucial for heart formation. Using stably transfected clones of the P19C16 embryonic carcinoma cell line, reported to differentiate efficiently into the cardiac lineage, we investigated whether Tbx5 is sufficient to enhance cardiogenesis and differentiation of chamber myocardium. TBX5-transfected clones started to beat earlier, however, a relation between transgenic TBX5 mRNA levels and the number of beating foci or levels of Serca2a mRNA, a myocardial marker, could not be observed. However, TBX5-transfected clones displayed significantly higher levels of atrial natriuretic factor (Anf) and Connexin (Cx)40 mRNAs, which are associated with the formation of chamber myocardium. This indicates that Tbx5 enhances cardiac maturation within this system rather than cardiogenesis.
Collapse
Affiliation(s)
- Arnoud C Fijnvandraat
- Experimental and Molecular Cardiology Group, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
85
|
Firulli AB, Thattaliyath BD. Transcription factors in cardiogenesis: the combinations that unlock the mysteries of the heart. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 214:1-62. [PMID: 11893163 DOI: 10.1016/s0074-7696(02)14002-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Heart formation is one of the first signs of organogenesis within the developing embryo and this process is conserved from flies to man. Completing the genetic roadmap of the molecular mechanisms that control the cell specification and differentiation of cells that form the developing heart has been an exciting and fast-moving area of research in the fields of molecular and developmental biology. At the core of these studies is an interest in the transcription factors that are responsible for initiation of a pluripotent cell to become programmed to the cardiac lineage and the subsequent transcription factors that implement the instructions set up by the cells commitment decision. To gain a better understanding of these pathways, cardiac-expressed transcription factors have been identified, cloned, overexpressed, and mutated to try to determine function. Although results vary depending on the gene in question, it is clear that there is a striking evolutionary conservation of the cardiogenic program among species. As we move up the evolutionary ladder toward man, we encounter cases of functional redundancy and combinatorial interactions that reflect the complex networks of gene expression that orchestrate heart development. This review focuses on what is known about the transcription factors implicated in heart formation and the role they play in this intricate genetic program.
Collapse
Affiliation(s)
- Anthony B Firulli
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio 78229, USA
| | | |
Collapse
|
86
|
Abstract
Development of the heart is a complex process involving primary and secondary heart fields that are set aside to generate myocardial and endocardial cell lineages. The molecular inductions that occur in the primary heart field appear to be recapitulated in induction and myocardial differentiation of the secondary heart field, which adds the conotruncal segments to the primary heart tube. While much is now known about the initial steps and factors involved in induction of myocardial differentiation, little is known about induction of endocardial development. Many of the genes expressed by nascent myocardial cells, which then become committed to a specific heart segment, have been identified and studied. In addition to the heart fields, several other "extracardiac" cell populations contribute to the fully functional mature heart. Less is known about the genetic programs of extracardiac cells as they enter the heart and take part in cardiogenesis. The molecular/genetic basis of many congenital cardiac defects has been elucidated in recent years as a result of new insights into the molecular control of developmental events.
Collapse
Affiliation(s)
- Margaret L Kirby
- Department of Pediatrics, Division of Neonatology, Duke University Medical Center, Box 3179, Durham, NC 27710, USA.
| |
Collapse
|
87
|
Rogerson PJ, Jamali M, Skerjanc IS. The C-terminus of myogenin, but not MyoD, targets upregulation of MEF2C expression. FEBS Lett 2002; 524:134-8. [PMID: 12135755 DOI: 10.1016/s0014-5793(02)03024-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The myogenic regulatory family of basic helix-loop-helix transcription factors, including MyoD and myogenin, functions cooperatively with the myocyte-specific enhancer binding factor 2 (MEF2) family during skeletal myogenesis. Previously, using aggregated P19 cells, we have shown that myogenin upregulates MEF2C expression while MyoD does not [Ridgeway et al., J. Biol. Chem. 275 (2000) 41-46]. In order to identify the domain of myogenin responsible for activating MEF2C expression, a series of chimeras of MyoD and myogenin were generated. Only chimeras containing the C-terminal region of myogenin were able to activate MEF2C in aggregated P19 cells, suggesting that the C-terminus of myogenin is responsible for the regulation of specific target genes.
Collapse
Affiliation(s)
- Parker J Rogerson
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
88
|
Paquin J, Danalache BA, Jankowski M, McCann SM, Gutkowska J. Oxytocin induces differentiation of P19 embryonic stem cells to cardiomyocytes. Proc Natl Acad Sci U S A 2002; 99:9550-5. [PMID: 12093924 PMCID: PMC123178 DOI: 10.1073/pnas.152302499] [Citation(s) in RCA: 169] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We recently discovered the existence of the oxytocin/oxytocin receptor (OT/OTR) system in the heart. Activation of cardiac OTR stimulates the release of atrial natriuretic peptide (ANP), which is involved in regulation of blood pressure and cell growth. Having observed elevated OT levels in the fetal and newborn heart at a stage of intense cardiomyocyte hyperplasia, we hypothesized a role for OT in cardiomyocyte differentiation. We used mouse P19 embryonic stem cells to substantiate this potential role. P19 cells give rise to the formation of cell derivatives of all germ layers. Treatment of P19 cell aggregates with dimethyl sulfoxide (DMSO) induces differentiation to cardiomyocytes. In this work, P19 cells were allowed to aggregate from day 0 to day 4 in the presence of 0.5% DMSO, 10(-7) M OT and/or 10(-7) M OT antagonist (OTA), and then cultured in the absence of these factors until day 14. OT alone stimulated the production of beating cell colonies in all 24 independently growing cultures by day 8 of the differentiation protocol, whereas the same result was obtained in cells induced by DMSO only after 12 days. Cells induced with OT exhibited increased ANP mRNA, had abundant mitochondria (i.e., they strongly absorbed rhodamine 123), and expressed sarcomeric myosin heavy chain and dihydropyridine receptor-alpha 1, confirming a cardiomyocyte phenotype. In addition, OT as well as DMSO increased OTR protein and OTR mRNA, and OTA completely inhibited the formation of cardiomyocytes in OT- and DMSO-supplemented cultures. These results suggest that the OT/OTR system plays an important role in cardiogenesis by promoting cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Joanne Paquin
- Laboratoire de Neuroendocrinologie Développementale, Département de Chimie et de Biochimie, Université du Québec, Montreal, QC, Canada H3C 3P8.
| | | | | | | | | |
Collapse
|
89
|
Eriksson M, Leppä S. Mitogen-activated protein kinases and activator protein 1 are required for proliferation and cardiomyocyte differentiation of P19 embryonal carcinoma cells. J Biol Chem 2002; 277:15992-6001. [PMID: 11884386 DOI: 10.1074/jbc.m107340200] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mitogen-activated protein kinases (MAPKs) have been implicated as regulators of differentiation. The biological effect of MAPK signaling in the nucleus is achieved by signal-responsive transcription factors. Here we have investigated MAPK signaling and activation of AP-1 transcription factors in P19 embryonal carcinoma cells undergoing cardiomyocyte differentiation. We show that aggregation and Me(2)SO treatment, which trigger the differentiation response, result in sustained activation of JNK1, p38, and ERK1/2 MAPKs and acquisition of AP-1 DNA binding activity. The induced AP-1 activity consists of c-Jun, JunD, and Fra-2 proteins and is accompanied with the increased expression of these proteins. JNK is involved in c-Jun phosphorylation, whereas ERK and p38 activities are essential for maximal c-Jun and Fra-2 expression, and AP-1 DNA binding activity. While the inhibition of ERK can partially prevent the formation of beating cardiomyocytes, the activity of p38 is absolutely required for the differentiation. Expression of dominant negative c-Jun(bZIP) in P19 cells can also inhibit the differentiation response. Surprisingly, however, expression of dominant negative SEK or JNK causes an inhibition of P19 cell proliferation. Taken together, the results show that ERK, JNK, p38, and AP-1 are activated in a coordinated and sustained manner, and contribute to proliferation and cardiomyocyte differentiation of P19 cells.
Collapse
Affiliation(s)
- Minna Eriksson
- Molecular Cancer Biology Research Program, Biomedicum Helsinki and Haartman Institute, University of Helsinki, P. O. Box 63, FIN-00014 Helsinki, Finland
| | | |
Collapse
|
90
|
Jamali M, Karamboulas C, Rogerson PJ, Skerjanc IS. BMP signaling regulates Nkx2-5 activity during cardiomyogenesis. FEBS Lett 2001; 509:126-30. [PMID: 11734219 DOI: 10.1016/s0014-5793(01)03151-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Nkx2-5 regulates the transcription of muscle-specific genes during cardiomyogenesis. Nkx2-5 expression can induce cardiomyogenesis in aggregated P19 cells but not in monolayer cultures. In order to investigate the mechanism by which cellular aggregation regulates Nkx2-5 function, we examined the role of bone morphogenetic protein 4 (BMP4). We showed that the expression of the BMP inhibitor, noggin, was sufficient to inhibit the induction of cardiomyogenesis by Nkx2-5 during cellular aggregation. Furthermore, soluble BMP4 could activate Nkx2-5 function in monolayer cultures, resulting in the formation of cardiomyocytes. Therefore, BMP signaling is necessary and sufficient for the regulation of Nkx2-5 activity during cardiomyogenesis in P19 cells.
Collapse
Affiliation(s)
- M Jamali
- Department of Biochemistry, Medical Sciences Building, The University of Western Ontario, London, ON N6A 5C1, Canada
| | | | | | | |
Collapse
|
91
|
Jamali M, Rogerson PJ, Wilton S, Skerjanc IS. Nkx2-5 activity is essential for cardiomyogenesis. J Biol Chem 2001; 276:42252-8. [PMID: 11526122 DOI: 10.1074/jbc.m107814200] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The homeobox transcription factor tinman is essential for heart vessel formation in Drosophila. In contrast, mice lacking the murine homologue Nkx2-5 are defective in cardiac looping but not in cardiac myocyte development. The lack of an essential role for Nkx2-5 in cardiomyogenesis in mammalian systems is most likely the result of genetic redundancy with family members. In this study, we used a dominant negative mutant of Nkx2-5, created by fusing the repressor domain of engrailed 2 to the Nkx2-5 homeodomain, termed Nkx/EnR. Expression of Nkx/EnR inhibited Me(2)SO-induced cardiomyogenesis in P19 cells but not skeletal myogenesis. Nkx/EnR inhibited expression of cardiomyoblast markers, such as GATA-4 and MEF2C, but not of mesoderm markers, such as Brachyury T and Wnt5b, or of skeletal lineage markers, such as MyoD and Mox1. To identify the minimal region of Nkx2-5 that can trigger cardiomyogenesis, we analyzed the activity of various Nkx2-5 deletion mutants. The C-terminal domain was not necessary for the ability of Nkx2-5 to induce cardiomyogenesis and loss of this domain did not enhance myogenesis. Therefore, Nkx2-5 function is essential for commitment of mesoderm into the cardiac muscle lineage, and the N-terminal region, together with the homeodomain, is sufficient for cardiomyogenesis in P19 cells.
Collapse
Affiliation(s)
- M Jamali
- Department of Biochemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | | | |
Collapse
|
92
|
Jamali M, Karamboulas C, Wilton S, Skerjanc IS. Factors in serum regulate Nkx2.5 and MEF2C function. In Vitro Cell Dev Biol Anim 2001; 37:635-7. [PMID: 11776966 DOI: 10.1290/1071-2690(2001)037<0635:fisrna>2.0.co;2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
93
|
Hiroi Y, Kudoh S, Monzen K, Ikeda Y, Yazaki Y, Nagai R, Komuro I. Tbx5 associates with Nkx2-5 and synergistically promotes cardiomyocyte differentiation. Nat Genet 2001; 28:276-80. [PMID: 11431700 DOI: 10.1038/90123] [Citation(s) in RCA: 416] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cardiac homeobox protein Nkx2-5 is essential in cardiac development, and mutations in Csx (which encodes Nkx2-5) cause various congenital heart diseases. Using the yeast two-hybrid system with Nkx2-5 as the 'bait', we isolated the T-box-containing transcription factor Tbx5; mutations in TBX5 cause heart and limb malformations in Holt-Oram syndrome (HOS). Co-transfection of Nkx2-5 and Tbx5 into COS-7 cells showed that they also associate with each other in mammalian cells. Glutathione S-transferase (GST) 'pull-down' assays indicated that the N-terminal domain and N-terminal part of the T-box of Tbx5 and the homeodomain of Nkx2-5 were necessary for their interaction. Tbx5 and Nkx2-5 directly bound to the promoter of the gene for cardiac-specific natriuretic peptide precursor type A (Nppa) in tandem, and both transcription factors showed synergistic activation. Deletion analysis showed that both the N-terminal domain and T-box of Tbx5 were important for this transactivation. A G80R mutation of Tbx5, which causes substantial cardiac defects with minor skeletal abnormalities in HOS, did not activate Nppa or show synergistic activation, whereas R237Q, which causes upper-limb malformations without cardiac abnormalities, activated the Nppa promoter to a similar extent to that of wildtype Tbx5. P19CL6 cell lines overexpressing wildtype Tbx5 started to beat earlier and expressed cardiac-specific genes more abundantly than did parental P19CL6 cells, whereas cell lines expressing the G80R mutant did not differentiate into beating cardiomyocytes. These results indicate that two different types of cardiac transcription factors synergistically induce cardiac development.
Collapse
Affiliation(s)
- Y Hiroi
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
94
|
Ridgeway AG, Skerjanc IS. Pax3 is essential for skeletal myogenesis and the expression of Six1 and Eya2. J Biol Chem 2001; 276:19033-9. [PMID: 11262400 DOI: 10.1074/jbc.m011491200] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Pax3 is a paired box transcription factor expressed during somitogenesis that has been implicated in initiating the expression of the myogenic regulatory factors during myogenesis. We find that Pax3 is necessary and sufficient to induce myogenesis in pluripotent stem cells. Pax3 induced the expression of the transcription factor Six1, its cofactor Eya2, and the transcription factor Mox1 prior to inducing the expression of MyoD and myogenin. Overexpression of a dominant negative Pax3, engineered by fusing the active transcriptional repression domain of mouse EN-2 in place of the Pax3 transcriptional activation domain, completely abolished skeletal myogenesis without inhibiting cardiogenesis. Expression of the dominant negative Pax3 resulted in a loss of expression of Six1, Eya2, and endogenous Pax3 as well as a down-regulation in the expression of Mox1. No effect was found on the expression of Gli2. These results indicate that Pax3 activity is essential for skeletal muscle development, the expression of Six1 and Eya2, and is involved in regulating its own expression. In summary, the combined approach of expressing both a wild type and dominant negative transcription factor in stem cells has identified a cascade of transcriptional events controlled by Pax3 that are necessary and sufficient for skeletal myogenesis.
Collapse
MESH Headings
- Blotting, Northern
- Cells, Cultured
- DNA-Binding Proteins/physiology
- Down-Regulation
- Gene Expression Regulation, Developmental
- Genes, Dominant
- Homeodomain Proteins/biosynthesis
- Humans
- Intracellular Signaling Peptides and Proteins
- Models, Biological
- Muscle, Skeletal/embryology
- Muscle, Skeletal/metabolism
- Muscles/cytology
- Muscles/metabolism
- MyoD Protein/biosynthesis
- Myogenin/biosynthesis
- NADH, NADPH Oxidoreductases/metabolism
- NADPH Oxidases
- Nuclear Proteins
- PAX3 Transcription Factor
- Paired Box Transcription Factors
- Plasmids/metabolism
- Protein Structure, Tertiary
- Protein Tyrosine Phosphatases
- Recombinant Fusion Proteins/metabolism
- Stem Cells/metabolism
- Trans-Activators/biosynthesis
- Transcription Factors
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- A G Ridgeway
- Department of Biochemistry, Medical Sciences Building, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | |
Collapse
|
95
|
Palmer S, Groves N, Schindeler A, Yeoh T, Biben C, Wang CC, Sparrow DB, Barnett L, Jenkins NA, Copeland NG, Koentgen F, Mohun T, Harvey RP. The small muscle-specific protein Csl modifies cell shape and promotes myocyte fusion in an insulin-like growth factor 1-dependent manner. J Cell Biol 2001; 153:985-98. [PMID: 11381084 PMCID: PMC2174333 DOI: 10.1083/jcb.153.5.985] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We have isolated a murine cDNA encoding a 9-kD protein, Chisel (Csl), in a screen for transcriptional targets of the cardiac homeodomain factor Nkx2-5. Csl transcripts were detected in atria and ventricles of the heart and in all skeletal muscles and smooth muscles of the stomach and pulmonary veins. Csl protein was distributed throughout the cytoplasm in fetal muscles, although costameric and M-line localization to the muscle cytoskeleton became obvious after further maturation. Targeted disruption of Csl showed no overt muscle phenotype. However, ectopic expression in C2C12 myoblasts induced formation of lamellipodia in which Csl protein became tethered to membrane ruffles. Migration of these cells was retarded in a monolayer wound repair assay. Csl-expressing myoblasts differentiated and fused normally, although in the presence of insulin-like growth factor (IGF)-1 they showed dramatically enhanced fusion, leading to formation of large dysmorphogenic "myosacs." The activities of transcription factors nuclear factor of activated T cells (NFAT) and myocyte enhancer-binding factor (MEF)2, were also enhanced in an IGF-1 signaling-dependent manner. The dynamic cytoskeletal localization of Csl and its dominant effects on cell shape and behavior and transcription factor activity suggest that Csl plays a role in the regulatory network through which muscle cells coordinate their structural and functional states during growth, adaptation, and repair.
Collapse
Affiliation(s)
- Steve Palmer
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Nicola Groves
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Aaron Schindeler
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Thomas Yeoh
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Christine Biben
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Cheng-Chun Wang
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Duncan B. Sparrow
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Louise Barnett
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3050, Australia
| | - Nancy A. Jenkins
- Mouse Cancer Genetics Program, National Cancer Institute-Frederick, Frederick, Maryland 21702
| | - Neal G. Copeland
- Mouse Cancer Genetics Program, National Cancer Institute-Frederick, Frederick, Maryland 21702
| | - Frank Koentgen
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3050, Australia
| | - Tim Mohun
- Medical Research Council Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- Faculties of Medicine and Life Sciences, University of New South Wales, Kensington, NSW 2051, Australia
| |
Collapse
|
96
|
Abstract
Building a vertebrate heart is a complex task and involves several tissues, including the myocardium, endocardium, neural crest, and epicardium. Interactions between these tissues result in the changes in function and morphology (and also in the extracellular matrix, which serves as a substrate for morphological change) that are requisite for development of the heart. Some of the signaling pathways that mediate these changes have now been identified and several investigators are now filling in the missing pieces in these pathways in hopes of ultimately understanding the molecular mechanisms that govern healthy heart development. In addition, transcription factors that regulate various aspects of heart development have been identified. Transcription factors of the GATA and Nkx2 families are of particular importance for early specification of the heart field and for regulating expression of genes that encode proteins of the contractile apparatus. This chapter highlights some of the most significant discoveries made in the rapidly expanding field of heart development.
Collapse
Affiliation(s)
- M J Farrell
- Developmental Biology Program, Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta 30912, USA
| | | |
Collapse
|
97
|
Ridgeway AG, Petropoulos H, Wilton S, Skerjanc IS. Wnt signaling regulates the function of MyoD and myogenin. J Biol Chem 2000; 275:32398-405. [PMID: 10915791 DOI: 10.1074/jbc.m004349200] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The myogenic regulatory factors (MRFs), MyoD and myogenin, can induce myogenesis in a variety of cell lines but not efficiently in monolayer cultures of P19 embryonal carcinoma stem cells. Aggregation of cells expressing MRFs, termed P19[MRF] cells, results in an approximately 30-fold enhancement of myogenesis. Here we examine molecular events occurring during P19 cell aggregation to identify potential mechanisms regulating MRF activity. Although myogenin protein was continually present in the nuclei of >90% of P19[myogenin] cells, only a fraction of these cells differentiated. Consequently, it appears that post-translational regulation controls myogenin activity in a cell lineage-specific manner. A correlation was obtained between the expression of factors involved in somite patterning, including Wnt3a, Wnt5b, BMP-2/4, and Pax3, and the induction of myogenesis. Co-culturing P19[Wnt3a] cells with P19[MRF] cells in monolayer resulted in a 5- to 8-fold increase in myogenesis. Neither BMP-4 nor Pax3 was efficient in enhancing MRF activity in unaggregated P19 cultures. Furthermore, BMP-4 abrogated the enhanced myogenesis induced by Wnt signaling. Consequently, signaling events resulting from Wnt3a expression but not BMP-4 signaling or Pax3 expression, regulate MRF function. Therefore, the P19 cell culture system can be used to study the link between somite patterning events and myogenesis.
Collapse
Affiliation(s)
- A G Ridgeway
- Department of Biochemistry, Medical Sciences Building, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | | | | | | |
Collapse
|
98
|
Abstract
Zinc finger-containing transcription factor GATA-4 and homeodomain Nkx-2.5 govern crucial developmental fates and have been found to promote cardiogenesis in embryonic cells exposed to the differentiating agent DMSO. Nevertheless, intracellular activators of these transcription factors are largely unknown. In this study, pluripotent P19 cells expressed the prodynorphin gene, an opioid gene encoding for the dynorphin family of opioid peptides. P19 cells were also able to synthesize and secrete dynorphin B, a biologically active end product of the prodynorphin gene. DMSO-primed GATA-4 and Nkx-2.5 gene expression was preceded by a marked increase in prodynorphin gene expression and dynorphin B synthesis and secretion. The DMSO effect occurred at the transcriptional level. In the absence of DMSO, dynorphin B triggered GATA-4 and Nkx-2.5 gene expression and led to the appearance of both alpha-myosin heavy chain and myosin light chain-2V transcripts, two markers of cardiac differentiation. Moreover, dynorphin B-exposed cells were positively stained in the presence of MF 20, a mouse monoclonal antibody raised against the alpha-myosin heavy chain. Opioid receptor antagonism and inhibition of opioid gene expression by a prodynorphin antisense phosphorothioate oligonucleotide blocked DMSO-induced cardiogenesis, suggesting an autocrine role of an opioid gene in developmental decisions.
Collapse
Affiliation(s)
- C Ventura
- Department of Biomedical Sciences, Division of Biochemistry, Laboratory of Cardiovascular Research, University of Sassari, Sassari, Italy.
| | | |
Collapse
|
99
|
Skerjanc IS, Wilton S. Myocyte enhancer factor 2C upregulates MASH-1 expression and induces neurogenesis in P19 cells. FEBS Lett 2000; 472:53-6. [PMID: 10781804 DOI: 10.1016/s0014-5793(00)01438-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
MEF2C is a transcription factor expressed in neural lineages. After transient transfection, the MEF2 family of factors can act synergistically with the neural-specific transcription factor, MASH-1, and activate exogenous neural-specific promoters. To determine whether MEF2C is capable of modulating endogenous gene expression, P19 cell lines were analyzed that overexpressed MEF2C, termed P19[MEF2C] cells. Here we show that P19[MEF2C] cells differentiate into neurons when aggregated with ME(2)SO. MEF2C-induced neurons expressed neurofilament protein, the nuclear antigen NeuN, as well as MASH-1. Our results indicate that MEF2C can directly or indirectly activate the expression of MASH-1, leading to neurogenesis.
Collapse
Affiliation(s)
- I S Skerjanc
- Department of Biochemistry, Medical Sciences Building, University of Western Ontario, London, Ont., Canada.
| | | |
Collapse
|
100
|
Davidson SM, Morange M. Hsp25 and the p38 MAPK pathway are involved in differentiation of cardiomyocytes. Dev Biol 2000; 218:146-60. [PMID: 10656759 DOI: 10.1006/dbio.1999.9596] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The small heat-shock protein HSP25 is expressed in the heart early during development, and although multiple roles for HSP25 have been proposed, its specific role during development and differentiation is not known. P19 is an embryonal carcinoma cell line which can be induced to differentiate in vitro into either cardiomyocytes or neurons. We have used P19 to examine the role of HSP25 in differentiation. We found that HSP25 expression is strongly increased in P19 cardiomyocytes. Antisense HSP25 expression reduced the extent of cardiomyocyte differentiation and resulted in reduced expression of cardiac actin and the intermediate filament desmin and reduced level of cardiac mRNAs. Thus, HSP25 is necessary for differentiation of P19 into cardiomyocytes. In contrast, P19 neurons did not express HSP25 and antisense HSP25 expression had no effect on neuronal differentiation. The phosphorylation of HSP25 by the p38/SAPK2 pathway is known to be important for certain of its functions. Inhibition of this pathway by the specific inhibitor SB203580 prevented cardiomyocyte differentiation of P19 cells. In contrast, PD90589, which inhibits the ERK1/2 pathway, had no effect. Surprisingly, cardiogenesis was only sensitive to SB203580 during the first 2 days of differentiation, before HSP25 expression increases. In contrast to the effect of antisense HSP25, SB203580 reduced the level of expression of the mesodermal marker Brachyury-T during differentiation. Therefore, we propose that the p38 pathway acts on an essential target during early cardiogenesis. Once this initial step is complete, HSP25 is necessary for the functional differentiation of P19 cardiomyocytes, but its phosphorylation by p38/SAPK2 is not required.
Collapse
Affiliation(s)
- S M Davidson
- Département de Biologie, Unité de Génétique Moléculaire, Ecole Normale Supérieure, 46 rue d'Ulm, Paris Cedex 05, 75230, France.
| | | |
Collapse
|