1
|
Derrien AC, Houy A, Ganier O, Dingli F, Ningarhari M, Mobuchon L, Espejo Díaz MI, Loew D, Cassoux N, Cussenot O, Cancel-Tassin G, Margueron R, Noirel J, Zucman-Rossi J, Rodrigues M, Stern MH. Functional characterization of 5p15.33 risk locus in uveal melanoma reveals rs452384 as a functional variant and NKX2.4 as an allele-specific interactor. Am J Hum Genet 2022; 109:2196-2209. [PMID: 36459980 PMCID: PMC9748249 DOI: 10.1016/j.ajhg.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 11/03/2022] [Indexed: 12/05/2022] Open
Abstract
The TERT/CLPTM1L risk locus on chromosome 5p15.33 is a pleiotropic cancer risk locus in which multiple independent risk alleles have been identified, across well over ten cancer types. We previously conducted a genome-wide association study in uveal melanoma (UM), which uncovered a role for the TERT/CLPTM1L risk locus in this intraocular tumor and identified multiple highly correlated risk alleles. Aiming to unravel the biological mechanisms in UM of this locus, which contains a domain enriched in active chromatin marks and enhancer elements, we demonstrated the allele-specific enhancer activity of this risk region using reporter assays. In UM, we identified the functional variant rs452384, of which the C risk allele is associated with higher gene expression, increased CLPTM1L expression in UM tumors, and a longer telomere length in peripheral blood mononuclear cells. Electrophoretic mobility shift assays and quantitative mass spectrometry identified NKX2.4 as an rs452384-T-specific binding protein, whereas GATA4 preferentially interacted with rs452384-C. Knockdown of NKX2.4 but not GATA4 resulted in increased TERT and CLPTM1L expression. In summary, the UM risk conferred by the 5p locus is at least partly due to rs452384, for which NKX2.4 presents strong differential binding activity and regulates CLPTM1L and TERT expression. Altogether, our work unraveled some of the complex regulatory mechanisms at the 5p15.33 susceptibility region in UM, and this might also shed light on shared mechanisms with other tumor types affected by this susceptibility region.
Collapse
Affiliation(s)
- Anne-Céline Derrien
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris 75005, France
| | - Alexandre Houy
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris 75005, France
| | - Olivier Ganier
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris 75005, France
| | - Florent Dingli
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, 26 rue d'Ulm, Paris 75248 Cedex 05, France
| | - Massih Ningarhari
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, 75006 Paris, France; Functional Genomics of Solid Tumors laboratory, Équipe labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, 75006 Paris, France
| | - Lenha Mobuchon
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris 75005, France
| | - María Isabel Espejo Díaz
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris 75005, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, 26 rue d'Ulm, Paris 75248 Cedex 05, France
| | - Nathalie Cassoux
- Department of Ocular Oncology, Institut Curie, Paris 75005, France; Factulty of Medicine, University of Paris Descartes, Paris 75005, France
| | - Olivier Cussenot
- CeRePP, Tenon Hospital, Paris 75020, France; Sorbonne University, GRC n°5 Predictive Onco-Urology, AP-HP, Tenon Hospital, Paris 75020, France; University of Oxford, Nuffield Department of Surgical Sciences, Oxford, UK
| | - Géraldine Cancel-Tassin
- CeRePP, Tenon Hospital, Paris 75020, France; Sorbonne University, GRC n°5 Predictive Onco-Urology, AP-HP, Tenon Hospital, Paris 75020, France
| | - Raphael Margueron
- Institut Curie, PSL Research University, Sorbonne University, Inserm U934/ CNRS UMR3215, 26, rue d'Ulm, 75005 Paris, France
| | - Josselin Noirel
- Laboratoire GBCM (EA7528), CNAM, HESAM Université, Paris, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, 75006 Paris, France; Functional Genomics of Solid Tumors laboratory, Équipe labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, 75006 Paris, France; Hôpital Européen Georges Pompidou, APHP, 75015, Paris, France
| | - Manuel Rodrigues
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris 75005, France; Institut Curie, PSL Research University, Department of Medical Oncology, Paris 75005, France
| | - Marc-Henri Stern
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, Paris 75005, France.
| |
Collapse
|
2
|
Deshpande A, Shetty PMV, Frey N, Rangrez AY. SRF: a seriously responsible factor in cardiac development and disease. J Biomed Sci 2022; 29:38. [PMID: 35681202 PMCID: PMC9185982 DOI: 10.1186/s12929-022-00820-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
The molecular mechanisms that regulate embryogenesis and cardiac development are calibrated by multiple signal transduction pathways within or between different cell lineages via autocrine or paracrine mechanisms of action. The heart is the first functional organ to form during development, which highlights the importance of this organ in later stages of growth. Knowledge of the regulatory mechanisms underlying cardiac development and adult cardiac homeostasis paves the way for discovering therapeutic possibilities for cardiac disease treatment. Serum response factor (SRF) is a major transcription factor that controls both embryonic and adult cardiac development. SRF expression is needed through the duration of development, from the first mesodermal cell in a developing embryo to the last cell damaged by infarction in the myocardium. Precise regulation of SRF expression is critical for mesoderm formation and cardiac crescent formation in the embryo, and altered SRF levels lead to cardiomyopathies in the adult heart, suggesting the vital role played by SRF in cardiac development and disease. This review provides a detailed overview of SRF and its partners in their various functions and discusses the future scope and possible therapeutic potential of SRF in the cardiovascular system.
Collapse
Affiliation(s)
- Anushka Deshpande
- Department of Internal Medicine III, Cardiology and Angiology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Prithviraj Manohar Vijaya Shetty
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
3
|
Nazari H, Heirani-Tabasi A, Esmaeili E, Kajbafzadeh AM, Hassannejad Z, Boroomand S, Shahsavari Alavijeh MH, Mishan MA, Ahmadi Tafti SH, Warkiani ME, Dadgar N. Decellularized human amniotic membrane reinforced by MoS2-Polycaprolactone nanofibers, a novel conductive scaffold for cardiac tissue engineering. J Biomater Appl 2022; 36:1527-1539. [DOI: 10.1177/08853282211063289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In order to regenerate myocardial tissues with functional characteristics, we need to copy some properties of the myocardium, such as its extracellular matrix and electrical conductivity. In this study, we synthesized nanosheets of Molybdenum disulfide (MoS2), and integrated them into polycaprolactone (PCL) and electrospun on the surface of decellularized human amniotic membrane (DHAM) with the purpose of improving the scaffolds mechanical properties and electrical conductivity. For in vitro studies, we seeded the mouse embryonic cardiac cells, mouse Embryonic Cardiac Cells (mECCs), on the scaffolds and then studied the MoS2 nanocomposites by scanning electron microscopy and Raman spectroscopy. In addition, we characterized the DHAM/PCL and DHAM/PCL-MoS2 by SEM, transmission electron microscopy, water contact angle measurement, electrical conductivity, and tensile test. Besides, we confirmed the scaffolds are biocompatible by 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide, MTT assay. Furthermore, by means of SEM images, it was shown that mECCs attached to the DHAM/PCL-MoS2 scaffold have more cell aggregations and elongated morphology. Furthermore, through the Real-Time PCR and immunostaining studies, we found out cardiac genes were maturated and upregulated, and they also included GATA-4, c-TnT, NKX 2.5, and alpha-myosin heavy chain in cells cultured on DHAM/PCL-MoS2 scaffold in comparison to DHAM/PCL and DHAM. Therefore, in terms of cardiac tissue engineering, DHAM nanofibrous scaffolds reinforced by PCL-MoS2 can be suggested as a proper candidate.
Collapse
Affiliation(s)
- Hojjallah Nazari
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Asieh Heirani-Tabasi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Esmaeili
- Stem Cell Technology Research Center, Tehran, Iran
- Arta Shimi Alborz Research Center, Tehran, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Safiye Boroomand
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- Institute of Molecular Medicine, Sechenov University, Moscow, 119991, Russia
| | - Neda Dadgar
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Colorectal Surgery, Digestive Disease Surgerical Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
4
|
Dixit R, Narasimhan C, Balekundri VI, Agrawal D, Kumar A, Mohapatra B. Functional analysis of novel genetic variants of NKX2-5 associated with nonsyndromic congenital heart disease. Am J Med Genet A 2021; 185:3644-3663. [PMID: 34214246 DOI: 10.1002/ajmg.a.62413] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 01/26/2023]
Abstract
NKX2-5, a master cardiac regulatory transcription factor was the first known genetic cause of congenital heart diseases (CHDs). To further investigate its role in CHD pathogenesis, we performed mutational screening of 285 CHD probands and 200 healthy controls. Five coding sequence variants were identified in six CHD cases (2.1%), including three in the N-terminal region (p.A61G, p.R95L, and p.E131K) and one each in homeodomain (HD) (p.A148E) and tyrosine-rich domain (p.P247A). Variant-p.A148E showed tertiary structure changes and differential DNA binding affinity of mutant compared to wild type. Two N-terminal variants-p.A61G and p.E131K along with HD variant p.A148E demonstrated significantly reduced transcriptional activity of Nppa and Actc1 promoters in dual luciferase promoter assay supported by their reduced expression in qRT-PCR. Nonetheless, variant p.R95L affected the synergy of NKX2-5 with serum response factor and TBX5 leading to significantly decreased Actc1 promoter activity depicting a distinctive role of this region. The aberrant expression of other target genes-Irx4, Mef2c, Bmp10, Myh6, Myh7, and Myocd is also observed in response to NKX2-5 variants, possibly due to the defective gene regulatory network. Severely impaired downstream promoter activities and abnormal expression of target genes due to N-terminal variants supports the emerging role of this region during cardiac-developmental pathways.
Collapse
Affiliation(s)
- Ritu Dixit
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Chitra Narasimhan
- Department of Pediatric Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, Karnataka, India
| | - Vijayalakshmi I Balekundri
- Super Speciality Hospital, Pradhan Mantri Swasthya Suraksha Yojana (PMSSY), Medical College and Research Institute, Bengaluru, Karnataka, India
| | - Damyanti Agrawal
- Department of Cardiothoracic and Vascular Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Pediatrics, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Bhagyalaxmi Mohapatra
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
5
|
Miranda MZ, Lichner Z, Szászi K, Kapus A. MRTF: Basic Biology and Role in Kidney Disease. Int J Mol Sci 2021; 22:ijms22116040. [PMID: 34204945 PMCID: PMC8199744 DOI: 10.3390/ijms22116040] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/21/2021] [Accepted: 05/30/2021] [Indexed: 12/23/2022] Open
Abstract
A lesser known but crucially important downstream effect of Rho family GTPases is the regulation of gene expression. This major role is mediated via the cytoskeleton, the organization of which dictates the nucleocytoplasmic shuttling of a set of transcription factors. Central among these is myocardin-related transcription factor (MRTF), which upon actin polymerization translocates to the nucleus and binds to its cognate partner, serum response factor (SRF). The MRTF/SRF complex then drives a large cohort of genes involved in cytoskeleton remodeling, contractility, extracellular matrix organization and many other processes. Accordingly, MRTF, activated by a variety of mechanical and chemical stimuli, affects a plethora of functions with physiological and pathological relevance. These include cell motility, development, metabolism and thus metastasis formation, inflammatory responses and—predominantly-organ fibrosis. The aim of this review is twofold: to provide an up-to-date summary about the basic biology and regulation of this versatile transcriptional coactivator; and to highlight its principal involvement in the pathobiology of kidney disease. Acting through both direct transcriptional and epigenetic mechanisms, MRTF plays a key (yet not fully appreciated) role in the induction of a profibrotic epithelial phenotype (PEP) as well as in fibroblast-myofibroblast transition, prime pathomechanisms in chronic kidney disease and renal fibrosis.
Collapse
Affiliation(s)
- Maria Zena Miranda
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
| | - Zsuzsanna Lichner
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| |
Collapse
|
6
|
Noori NM, shahraki Z, Karimi F, Miri-Moghaddam E. Rs4841587 in GATA4 and rs6999593 in DNMT1 gene associated with congenital heart diseases in the southeast of Iran. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
7
|
Abstract
Various strategies have been applied to replace the loss of cardiomyocytes in order to restore reduced cardiac function and prevent the progression of heart disease. Intensive research efforts in the field of cellular reprogramming and cell transplantation may eventually lead to efficient in vivo applications for the treatment of cardiac injuries, representing a novel treatment strategy for regenerative medicine. Modulation of cardiac transcription factor (TF) networks by chemical entities represents another viable option for therapeutic interventions. Comprehensive screening projects have revealed a number of molecular entities acting on molecular pathways highly critical for cellular lineage commitment and differentiation, including compounds targeting Wnt- and transforming growth factor beta (TGFβ)-signaling. Furthermore, previous studies have demonstrated that GATA4 and NKX2-5 are essential TFs in gene regulation of cardiac development and hypertrophy. For example, both of these TFs are required to fully activate mechanical stretch-responsive genes such as atrial natriuretic peptide and brain natriuretic peptide (BNP). We have previously reported that the compound 3i-1000 efficiently inhibited the synergy of the GATA4-NKX2-5 interaction. Cellular effects of 3i-1000 have been further characterized in a number of confirmatory in vitro bioassays, including rat cardiac myocytes and animal models of ischemic injury and angiotensin II-induced pressure overload, suggesting the potential for small molecule-induced cardioprotection.
Collapse
Affiliation(s)
- Mika J. Välimäki
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| | - Heikki J. Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of PharmacyUniversity of HelsinkiHelsinki, Finland
| |
Collapse
|
8
|
Miksiunas R, Mobasheri A, Bironaite D. Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac Development, Degeneration and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:155-178. [PMID: 30945165 DOI: 10.1007/5584_2019_349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are the most common cause of human death in the developing world. Extensive evidence indicates that various toxic environmental factors and unhealthy lifestyle choices contribute to the risk, incidence and severity of cardiovascular diseases. Alterations in the genetic level of myocardium affects normal heart development and initiates pathological processes leading to various types of cardiac diseases. Homeobox genes are a large and highly specialized family of closely related genes that direct the formation of body structure, including cardiac development. Homeobox genes encode homeodomain proteins that function as transcription factors with characteristic structures that allow them to bind to DNA, regulate gene expression and subsequently control the proper physiological function of cells, tissues and organs. Mutations in homeobox genes are rare and usually lethal with evident alterations in cardiac function at or soon after the birth. Our understanding of homeobox gene family expression and function has expanded significantly during the recent years. However, the involvement of homeobox genes in the development of human and animal cardiac tissue requires further investigation. The phenotype of human congenital heart defects unveils only some aspects of human heart development. Therefore, mouse models are often used to gain a better understanding of human heart function, pathology and regeneration. In this review, we have focused on the role of homeobox genes in the development and pathology of human heart as potential tools for the future development of targeted regenerative strategies for various heart malfunctions.
Collapse
Affiliation(s)
- Rokas Miksiunas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| |
Collapse
|
9
|
Tremblay M, Sanchez-Ferras O, Bouchard M. GATA transcription factors in development and disease. Development 2018; 145:145/20/dev164384. [DOI: 10.1242/dev.164384] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT
The GATA family of transcription factors is of crucial importance during embryonic development, playing complex and widespread roles in cell fate decisions and tissue morphogenesis. GATA proteins are essential for the development of tissues derived from all three germ layers, including the skin, brain, gonads, liver, hematopoietic, cardiovascular and urogenital systems. The crucial activity of GATA factors is underscored by the fact that inactivating mutations in most GATA members lead to embryonic lethality in mouse models and are often associated with developmental diseases in humans. In this Primer, we discuss the unique and redundant functions of GATA proteins in tissue morphogenesis, with an emphasis on their regulation of lineage specification and early organogenesis.
Collapse
Affiliation(s)
- Mathieu Tremblay
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal H3A 1A3, Canada
| | - Oraly Sanchez-Ferras
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal H3A 1A3, Canada
| | - Maxime Bouchard
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal H3A 1A3, Canada
| |
Collapse
|
10
|
Polyaniline cryogels: Biocompatibility of novel conducting macroporous material. Sci Rep 2018; 8:135. [PMID: 29317683 PMCID: PMC5760658 DOI: 10.1038/s41598-017-18290-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/05/2017] [Indexed: 12/22/2022] Open
Abstract
Polyaniline cryogel is a new unique form of polyaniline combining intrinsic electrical conductivity and the material properties of hydrogels. It is prepared by the polymerization of aniline in frozen poly(vinyl alcohol) solutions. The biocompatibility of macroporous polyaniline cryogel was demonstrated by testing its cytotoxicity on mouse embryonic fibroblasts and via the test of embryotoxicity based on the formation of beating foci within spontaneous differentiating embryonic stem cells. Good biocompatibility was related to low contents of low-molecular-weight impurities in polyaniline cryogel, which was confirmed by liquid chromatography. The adhesion and growth of embryonic stem cells, embryoid bodies, cardiomyocytes, and neural progenitors prove that polyaniline cryogel has the potential to be used as a carrier for cells in tissue engineering or bio-sensing. The surface energy as well as the elasticity and porosity of cryogel mimic tissue properties. Polyaniline cryogel can therefore be applied in bio-sensing or regenerative medicine in general, and mainly in the tissue engineering of electrically excitable tissues.
Collapse
|
11
|
Välimäki MJ, Tölli MA, Kinnunen SM, Aro J, Serpi R, Pohjolainen L, Talman V, Poso A, Ruskoaho HJ. Discovery of Small Molecules Targeting the Synergy of Cardiac Transcription Factors GATA4 and NKX2-5. J Med Chem 2017; 60:7781-7798. [PMID: 28858485 DOI: 10.1021/acs.jmedchem.7b00816] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Transcription factors are pivotal regulators of gene transcription, and many diseases are associated with the deregulation of transcriptional networks. In the heart, the transcription factors GATA4 and NKX2-5 are required for cardiogenesis. GATA4 and NKX2-5 interact physically, and the activation of GATA4, in cooperation with NKX2-5, is essential for stretch-induced cardiomyocyte hypertrophy. Here, we report the identification of four small molecule families that either inhibit or enhance the GATA4-NKX2-5 transcriptional synergy. A fragment-based screening, reporter gene assay, and pharmacophore search were utilized for the small molecule screening, identification, and optimization. The compounds modulated the hypertrophic agonist-induced cardiac gene expression. The most potent hit compound, N-[4-(diethylamino)phenyl]-5-methyl-3-phenylisoxazole-4-carboxamide (3, IC50 = 3 μM), exhibited no activity on the protein kinases involved in the regulation of GATA4 phosphorylation. The identified and chemically and biologically characterized active compound, and its derivatives may provide a novel class of small molecules for modulating heart regeneration.
Collapse
Affiliation(s)
- Mika J Välimäki
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland.,Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Marja A Tölli
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Sini M Kinnunen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland.,Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Jani Aro
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Raisa Serpi
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| | - Lotta Pohjolainen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland
| | - Virpi Talman
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland
| | - Antti Poso
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland , Kuopio FI-70211, Finland
| | - Heikki J Ruskoaho
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki , Helsinki FI-00014, Finland.,Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu , Oulu FI-90014, Finland
| |
Collapse
|
12
|
Xu JH, Gu JY, Guo YH, Zhang H, Qiu XB, Li RG, Shi HY, Liu H, Yang XX, Xu YJ, Qu XK, Yang YQ. Prevalence and Spectrum of NKX2-5 Mutations Associated With Sporadic Adult-Onset Dilated Cardiomyopathy. Int Heart J 2017; 58:521-529. [PMID: 28690296 DOI: 10.1536/ihj.16-440] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dilated cardiomyopathy (DCM), the most common form of primary myocardial disease, is a leading cause of congestive heart failure and the most common indication for heart transplantation. Recently, NKX2-5 mutations have been involved in the pathogenesis of familial DCM. However, the prevalence and spectrum of NKX2-5 mutations associated with sporadic DCM remain to be evaluated. In this study, the coding regions and flanking introns of the NKX2-5 gene, which encodes a cardiac transcription factor pivotal for cardiac development and structural remodeling, were sequenced in 210 unrelated patients with sporadic adult-onset DCM. A total of 300 unrelated healthy individuals used as controls were also genotyped for NKX2-5. The functional effect of the mutant NKX2-5 was investigated using a dual-luciferase reporter assay system. As a result, two novel heterozygous NKX2-5 mutations, p.R139W and p.E167X, were identified in 2 unrelated patients with sporadic adult-onset DCM, with a mutational prevalence of approximately 0.95%. The mutations were absent in 600 referential chromosomes and the altered amino acids were completely conserved evolutionarily across species. Functional assays revealed that the NKX2-5 mutants were associated with significantly reduced transcriptional activity. Furthermore, the mutations abrogated the synergistic activation between NKX2-5 and GATA4 as well as TBX20, two other cardiac key transcription factors that have been causally linked to adult-onset DCM. This study is the first to associate NKX2-5 loss-of-function mutations with enhanced susceptibility to sporadic DCM, which provides novel insight into the molecular etiology underpinning DCM, and suggests the potential implications for the genetic counseling and personalized treatment of the DCM patients.
Collapse
Affiliation(s)
- Jia-Hong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Jian-Yun Gu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Yu-Han Guo
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Hong Zhang
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Hong-Yu Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Hua Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Xiao-Xiao Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Xin-Kai Qu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University.,Department of Cardiovascular Research Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University.,Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University
| |
Collapse
|
13
|
Ranjbarvaziri S, Park S, Nguyen NB, Gilmore WB, Zhao P, Ardehali R. Generation of Nkx2-5/CreER transgenic mice for inducible Cre expression in developing hearts. Genesis 2017; 55. [PMID: 28589709 DOI: 10.1002/dvg.23041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 11/11/2022]
Abstract
Nkx2-5 is a homeobox-containing transcriptional regulator that serves as one of the earliest markers of cardiac lineage commitment. To study the role of Nkx2-5-expressing progenitors at specific time points in cardiac development, we have generated a novel and inducible NKX2-5 mouse line by knocking in a CreER cassette into the Nkx2-5 genomic locus, while preserving the endogenous Nkx2-5 gene to avoid haploinsufficiency. We evaluated the specificity and efficiency of CreER activity after 4-OHT injection by crossing Nkx2-5CreER/+ mice with a Rosa26tdT/+ reporter strain. Our immunohistochemistry results confirmed Cre-induced tdTomato expression specifically in cells expressing Nkx2-5. These cells were mainly cardiomyocytes and were observed in the embryonic heart as early as day 9.5. Additionally, quantitative polymerase chain reaction on postnatal hearts showed enriched expression of Nkx2-5 in isolated tdTomato-expressing cells. No tdTomato expression was observed in Nkx2-5CreER/+ ;Rosa26tdT/+ mice in the absence of 4-OHT, confirming the inducible nature of CreER activity. The Nkx2-5/CreER mouse model described in this article will serve as an invaluable tool to trace myocardial lineage and to temporally induce genetic manipulation in a selective population of cardiac progenitors during embryonic development and in the adult heart.
Collapse
Affiliation(s)
- Sara Ranjbarvaziri
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, 90095.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, 90095.,Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, California, 90095
| | - Shuin Park
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, 90095.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, 90095.,Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, California, 90095
| | - Ngoc B Nguyen
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, 90095.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, 90095.,Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, California, 90095
| | - William B Gilmore
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, 90095.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, 90095
| | - Peng Zhao
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, 90095.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, 90095.,Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, California, 90095
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, 90095.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, 90095.,Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, Los Angeles, California, 90095
| |
Collapse
|
14
|
Maskell LJ, Qamar K, Babakr AA, Hawkins TA, Heads RJ, Budhram-Mahadeo VS. Essential but partially redundant roles for POU4F1/Brn-3a and POU4F2/Brn-3b transcription factors in the developing heart. Cell Death Dis 2017; 8:e2861. [PMID: 28594399 PMCID: PMC5520879 DOI: 10.1038/cddis.2017.185] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 01/15/2023]
Abstract
Congenital heart defects contribute to embryonic or neonatal lethality but due to the complexity of cardiac development, the molecular changes associated with such defects are not fully understood. Here, we report that transcription factors (TFs) Brn-3a (POU4F1) and Brn-3b (POU4F2) are important for normal cardiac development. Brn-3a directly represses Brn-3b promoter in cardiomyocytes and consequently Brn-3a knockout (KO) mutant hearts express increased Brn-3b mRNA during mid-gestation, which is linked to hyperplastic growth associated with elevated cyclin D1, a known Brn-3b target gene. However, during late gestation, Brn-3b can cooperate with p53 to enhance transcription of pro-apoptotic genes e.g. Bax, thereby increasing apoptosis and contribute to morphological defects such as non-compaction, ventricular wall/septal thinning and increased crypts/fissures, which may cause lethality of Brn-3a KO mutants soon after birth. Despite this, early embryonic lethality in e9.5 double KO (Brn-3a-/- : Brn-3b-/-) mutants indicate essential functions with partial redundancy during early embryogenesis. High conservation between mammals and zebrafish (ZF) Brn-3b (87%) or Brn-3a (76%) facilitated use of ZF embryos to study potential roles in developing heart. Double morphant embryos targeted with morpholino oligonucleotides to both TFs develop significant cardiac defects (looping abnormalities and valve defects) suggesting essential roles for Brn-3a and Brn-3b in developing hearts.
Collapse
Affiliation(s)
- Lauren J Maskell
- Medical Molecular Biology Unit, Institute of Cardiovascular Science, University College London, UCL Rayne Building, London, UK
| | - Kashif Qamar
- Medical Molecular Biology Unit, Institute of Cardiovascular Science, University College London, UCL Rayne Building, London, UK
| | - Aram A Babakr
- Medical Molecular Biology Unit, Institute of Cardiovascular Science, University College London, UCL Rayne Building, London, UK
| | - Thomas A Hawkins
- Division of Biosciences, Cell and Developmental Biology, UCL, London, UK
| | - Richard J Heads
- Cardiovascular Division, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Vishwanie S Budhram-Mahadeo
- Medical Molecular Biology Unit, Institute of Cardiovascular Science, University College London, UCL Rayne Building, London, UK
| |
Collapse
|
15
|
Al-Maqtari T, Hong KU, Vajravelu BN, Moktar A, Cao P, Moore JB, Bolli R. Transcription factor-induced activation of cardiac gene expression in human c-kit+ cardiac progenitor cells. PLoS One 2017; 12:e0174242. [PMID: 28355297 PMCID: PMC5371315 DOI: 10.1371/journal.pone.0174242] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/06/2017] [Indexed: 12/11/2022] Open
Abstract
Although transplantation of c-kit+ cardiac progenitor cells (CPCs) significantly alleviates post-myocardial infarction left ventricular dysfunction, generation of cardiomyocytes by exogenous CPCs in the recipient heart has often been limited. Inducing robust differentiation would be necessary for improving the efficacy of the regenerative cardiac cell therapy. We assessed the hypothesis that differentiation of human c-kit+ CPCs can be enhanced by priming them with cardiac transcription factors (TFs). We introduced five different TFs (Gata4, MEF2C, NKX2.5, TBX5, and BAF60C) into CPCs, either alone or in combination, and then examined the expression of marker genes associated with the major cardiac cell types using quantitative RT-PCR. When introduced individually, Gata4 and TBX5 induced a subset of myocyte markers. Moreover, Gata4 alone significantly induced smooth muscle cell and fibroblast markers. Interestingly, these gene expression changes brought by Gata4 were also accompanied by morphological changes. In contrast, MEF2C and NKX2.5 were largely ineffective in initiating cardiac gene expression in CPCs. Surprisingly, introduction of multiple TFs in different combinations mostly failed to act synergistically. Likewise, addition of BAF60C to Gata4 and/or TBX5 did not further potentiate their effects on cardiac gene expression. Based on our results, it appears that GATA4 is able to potentiate gene expression programs associated with multiple cardiovascular lineages in CPCs, suggesting that GATA4 may be effective in priming CPCs for enhanced differentiation in the setting of stem cell therapy.
Collapse
Affiliation(s)
- Tareq Al-Maqtari
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Kyung U. Hong
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Bathri N. Vajravelu
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Afsoon Moktar
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Pengxiao Cao
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Joseph B. Moore
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
- * E-mail:
| |
Collapse
|
16
|
Ramialison M, Waardenberg AJ, Schonrock N, Doan T, de Jong D, Bouveret R, Harvey RP. Analysis of steric effects in DamID profiling of transcription factor target genes. Genomics 2017; 109:75-82. [DOI: 10.1016/j.ygeno.2017.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/19/2017] [Accepted: 01/29/2017] [Indexed: 01/08/2023]
|
17
|
Laemmle LL, Cohen JB, Glorioso JC. Constitutive Expression of GATA4 Dramatically Increases the Cardiogenic Potential of D3 Mouse Embryonic Stem Cells. ACTA ACUST UNITED AC 2016; 10:248-257. [PMID: 27441042 PMCID: PMC4948750 DOI: 10.2174/1874070701610010248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The transcription factor GATA binding protein 4 (GATA4) is a vital regulator of cardiac programming that acts by inducing the expression of many different genes involved in cardiomyogenesis. Here we generated a D3 mouse embryonic stem cell line that constitutively expresses high levels of GATA4 and show that these cells have dramatically increased cardiogenic potential compared to an eGFP-expressing control cell line. Embryoid bodies (EB) derived from the D3-GATA4 line displayed increased levels of cardiac gene expression and showed more abundant cardiomyocyte differentiation than control eGFP EB. These cells and two additional lines expressing lower levels of GATA4 provide a platform to screen previously untested cardiac genes and gene combinations for their ability to further increase the efficiency of cardiomyocyte differentiation beyond that achieved by transgenic GATA4 alone. Non-integrative delivery of identified gene combinations will aid in the production of differentiated cells for the treatment of ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Lillian L Laemmle
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15219, USA
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15219, USA
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
18
|
HIF-1alpha Deficiency Attenuates the Cardiomyogenesis of Mouse Embryonic Stem Cells. PLoS One 2016; 11:e0158358. [PMID: 27355368 PMCID: PMC4927095 DOI: 10.1371/journal.pone.0158358] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/14/2016] [Indexed: 01/09/2023] Open
Abstract
Cardiac cell formation, cardiomyogenesis, is critically dependent on oxygen availability. It is known that hypoxia, a reduced oxygen level, modulates the in vitro differentiation of pluripotent cells into cardiomyocytes via hypoxia inducible factor-1alpha (HIF-1α)-dependent mechanisms. However, the direct impact of HIF-1α deficiency on the formation and maturation of cardiac-like cells derived from mouse embryonic stem cells (mESC) in vitro remains to be elucidated. In the present study, we demonstrated that HIF-1α deficiency significantly altered the quality and quantity of mESC-derived cardiomyocytes. It was accompanied with lower mRNA and protein levels of cardiac cell specific markers (myosin heavy chains 6 and 7) and with a decreasing percentage of myosin heavy chain α and β, and cardiac troponin T-positive cells. As to structural aspects of the differentiated cardiomyocytes, the localization of contractile proteins (cardiac troponin T, myosin heavy chain α and β) and the organization of myofibrils were also different. Simultaneously, HIF-1α deficiency was associated with a lower percentage of beating embryoid bodies. Interestingly, an observed alteration in the in vitro differentiation scheme of HIF-1α deficient cells was accompanied with significantly lower expression of the endodermal marker (hepatic nuclear factor 4 alpha). These findings thus suggest that HIF-1α deficiency attenuates spontaneous cardiomyogenesis through the negative regulation of endoderm development in mESC differentiating in vitro.
Collapse
|
19
|
A Matter of the Heart: The African Clawed Frog Xenopus as a Model for Studying Vertebrate Cardiogenesis and Congenital Heart Defects. J Cardiovasc Dev Dis 2016; 3:jcdd3020021. [PMID: 29367567 PMCID: PMC5715680 DOI: 10.3390/jcdd3020021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/25/2016] [Accepted: 05/30/2016] [Indexed: 12/20/2022] Open
Abstract
The African clawed frog, Xenopus, is a valuable non-mammalian model organism to investigate vertebrate heart development and to explore the underlying molecular mechanisms of human congenital heart defects (CHDs). In this review, we outline the similarities between Xenopus and mammalian cardiogenesis, and provide an overview of well-studied cardiac genes in Xenopus, which have been associated with congenital heart conditions. Additionally, we highlight advantages of modeling candidate genes derived from genome wide association studies (GWAS) in Xenopus and discuss commonly used techniques.
Collapse
|
20
|
Furtado MB, Wilmanns JC, Chandran A, Tonta M, Biben C, Eichenlaub M, Coleman HA, Berger S, Bouveret R, Singh R, Harvey RP, Ramialison M, Pearson JT, Parkington HC, Rosenthal NA, Costa MW. A novel conditional mouse model for Nkx2-5 reveals transcriptional regulation of cardiac ion channels. Differentiation 2016; 91:29-41. [PMID: 26897459 DOI: 10.1016/j.diff.2015.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 01/30/2023]
Abstract
Nkx2-5 is one of the master regulators of cardiac development, homeostasis and disease. This transcription factor has been previously associated with a suite of cardiac congenital malformations and impairment of electrical activity. When disease causative mutations in transcription factors are considered, NKX2-5 gene dysfunction is the most common abnormality found in patients. Here we describe a novel mouse model and subsequent implications of Nkx2-5 loss for aspects of myocardial electrical activity. In this work we have engineered a new Nkx2-5 conditional knockout mouse in which flox sites flank the entire Nkx2-5 locus, and validated this line for the study of heart development, differentiation and disease using a full deletion strategy. While our homozygous knockout mice show typical embryonic malformations previously described for the lack of the Nkx2-5 gene, hearts of heterozygous adult mice show moderate morphological and functional abnormalities that are sufficient to sustain blood supply demands under homeostatic conditions. This study further reveals intriguing aspects of Nkx2-5 function in the control of cardiac electrical activity. Using a combination of mouse genetics, biochemistry, molecular and cell biology, we demonstrate that Nkx2-5 regulates the gene encoding Kcnh2 channel and others, shedding light on potential mechanisms generating electrical abnormalities observed in patients bearing NKX2-5 dysfunction and opening opportunities to the study of novel therapeutic targets for anti-arrhythmogenic therapies.
Collapse
Affiliation(s)
- Milena B Furtado
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia; The Jackson Laboratory, ME 04609, United States
| | - Julia C Wilmanns
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia; Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Anjana Chandran
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia
| | - Mary Tonta
- Department of Physiology, Monash University, Clayton, Vic 3800, Australia
| | - Christine Biben
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Vic 3052, Australia
| | - Michael Eichenlaub
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia
| | - Harold A Coleman
- Department of Physiology, Monash University, Clayton, Vic 3800, Australia
| | - Silke Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia
| | - Romaric Bouveret
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Reena Singh
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia
| | - James T Pearson
- Department of Physiology, Monash University, Clayton, Vic 3800, Australia; Monash Biomedical Imaging, Monash University, Clayton, Vic 3800, Australia
| | | | - Nadia A Rosenthal
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia; National Heart and Lung Institute, Imperial College London, SW3 6LY England, UK; The Jackson Laboratory, ME 04609, United States
| | - Mauro W Costa
- Australian Regenerative Medicine Institute, Monash University, Clayton, Vic 3800, Australia; The Jackson Laboratory, ME 04609, United States.
| |
Collapse
|
21
|
Kinnunen S, Välimäki M, Tölli M, Wohlfahrt G, Darwich R, Komati H, Nemer M, Ruskoaho H. Nuclear Receptor-Like Structure and Interaction of Congenital Heart Disease-Associated Factors GATA4 and NKX2-5. PLoS One 2015; 10:e0144145. [PMID: 26642209 PMCID: PMC4671672 DOI: 10.1371/journal.pone.0144145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/13/2015] [Indexed: 01/24/2023] Open
Abstract
AIMS Transcription factor GATA4 is a dosage sensitive regulator of heart development and alterations in its level or activity lead to congenital heart disease (CHD). GATA4 has also been implicated in cardiac regeneration and repair. GATA4 action involves combinatorial interaction with other cofactors such as NKX2-5, another critical cardiac regulator whose mutations also cause CHD. Despite its critical importance to the heart and its evolutionary conservation across species, the structural basis of the GATA4-NKX2-5 interaction remains incompletely understood. METHODS AND RESULTS A homology model was constructed and used to identify surface amino acids important for the interaction of GATA4 and NKX2-5. These residues were subjected to site-directed mutagenesis, and the mutant proteins were characterized for their ability to bind DNA and to physically and functionally interact with NKX2-5. The studies identify 5 highly conserved amino acids in the second zinc finger (N272, R283, Q274, K299) and its C-terminal extension (R319) that are critical for physical and functional interaction with the third alpha helix of NKX2-5 homeodomain. Integration of the experimental data with computational modeling suggests that the structural arrangement of the zinc finger-homeodomain resembles the architecture of the conserved DNA binding domain of nuclear receptors. CONCLUSIONS The results provide novel insight into the structural basis for protein-protein interactions between two important classes of transcription factors. The model proposed will help to elucidate the molecular basis for disease causing mutations in GATA4 and NKX2-5 and may be relevant to other members of the GATA and NK classes of transcription factors.
Collapse
Affiliation(s)
- Sini Kinnunen
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Mika Välimäki
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Marja Tölli
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Gerd Wohlfahrt
- Orion Pharma, Computer-Aided Drug Design, Espoo, Finland
| | - Rami Darwich
- Laboratory of Cardiac Development and Differentiation, Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, Canada
| | - Hiba Komati
- Laboratory of Cardiac Development and Differentiation, Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, Canada
| | - Mona Nemer
- Laboratory of Cardiac Development and Differentiation, Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, Canada
- * E-mail: (HR); (MN)
| | - Heikki Ruskoaho
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
- * E-mail: (HR); (MN)
| |
Collapse
|
22
|
Lovato TL, Sensibaugh CA, Swingle KL, Martinez MM, Cripps RM. The Drosophila Transcription Factors Tinman and Pannier Activate and Collaborate with Myocyte Enhancer Factor-2 to Promote Heart Cell Fate. PLoS One 2015. [PMID: 26225919 PMCID: PMC4520567 DOI: 10.1371/journal.pone.0132965] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Expression of the MADS domain transcription factor Myocyte Enhancer Factor 2 (MEF2) is regulated by numerous and overlapping enhancers which tightly control its transcription in the mesoderm. To understand how Mef2 expression is controlled in the heart, we identified a late stage Mef2 cardiac enhancer that is active in all heart cells beginning at stage 14 of embryonic development. This enhancer is regulated by the NK-homeodomain transcription factor Tinman, and the GATA transcription factor Pannier through both direct and indirect interactions with the enhancer. Since Tinman, Pannier and MEF2 are evolutionarily conserved from Drosophila to vertebrates, and since their vertebrate homologs can convert mouse fibroblast cells to cardiomyocytes in different activator cocktails, we tested whether over-expression of these three factors in vivo could ectopically activate known cardiac marker genes. We found that mesodermal over-expression of Tinman and Pannier resulted in approximately 20% of embryos with ectopic Hand and Sulphonylurea receptor (Sur) expression. By adding MEF2 alongside Tinman and Pannier, a dramatic expansion in the expression of Hand and Sur was observed in almost all embryos analyzed. Two additional cardiac markers were also expanded in their expression. Our results demonstrate the ability to initiate ectopic cardiac fate in vivo by the combination of only three members of the conserved Drosophila cardiac transcription network, and provide an opportunity for this genetic model system to be used to dissect the mechanisms of cardiac specification.
Collapse
Affiliation(s)
- TyAnna L. Lovato
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Cheryl A. Sensibaugh
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Kirstie L. Swingle
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Melody M. Martinez
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
| | - Richard M. Cripps
- Department of Biology, University of New Mexico, Albuquerque, NM 87131–1091, United States of America
- * E-mail:
| |
Collapse
|
23
|
GATA-dependent transcriptional and epigenetic control of cardiac lineage specification and differentiation. Cell Mol Life Sci 2015; 72:3871-81. [PMID: 26126786 PMCID: PMC4575685 DOI: 10.1007/s00018-015-1974-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/14/2022]
Abstract
Heart progenitor cells differentiate into various cell types including pacemaker and working cardiomyocytes. Cell-type specific gene expression is achieved by combinatorial interactions between tissue-specific transcription factors (TFs), co-factors, and chromatin remodelers and DNA binding elements in regulatory regions. Dysfunction of these transcriptional networks may result in congenital heart defects. Functional analysis of the regulatory DNA sequences has contributed substantially to the identification of the transcriptional network components and combinatorial interactions regulating the tissue-specific gene programs. GATA TFs have been identified as central players in these networks. In particular, GATA binding elements have emerged as a platform to recruit broadly active histone modification enzymes and cell-type-specific co-factors to drive cell-type-specific gene programs. Here, we discuss the role of GATA factors in cell fate decisions and differentiation in the developing heart.
Collapse
|
24
|
Forward Programming of Cardiac Stem Cells by Homogeneous Transduction with MYOCD plus TBX5. PLoS One 2015; 10:e0125384. [PMID: 26047103 PMCID: PMC4457652 DOI: 10.1371/journal.pone.0125384] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/23/2015] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Adult cardiac stem cells (CSCs) express many endogenous cardiogenic transcription factors including members of the Gata, Hand, Mef2, and T-box family. Unlike its DNA-binding targets, Myocardin (Myocd)-a co-activator not only for serum response factor, but also for Gata4 and Tbx5-is not expressed in CSCs. We hypothesised that its absence was a limiting factor for reprogramming. Here, we sought to investigate the susceptibility of adult mouse Sca1+ side population CSCs to reprogramming by supplementing the triad of GATA4, MEF2C, and TBX5 (GMT), and more specifically by testing the effect of the missing co-activator, Myocd. Exogenous factors were expressed via doxycycline-inducible lentiviral vectors in various combinations. High throughput quantitative RT-PCR was used to test expression of 29 cardiac lineage markers two weeks post-induction. GMT induced more than half the analysed cardiac transcripts. However, no protein was detected for the induced sarcomeric genes Actc1, Myh6, and Myl2. Adding MYOCD to GMT affected only slightly the breadth and level of gene induction, but, importantly, triggered expression of all three proteins examined (α-cardiac actin, atrial natriuretic peptide, sarcomeric myosin heavy chains). MYOCD + TBX was the most effective pairwise combination in this system. In clonal derivatives homogenously expressing MYOCD + TBX at high levels, 93% of cardiac transcripts were up-regulated and all five proteins tested were visualized. IN SUMMARY (1) GMT induced cardiac genes in CSCs, but not cardiac proteins under the conditions used. (2) Complementing GMT with MYOCD induced cardiac protein expression, indicating a more complete cardiac differentiation program. (3) Homogeneous transduction with MYOCD + TBX5 facilitated the identification of differentiating cells and the validation of this combinatorial reprogramming strategy. Together, these results highlight the pivotal importance of MYOCD in driving CSCs toward a cardiac muscle fate.
Collapse
|
25
|
Chaturvedi P, Kalani A, Familtseva A, Kamat PK, Metreveli N, Tyagi SC. Cardiac tissue inhibitor of matrix metalloprotease 4 dictates cardiomyocyte contractility and differentiation of embryonic stem cells into cardiomyocytes: Road to therapy. Int J Cardiol 2015; 184:350-363. [PMID: 25745981 PMCID: PMC4417452 DOI: 10.1016/j.ijcard.2015.01.091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 01/08/2015] [Accepted: 01/24/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND TIMP4 (Tissue Inhibitors of Matrix Metalloprotease 4), goes down in failing hearts and mice lacking TIMP4 show poor regeneration capacity after myocardial infarction (MI). This study is based on our previous observation that administration of cardiac inhibitor of metalloproteinase (~TIMP4) attenuates oxidative stress and remodeling in failing hearts. Therefore, we hypothesize that TIMP4 helps in cardiac regeneration by augmenting contractility and inducing the differentiation of cardiac progenitor cells into cardiomyocytes. METHODS To validate this hypothesis, we transfected mouse cardiomyocytes with TIMP4 and TIMP4-siRNA and performed contractility studies in the TIMP4 transfected cardiomyocytes as compared to siRNA-TIMP4 transfected cardiomyocytes. We evaluated the calcium channel gene serca2a (sarcoplasmic reticulum calcium ATPase2a) and mir122a which tightly regulates serca2a to explain the changes in contractility. We treated mouse embryonic stem cells with cardiac extract and cardiac extract minus TIMP4 (using TIMP4 monoclonal antibody) to examine the effect of TIMP4 on differentiation of cardiac progenitor cells. RESULTS Contractility was augmented in the TIMP4 transfected cardiomyocytes as compared to siRNA-TIMP4 transfected cardiomyocytes. There was elevated expression of serca2a in the TIMP4 transformed myocytes and down regulation of mir122a. The cells treated with cardiac extract containing TIMP4 showed cardiac phenotype in terms of Ckit+, GATA4+ and Nkx2.5 expression. CONCLUSION This is a novel report suggesting that TIMP4 augments contractility and induces differentiation of progenitor cells into cardiac phenotype. In view of the failure of MMP9 inhibitors for cardiac therapy, TIMP4 provides an alternative approach, being an indigenous molecule and a natural inhibitor of MMP9.
Collapse
Affiliation(s)
- Pankaj Chaturvedi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA.
| | - Anuradha Kalani
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Anastasia Familtseva
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Pradip Kumar Kamat
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Naira Metreveli
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| |
Collapse
|
26
|
Cardiac transcription factor Nkx2.5 interacts with p53 and modulates its activity. Arch Biochem Biophys 2015; 569:45-53. [DOI: 10.1016/j.abb.2015.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/01/2015] [Indexed: 01/30/2023]
|
27
|
The regulation of troponins I, C and ANP by GATA4 and Nkx2-5 in heart of hibernating thirteen-lined ground squirrels, Ictidomys tridecemlineatus. PLoS One 2015; 10:e0117747. [PMID: 25679215 PMCID: PMC4334527 DOI: 10.1371/journal.pone.0117747] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 01/02/2015] [Indexed: 02/06/2023] Open
Abstract
Hibernation is an adaptive strategy used by various mammals to survive the winter under situations of low ambient temperatures and limited or no food availability. The heart of hibernating thirteen-lined ground squirrels (Ictidomys tridecemlineatus) has the remarkable ability to descend to low, near 0°C temperatures without falling into cardiac arrest. We hypothesized that the transcription factors GATA4 and Nkx2-5 may play a role in cardioprotection by facilitating the expression of key downstream targets such as troponin I, troponin C, and ANP (atrial natriuretic peptide). This study measured relative changes in transcript levels, protein levels, protein post-translational modifications, and transcription factor binding over six stages: euthermic control (EC), entrance into torpor (EN), early torpor (ET), late torpor (LT), early arousal (EA), and interbout arousal (IA). We found differential regulation of GATA4 whereby transcript/protein expression, post-translational modification (phosphorylation of serine 261), and DNA binding were enhanced during the transitory phases (entrance and arousal) of hibernation. Activation of GATA4 was paired with increases in cardiac troponin I, troponin C and ANP protein levels during entrance, while increases in p-GATA4 DNA binding during early arousal was paired with decreases in troponin I and no changes in troponin C and ANP protein levels. Unlike its binding partner, the relative mRNA/protein expression and DNA binding of Nkx2-5 did not change during hibernation. This suggests that either Nkx2-5 does not play a substantial role or other regulatory mechanisms not presently studied (e.g. posttranslational modifications) are important during hibernation. The data suggest a significant role for GATA4-mediated gene transcription in the differential regulation of genes which aid cardiac-specific challenges associated with torpor-arousal.
Collapse
|
28
|
Yuan F, Qiu XB, Li RG, Qu XK, Wang J, Xu YJ, Liu X, Fang WY, Yang YQ, Liao DN. A novel NKX2-5 loss-of-function mutation predisposes to familial dilated cardiomyopathy and arrhythmias. Int J Mol Med 2014; 35:478-86. [PMID: 25503402 DOI: 10.3892/ijmm.2014.2029] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 12/05/2014] [Indexed: 01/08/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is the most prevalent type of primary myocardial disease, which is the third most common cause of heart failure and the most frequent reason for heart transplantation. Aggregating evidence demonstrates that genetic risk factors are involved in the pathogenesis of idiopathic DCM. Nevertheless, DCM is of remarkable genetic heterogeneity and the genetic defects underpinning DCM in an overwhelming majority of patients remain unknown. In the present study, the whole coding exons and splice junction sites of the NKX2-5 gene, which encodes a homeodomain transcription factor crucial for cardiac development and structural remodeling, were sequenced in 130 unrelated patients with idiopathic DCM. The available relatives of the index patient harboring an identified mutation and 200 unrelated ethnically matched healthy individuals used as controls were genotyped for the NKX2-5 gene. The functional effect of the mutant NKX2-5 was characterized in contrast to its wild-type counterpart using a dual-luciferase reporter assay system. As a result, a novel heterozygous NKX2-5 mutation, p.S146W, was identified in a family with DCM inherited as an autosomal dominant trait, which co-segregated with DCM in the family with complete penetrance. Notably, the mutation carriers also had arrhythmias, such as paroxysmal atrial fibrillation and atrioventricular block. The missense mutation was absent in 400 reference chromosomes and the altered amino acid was completely conserved evolutionarily among species. Functional analysis revealed that the NKX2-5 mutant was associated with a significantly reduced transcriptional activity. The findings expand the mutational spectrum of NKX2-5 linked to DCM and provide novel insight into the molecular mechanisms underlying DCM, contributing to the antenatal prophylaxis and allele-specific management of DCM.
Collapse
Affiliation(s)
- Fang Yuan
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xin-Kai Qu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Juan Wang
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xu Liu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Wei-Yi Fang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - De-Ning Liao
- Department of Cardiology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
29
|
Kempf H, Olmer R, Kropp C, Rückert M, Jara-Avaca M, Robles-Diaz D, Franke A, Elliott DA, Wojciechowski D, Fischer M, Roa Lara A, Kensah G, Gruh I, Haverich A, Martin U, Zweigerdt R. Controlling expansion and cardiomyogenic differentiation of human pluripotent stem cells in scalable suspension culture. Stem Cell Reports 2014; 3:1132-46. [PMID: 25454631 PMCID: PMC4264033 DOI: 10.1016/j.stemcr.2014.09.017] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/13/2022] Open
Abstract
To harness the potential of human pluripotent stem cells (hPSCs), an abundant supply of their progenies is required. Here, hPSC expansion as matrix-independent aggregates in suspension culture was combined with cardiomyogenic differentiation using chemical Wnt pathway modulators. A multiwell screen was scaled up to stirred Erlenmeyer flasks and subsequently to tank bioreactors, applying controlled feeding strategies (batch and cyclic perfusion). Cardiomyogenesis was sensitive to the GSK3 inhibitor CHIR99021 concentration, whereas the aggregate size was no prevailing factor across culture platforms. However, in bioreactors, the pattern of aggregate formation in the expansion phase dominated subsequent differentiation. Global profiling revealed a culture-dependent expression of BMP agonists/antagonists, suggesting their decisive role in cell-fate determination. Furthermore, metallothionein was discovered as a potentially stress-related marker in hPSCs. In 100 ml bioreactors, the production of 40 million predominantly ventricular-like cardiomyocytes (up to 85% purity) was enabled that were directly applicable to bioartificial cardiac tissue formation. Efficient cardiac differentiation protocol in suspension by chemical Wnt modulators Differentiation is CHIR concentration dependent, but aggregate size independent Bioreactor-controlled hPSC expansion dictates subsequent lineage differentiation Metallothionein is a potentially stress-induced marker of hPSC culture
Collapse
Affiliation(s)
- Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; Member of the Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Christina Kropp
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Michael Rückert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Monica Jara-Avaca
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Diana Robles-Diaz
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - David A Elliott
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Flemington Road, Parkville, VIC 3052, Australia
| | - Daniel Wojciechowski
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Martin Fischer
- Institute for Neurophysiology, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Angelica Roa Lara
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - George Kensah
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Ina Gruh
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; Member of the Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation, and Vascular Surgery (HTTG), Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany; REBIRTH-Cluster of Excellence, Hannover Medical School, Carl-Neuberg-Straβe 1, 30625 Hannover, Germany.
| |
Collapse
|
30
|
Directed differentiation of patient-specific induced pluripotent stem cells identifies the transcriptional repression and epigenetic modification of NKX2-5, HAND1, and NOTCH1 in hypoplastic left heart syndrome. PLoS One 2014; 9:e102796. [PMID: 25050861 PMCID: PMC4106834 DOI: 10.1371/journal.pone.0102796] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 06/24/2014] [Indexed: 11/19/2022] Open
Abstract
The genetic basis of hypoplastic left heart syndrome (HLHS) remains unknown, and the lack of animal models to reconstitute the cardiac maldevelopment has hampered the study of this disease. This study investigated the altered control of transcriptional and epigenetic programs that may affect the development of HLHS by using disease-specific induced pluripotent stem (iPS) cells. Cardiac progenitor cells (CPCs) were isolated from patients with congenital heart diseases to generate patient-specific iPS cells. Comparative gene expression analysis of HLHS- and biventricle (BV) heart-derived iPS cells was performed to dissect the complex genetic circuits that may promote the disease phenotype. Both HLHS- and BV heart-derived CPCs were reprogrammed to generate disease-specific iPS cells, which showed characteristic human embryonic stem cell signatures, expressed pluripotency markers, and could give rise to cardiomyocytes. However, HLHS-iPS cells exhibited lower cardiomyogenic differentiation potential than BV-iPS cells. Quantitative gene expression analysis demonstrated that HLHS-derived iPS cells showed transcriptional repression of NKX2-5, reduced levels of TBX2 and NOTCH/HEY signaling, and inhibited HAND1/2 transcripts compared with control cells. Although both HLHS-derived CPCs and iPS cells showed reduced SRE and TNNT2 transcriptional activation compared with BV-derived cells, co-transfection of NKX2-5, HAND1, and NOTCH1 into HLHS-derived cells resulted in synergistic restoration of these promoters activation. Notably, gain- and loss-of-function studies revealed that NKX2-5 had a predominant impact on NPPA transcriptional activation. Moreover, differentiated HLHS-derived iPS cells showed reduced H3K4 dimethylation as well as histone H3 acetylation but increased H3K27 trimethylation to inhibit transcriptional activation on the NKX2-5 promoter. These findings suggest that patient-specific iPS cells may provide molecular insights into complex transcriptional and epigenetic mechanisms, at least in part, through combinatorial expression of NKX2-5, HAND1, and NOTCH1 that coordinately contribute to cardiac malformations in HLHS.
Collapse
|
31
|
Wang J, Sontag D, Cattini PA. Heart-specific expression of FGF-16 and a potential role in postnatal cardioprotection. Cytokine Growth Factor Rev 2014; 26:59-66. [PMID: 25106133 DOI: 10.1016/j.cytogfr.2014.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 10/25/2022]
Abstract
Fibroblast growth factor 16 (FGF-16) was originally cloned from rat heart. Subsequent investigation of mouse FGF-16, including generation of null mice, revealed a specific pattern of expression in the endocardium and epicardium, and role for FGF-16 during embryonic heart development. FGF-16 is expressed mainly in brown adipose tissue during rat embryonic development, but is expressed mainly in the murine heart after birth. There is also an apparent switch from limited endocardial and epicardial expression in the embryo to the myocardium in the perinatal period. The FGF-16 gene and its location on the X chromosome are conserved between human and murine species, and no other member of the FGF family shows this pattern of spatial and temporal expression. The human and murine FGF-16 gene promoter regions also share an equivalent location for TATA sequences, as well as adjacent putative binding sites for transcription factors linked to cardiac expression and response to stress. Recent evidence has implicated nonsense mutation of FGF-16 with increased cardiovascular risk, and FGF-16 supplementation with cardioprotection. Here we review the important role of FGF-16 in embryonic heart development, its gene regulation, and evidence for FGF-16 as an endogenous and exogenous cardiac-specific and protective factor in the postnatal heart. Moreover, given the conservation of the FGF-16 gene and its chromosomal location between species, the question of support for a cardiac role in the human population is also considered.
Collapse
Affiliation(s)
- Jie Wang
- Department of Physiology & Pathophysiology, University of Manitoba, Manitoba, Canada.
| | - David Sontag
- Department of Physiology & Pathophysiology, University of Manitoba, Manitoba, Canada
| | - Peter A Cattini
- Department of Physiology & Pathophysiology, University of Manitoba, Manitoba, Canada
| |
Collapse
|
32
|
Clowes C, Boylan MGS, Ridge LA, Barnes E, Wright JA, Hentges KE. The functional diversity of essential genes required for mammalian cardiac development. Genesis 2014; 52:713-37. [PMID: 24866031 PMCID: PMC4141749 DOI: 10.1002/dvg.22794] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 01/04/2023]
Abstract
Genes required for an organism to develop to maturity (for which no other gene can compensate) are considered essential. The continuing functional annotation of the mouse genome has enabled the identification of many essential genes required for specific developmental processes including cardiac development. Patterns are now emerging regarding the functional nature of genes required at specific points throughout gestation. Essential genes required for development beyond cardiac progenitor cell migration and induction include a small and functionally homogenous group encoding transcription factors, ligands and receptors. Actions of core cardiogenic transcription factors from the Gata, Nkx, Mef, Hand, and Tbx families trigger a marked expansion in the functional diversity of essential genes from midgestation onwards. As the embryo grows in size and complexity, genes required to maintain a functional heartbeat and to provide muscular strength and regulate blood flow are well represented. These essential genes regulate further specialization and polarization of cell types along with proliferative, migratory, adhesive, contractile, and structural processes. The identification of patterns regarding the functional nature of essential genes across numerous developmental systems may aid prediction of further essential genes and those important to development and/or progression of disease. genesis 52:713–737, 2014.
Collapse
Affiliation(s)
- Christopher Clowes
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, United Kingdom
| | | | | | | | | | | |
Collapse
|
33
|
Madonna R, Geng YJ, Bolli R, Rokosh G, Ferdinandy P, Patterson C, De Caterina R. Co-activation of nuclear factor-κB and myocardin/serum response factor conveys the hypertrophy signal of high insulin levels in cardiac myoblasts. J Biol Chem 2014; 289:19585-98. [PMID: 24855642 DOI: 10.1074/jbc.m113.540559] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Hyperinsulinemia contributes to cardiac hypertrophy and heart failure in patients with the metabolic syndrome and type 2 diabetes. Here, high circulating levels of tumor necrosis factor (TNF)-α may synergize with insulin in signaling inflammation and cardiac hypertrophy. We tested whether high insulin affects activation of TNF-α-induced NF-κB and myocardin/serum response factor (SRF) to convey hypertrophy signaling in cardiac myoblasts. In canine cardiac myoblasts, treatment with high insulin (10(-8) to 10(-7) m) for 0-24 h increased insulin receptor substrate (IRS)-1 phosphorylation at Ser-307, decreased protein levels of chaperone-associated ubiquitin (Ub) E3 ligase C terminus of heat shock protein 70-interacting protein (CHIP), increased SRF activity, as well as β-myosin heavy chain (MHC) and myocardin expressions. Here siRNAs to myocardin or NF-κB, as well as CHIP overexpression prevented (while siRNA-mediated CHIP disruption potentiated) high insulin-induced SR element (SRE) activation and β-MHC expression. Insulin markedly potentiated TNF-α-induced NF-κB activation. Compared with insulin alone, insulin+TNF-α increased SRF/SRE binding and β-MHC expression, which was reversed by the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and by NF-κB silencing. In the hearts of db/db diabetic mice, in which Akt phosphorylation was decreased, p38MAPK, Akt1, and IRS-1 phosphorylation at Ser-307 were increased, together with myocardin expression as well as SRE and NF-κB activities. In response to high insulin, cardiac myoblasts increase the expression or the promyogenic transcription factors myocardin/SRF in a CHIP-dependent manner. Insulin potentiates TNF-α in inducing NF-κB and SRF/SRE activities. In hyperinsulinemic states, myocardin may act as a nuclear effector of insulin, promoting cardiac hypertrophy.
Collapse
Affiliation(s)
- Rosalinda Madonna
- From the Texas Heart Institute and University of Texas Medical School in Houston, Houston, Texas 77030, the Institute of Cardiology, and Center of Excellence on Aging, "G. d'Annunzio" University, 66100 Chieti, Italy
| | - Yong-Jian Geng
- From the Texas Heart Institute and University of Texas Medical School in Houston, Houston, Texas 77030
| | - Roberto Bolli
- the Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky 40202
| | - Gregg Rokosh
- the Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky 40202
| | - Peter Ferdinandy
- the Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1085 Budapest, Hungary, and
| | - Cam Patterson
- the Center for Molecular Cardiology, The University of Texas Medical Branch at Galveston, Galveston, Texas 77555
| | - Raffaele De Caterina
- the Institute of Cardiology, and Center of Excellence on Aging, "G. d'Annunzio" University, 66100 Chieti, Italy,
| |
Collapse
|
34
|
Transcriptional Regulation of cGMP-Dependent Protein Kinase II (cGK-II) in Chondrocytes. Biosci Biotechnol Biochem 2014; 74:44-9. [DOI: 10.1271/bbb.90529] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
Reineke EL, Benham A, Soibam B, Stashi E, Taegtmeyer H, Entman ML, Schwartz RJ, O'Malley BW. Steroid receptor coactivator-2 is a dual regulator of cardiac transcription factor function. J Biol Chem 2014; 289:17721-31. [PMID: 24811170 DOI: 10.1074/jbc.m113.539908] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We have previously demonstrated the potential role of steroid receptor coactivator-2 (SRC-2) as a co-regulator in the transcription of critical molecules modulating cardiac function and metabolism in normal and stressed hearts. The present study seeks to extend the previous information by demonstrating SRC-2 fulfills this role by serving as a critical coactivator for the transcription and activity of critical transcription factors known to control cardiac growth and metabolism as well as in their downstream signaling. This knowledge broadens our understanding of the mechanism by which SRC-2 acts in normal and stressed hearts and allows further investigation of the transcriptional modifications mediating different types and degrees of cardiac stress. Moreover, the genetic manipulation of SRC-2 in this study is specific for the heart and thereby eliminating potential indirect effects of SRC-2 deletion in other organs. We have shown that SRC-2 is critical to transcriptional control modulated by MEF2, GATA-4, and Tbx5, thereby enhancing gene expression associated with cardiac growth. Additionally, we describe SRC-2 as a novel regulator of PPARα expression, thus controlling critical steps in metabolic gene expression. We conclude that through regulation of cardiac transcription factor expression and activity, SRC-2 is a critical transcriptional regulator of genes important for cardiac growth, structure, and metabolism, three of the main pathways altered during the cardiac stress response.
Collapse
Affiliation(s)
- Erin L Reineke
- From the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Ashley Benham
- the Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas 77030
| | - Benjamin Soibam
- the Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas 77030
| | - Erin Stashi
- From the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Heinrich Taegtmeyer
- the Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, Houston, Texas 77030
| | - Mark L Entman
- the Department of Medicine, Division of Cardiovascular Sciences, Baylor College of Medicine, Houston, Texas 77030, and
| | - Robert J Schwartz
- the Stem Cell Engineering Department, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas 77030, the Department of Biology and Biochemistry, University of Houston, Houston, Texas 77004
| | - Bert W O'Malley
- From the Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030,
| |
Collapse
|
36
|
Brody MJ, Cho E, Mysliwiec MR, Kim TG, Carlson CD, Lee KH, Lee Y. Lrrc10 is a novel cardiac-specific target gene of Nkx2-5 and GATA4. J Mol Cell Cardiol 2013; 62:237-46. [PMID: 23751912 PMCID: PMC3940241 DOI: 10.1016/j.yjmcc.2013.05.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/11/2013] [Accepted: 05/30/2013] [Indexed: 10/26/2022]
Abstract
Cardiac gene expression is precisely regulated and its perturbation causes developmental defects and heart disease. Leucine-rich repeat containing 10 (Lrrc10) is a cardiac-specific factor that is crucial for proper cardiac development and deletion of Lrrc10 in mice results in dilated cardiomyopathy. However, the mechanisms regulating Lrrc10 expression in cardiomyocytes remain unknown. Therefore, we set out to determine trans-acting factors and cis-elements critical for mediating Lrrc10 expression. We identify Lrrc10 as a transcriptional target of Nkx2-5 and GATA4. The Lrrc10 promoter region contains two highly conserved cardiac regulatory elements, which are functional in cardiomyocytes but not in fibroblasts. In vivo, Nkx2-5 and GATA4 endogenously occupy the proximal and distal cardiac regulatory elements of Lrrc10 in the heart. Moreover, embryonic hearts of Nkx2-5 knockout mice have dramatically reduced expression of Lrrc10. These data demonstrate the importance of Nkx2-5 and GATA4 in regulation of Lrrc10 expression in vivo. The proximal cardiac regulatory element located at around -200bp is synergistically activated by Nkx2-5 and GATA4 while the distal cardiac regulatory element present around -3kb requires SRF in addition to Nkx2-5 and GATA4 for synergistic activation. Mutational analyses identify a pair of adjacent Nkx2-5 and GATA binding sites within the proximal cardiac regulatory element that are necessary to induce expression of Lrrc10. In contrast, only the GATA site is functional in the distal regulatory element. Taken together, our data demonstrate that the transcription factors Nkx2-5 and GATA4 cooperatively regulate cardiac-specific expression of Lrrc10.
Collapse
Affiliation(s)
- Matthew J. Brody
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, WI 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, WI 53706, USA
| | - Eunjin Cho
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, WI 53706, USA
- Molecular and Cellular Pharmacology, University of Wisconsin-Madison, WI 53706, USA
| | - Matthew R. Mysliwiec
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, WI 53706, USA
| | - Tae-gyun Kim
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, WI 53706, USA
| | - Clayton D. Carlson
- Department of Biochemistry and the Genome Center of Wisconsin, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kyu-Ho Lee
- Department of Pediatrics, Division of Pediatric Cardiology, Children’s Hospital, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Youngsook Lee
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, WI 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, WI 53706, USA
- Molecular and Cellular Pharmacology, University of Wisconsin-Madison, WI 53706, USA
| |
Collapse
|
37
|
Myocardin-A enhances expression of promyogenic genes without depressing telomerase activity in adipose tissue-derived mesenchymal stem cells. Int J Cardiol 2013; 167:2912-21. [DOI: 10.1016/j.ijcard.2012.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 07/15/2012] [Accepted: 07/21/2012] [Indexed: 01/16/2023]
|
38
|
Clark CD, Zhang B, Lee B, Evans SI, Lassar AB, Lee KH. Evolutionary conservation of Nkx2.5 autoregulation in the second heart field. Dev Biol 2013; 374:198-209. [PMID: 23165293 PMCID: PMC3549048 DOI: 10.1016/j.ydbio.2012.11.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 10/22/2012] [Accepted: 11/09/2012] [Indexed: 11/28/2022]
Abstract
The cardiac homeobox gene Nkx2.5 plays a key and dosage-sensitive role in the differentiation of outflow tract and right ventricle from progenitors of the second heart field (SHF) and Nkx2.5 mutation is strongly associated with human outflow tract congenital heart disease (OFT CHD). Therefore defining the regulatory mechanisms controlling Nkx2.5 expression in SHF populations serves an important function in understanding the etiology of complex CHD. Through a comparative analysis of regulatory elements controlling SHF expression of Nkx2.5 in the chicken and mouse, we have found evidence that Nkx2.5 autoregulation is important for maintaining Nkx2.5 expression during SHF differentiation in both species. However the mechanism of Nkx2.5 maintenance differs between placental mammals and non-mammalian vertebrates: in chick Nkx2.5 binds directly to a genomic enhancer element that is required to maintain Nkx2.5 expression in the SHF. In addition, it is likely that this is true in other non-mammalian vertebrates given that they possess a similar genomic organization. By contrast, in placental mammals, Nkx2.5 autoregulation in the SHF functions indirectly through Mef2c. These data underscore a tight relationship in mammals between Nkx2.5 and Mef2c in SHF transcriptional regulation, and highlight the potential for evolutionary cis-regulatory analysis to identify core, conserved components of the gene networks controlling heart development.
Collapse
Affiliation(s)
- Christopher D. Clark
- Regenerative Medicine, Cell Biology and Anatomy Department, Medical University of South Carolina, Charleston, SC
| | - Boding Zhang
- Regenerative Medicine, Cell Biology and Anatomy Department, Medical University of South Carolina, Charleston, SC
| | - Benjamin Lee
- Regenerative Medicine, Cell Biology and Anatomy Department, Medical University of South Carolina, Charleston, SC
| | - Samuel I. Evans
- Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Andrew B. Lassar
- Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Kyu-Ho Lee
- Regenerative Medicine, Cell Biology and Anatomy Department, Medical University of South Carolina, Charleston, SC
- Department of Pediatrics, Division of Pediatric Cardiology, Children’s Hospital, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
39
|
Abstract
Differentiated adult cardiomyocytes (CMs) lack significant regenerative potential, which is one reason why degenerative heart diseases are the leading cause of death in the western world. For future cardiac repair, stem cell-based therapeutic strategies may become alternatives to donor heart transplantation. The principle of reprogramming adult terminally differentiated cells (iPSC) had a major impact on stem cell biology. One can now generate autologous pluripotent cells that highly resemble embryonic stem cells (ESC) and that are ethically inoffensive as opposed to human ESC. Yet, due to genetic and epigenetic aberrations arising during the full reprogramming process, it is questionable whether iPSC will enter the clinic in the near future. Therefore, the recent achievement of directly reprogramming fibroblasts into cardiomyocytes via a milder approach, thereby avoiding an initial pluripotent state, may become of great importance. In addition, various clinical scenarios will depend on the availability of specific cardiac cellular subtypes, for which a first step was achieved via our own programming approach to achieve cardiovascular cell subtypes. In this review, we discuss recent progress in the cardiovascular stem cell field addressing the above mentioned aspects.
Collapse
Affiliation(s)
- Robert David
- 1st Medical Department, University of Munich, Campus Grosshadern, Munich, Germany
| | | |
Collapse
|
40
|
Hwang JTK, Kelly GM. GATA6 and FOXA2 regulate Wnt6 expression during extraembryonic endoderm formation. Stem Cells Dev 2012; 21:3220-32. [PMID: 22607194 DOI: 10.1089/scd.2011.0492] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
One of the earliest epithelial-to-mesenchymal transitions in mouse embryogenesis involves the differentiation of inner cell mass cells into primitive and then into parietal endoderm. These processes can be recapitulated in vitro using F9 teratocarcinoma cells, which differentiate into primitive endoderm when treated with retinoic acid (RA) and into parietal endoderm with subsequent treatment with dibutyryl cyclic adenosine monophosphate (db-cAMP). Our previous work on how primitive endoderm develops revealed that the Wnt6 gene is upregulated by RA, leading to the activation of the canonical WNT-β-catenin pathway. The mechanism by which Wnt6 is regulated was not determined, but in silico analysis of the human WNT6 promoter region had suggested that the GATA6 and FOXA2 transcription factors might be involved [1]. Subsequent analysis determined that both Gata6 and Foxa2 mRNA are upregulated in F9 cells treated with RA or RA and db-cAMP. More specifically, overexpression of Gata6 or Foxa2 alone induced molecular and morphological markers of primitive endoderm, which occurred concomitantly with the upregulation of the Wnt6 gene. Gata6- or Foxa2-overexpressing cells were also found to have increased levels in T-cell factor (TCF)-dependent transcription, and when these cells were treated with db-cAMP, they developed into parietal endoderm. Chromatin immunoprecipitation analysis revealed that GATA6 and FOXA2 were bound to the Wnt6 promoter, and overexpression studies showed that these transcription factors were sufficient to switch on the gene expression of a Wnt6 reporter construct. Together, these results provide evidence for the direct regulation of Wnt6 that leads to the activation of the canonical WNT-β-catenin pathway and subsequent induction of primitive extraembryonic endoderm.
Collapse
Affiliation(s)
- Jason T K Hwang
- Molecular Genetics Unit, Department of Biology, Child Health Research Institute, Western University, London, Ontario, Canada
| | | |
Collapse
|
41
|
Mooney E, Mackle JN, Blond DJP, O'Cearbhaill E, Shaw G, Blau WJ, Barry FP, Barron V, Murphy JM. The electrical stimulation of carbon nanotubes to provide a cardiomimetic cue to MSCs. Biomaterials 2012; 33:6132-9. [PMID: 22681974 DOI: 10.1016/j.biomaterials.2012.05.032] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 05/15/2012] [Indexed: 12/29/2022]
Abstract
Once damaged, cardiac muscle has little intrinsic repair capability due to the poor regeneration potential of remaining cardiomyocytes. One method of overcoming this issue is to deliver functional cells to the injured myocardium to promote repair. To address this limitation we sought to test the hypothesis that electroactive carbon nanotubes (CNT) could be employed to direct mesenchymal stem cell (MSC) differentiation towards a cardiomyocyte lineage. Using a two-pronged approach, MSCs exposed to medium containing CNT and MSCs seeded on CNT based polylactic acid scaffolds were electrically stimulated in an electrophysiological bioreactor. After electrical stimulation the cells reoriented perpendicular to the direction of the current and adopted an elongated morphology. Using qPCR, an upregulation in a range of cardiac markers was detected, the greatest of which was observed for cardiac myosin heavy chain (CMHC), where a 40-fold increase was observed for the electrically stimulated cells after 14 days, and a 12-fold increase was observed for the electrically stimulated cells seeded on the PLA scaffolds after 10 days. Differentiation towards a cardioprogenitor cell was more evident from the western blot analysis, where upregulation of Nkx2.5, GATA-4, cardiac troponin t (CTT) and connexin43 (C43) was seen to occur. This was echoed in immunofluorescent staining, where increased levels of CTT, CMHC and C43 protein expression were observed after electrical stimulation for both cells and cell-seeded scaffolds. More interestingly, there was evidence of increased cross talk between the cells as shown by the pattern of C43 staining after electrical stimulation. These results establish a paradigm for nanoscale biomimetic cues that can be readily translated to other electroactive tissue repair applications.
Collapse
Affiliation(s)
- Emma Mooney
- Regenerative Medicine Institute (REMEDI), Orbsen Building, National University of Ireland, Galway, University Road, Galway, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Belaguli NS, Zhang M, Garcia AH, Berger DH. PIAS1 is a GATA4 SUMO ligase that regulates GATA4-dependent intestinal promoters independent of SUMO ligase activity and GATA4 sumoylation. PLoS One 2012; 7:e35717. [PMID: 22539995 PMCID: PMC3334497 DOI: 10.1371/journal.pone.0035717] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 03/20/2012] [Indexed: 01/12/2023] Open
Abstract
GATA4 confers cell type-specific gene expression on genes expressed in cardiovascular, gastro-intestinal, endocrine and neuronal tissues by interacting with various ubiquitous and cell-type-restricted transcriptional regulators. By using yeast two-hybrid screening approach, we have identified PIAS1 as an intestine-expressed GATA4 interacting protein. The physical interaction between GATA4 and PIAS1 was confirmed in mammalian cells by coimmunoprecipitation and two-hybrid analysis. The interacting domains were mapped to the second zinc finger and the adjacent C-terminal basic region of GATA4 and the RING finger and the adjoining C-terminal 60 amino acids of PIAS1. PIAS1 and GATA4 synergistically activated IFABP and SI promoters but not LPH promoters suggesting that PIAS1 differentially activates GATA4 targeted promoters. In primary murine enterocytes PIAS1 was recruited to the GATA4-regulated IFABP promoter. PIAS1 promoted SUMO-1 modification of GATA4 on lysine 366. However, sumoylation was not required for the nuclear localization and stability of GATA4. Further, neither GATA4 sumoylation nor the SUMO ligase activity of PIAS1 was required for coactivation of IFABP promoter by GATA4 and PIAS1. Together, our results demonstrate that PIAS1 is a SUMO ligase for GATA4 that differentially regulates GATA4 transcriptional activity independent of SUMO ligase activity and GATA4 sumoylation.
Collapse
Affiliation(s)
- Narasimhaswamy S. Belaguli
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, Texas, United States of America
- * E-mail: (NSB); (DHB)
| | - Mao Zhang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, Texas, United States of America
| | - Andres-Hernandez Garcia
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, Texas, United States of America
| | - David H. Berger
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, Texas, United States of America
- * E-mail: (NSB); (DHB)
| |
Collapse
|
43
|
Junion G, Spivakov M, Girardot C, Braun M, Gustafson E, Birney E, Furlong E. A Transcription Factor Collective Defines Cardiac Cell Fate and Reflects Lineage History. Cell 2012; 148:473-86. [DOI: 10.1016/j.cell.2012.01.030] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 08/16/2011] [Accepted: 01/17/2012] [Indexed: 11/28/2022]
|
44
|
Abstract
Transcription factors regulate formation and function of the heart, and perturbation of transcription factor expression and regulation disrupts normal heart structure and function. Multiple mechanisms regulate the level and locus-specific activity of transcription factors, including transcription, translation, subcellular localization, posttranslational modifications, and context-dependent interactions with other transcription factors, chromatin remodeling enzymes, and epigenetic regulators. The zinc finger transcription factor GATA4 is among the best-studied cardiac transcriptional factors. This review focuses on molecular mechanisms that regulate GATA4 transcriptional activity in the cardiovascular system, providing a framework to investigate and understand the molecular regulation of cardiac gene transcription by other transcription factors.
Collapse
|
45
|
Lee S, Lee JW, Lee SK. UTX, a histone H3-lysine 27 demethylase, acts as a critical switch to activate the cardiac developmental program. Dev Cell 2011; 22:25-37. [PMID: 22192413 DOI: 10.1016/j.devcel.2011.11.009] [Citation(s) in RCA: 199] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 10/26/2011] [Accepted: 11/21/2011] [Indexed: 10/14/2022]
Abstract
The removal of histone H3 lysine27 (H3K27) trimethylation mark is important for the robust induction of many cell type-specific genes during differentiation. Here we show that UTX, a H3K27 demethylase, acts as a critical switch to promote a cardiac-specific gene program. UTX-deficient ESCs failed to develop heart-like rhythmic contractions under a cardiac differentiation condition. UTX-deficient mice show severe defects in heart development and embryonic lethality. We found that UTX is recruited to cardiac-specific enhancers by associating with core cardiac transcription factors and demethylates H3K27 residues in cardiac genes. In addition, UTX facilitates the recruitment of Brg1 to the cardiac-specific enhancers. Together, our data reveal key roles for UTX in a timely transition from poised to active chromatin in cardiac genes during heart development and a fundamental mechanism by which a H3K27 demethylase triggers tissue-specific chromatin changes.
Collapse
Affiliation(s)
- Seunghee Lee
- Pediatric Neuroscience Research Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | |
Collapse
|
46
|
Abstract
NK-like (NKL) homeobox genes code for transcription factors, which can act as key regulators in fundamental cellular processes. NKL genes have been implicated in divergent types of cancer. In this review, we summarize the involvement of NKL genes in cancer and leukemia in particular. NKL genes can act as tumor-suppressor genes and as oncogenes, depending on tissue type. Aberrant expression of NKL genes is especially common in T-cell acute lymphoblastic leukemia (T-ALL). In T-ALL, 8 NKL genes have been reported to be highly expressed in specific T-ALL subgroups, and in ~30% of cases, high expression is caused by chromosomal rearrangement of 1 of 5 NKL genes. Most of these NKL genes are normally not expressed in T-cell development. We hypothesize that the NKL genes might share a similar downstream effect that promotes leukemogenesis, possibly due to mimicking a NKL gene that has a physiological role in early hematopoietic development, such as HHEX. All eight NKL genes posses a conserved Eh1 repressor motif, which has an important role in regulating downstream targets in hematopoiesis and possibly in leukemogenesis as well. Identification of a potential common leukemogenic NKL downstream pathway will provide a promising subject for future studies.
Collapse
|
47
|
Orza A, Soritau O, Olenic L, Diudea M, Florea A, Rus Ciuca D, Mihu C, Casciano D, Biris AS. Electrically conductive gold-coated collagen nanofibers for placental-derived mesenchymal stem cells enhanced differentiation and proliferation. ACS NANO 2011; 5:4490-4503. [PMID: 21609025 DOI: 10.1021/nn1035312] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Gold-coated collagen nanofibers (GCNFs) were produced by a single-step reduction process and used for the growth and differentiation of human adult stem cells. The nanomaterials were characterized by a number of analytical techniques including electron microscopy and spectroscopy. They were found to be biocompatible and to improve the myocardial and neuronal differentiation process of the mesenchymal stem cells isolated from the placental chorionic component. The expression of specific differentiation markers (atrium, natriuretic peptide, actin F and actin monomer, glial fibrilary acidic protein, and neurofilaments) was investigated by immunocytochemistry.
Collapse
Affiliation(s)
- Anamaria Orza
- Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, Cluj-Napoca, Romania.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Miranda-Carboni GA, Guemes M, Bailey S, Anaya E, Corselli M, Peault B, Krum SA. GATA4 regulates estrogen receptor-alpha-mediated osteoblast transcription. Mol Endocrinol 2011; 25:1126-36. [PMID: 21566084 DOI: 10.1210/me.2010-0463] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Estrogens regulate osteoblast differentiation and mineralization. We identified GATA4 as a transcription factor expressed in osteoblasts and directly regulated by 17β-estradiol in this cell type but not in breast cancer cells, another estrogen-responsive tissue. Chromatin immunoprecipitation sequencing (chromatin immunoprecipitation sequencing) reveals that estrogen receptor α (ERα) binds to chromatin near GATA4 at five different enhancers. GATA4 and ERα are both recruited to ERα binding sites near genes that are specifically expressed in osteoblasts and control osteoblast differentiation. Maximal binding of GATA4 precedes ERα binding, and GATA4 is necessary for histone 3 lysine 4 dimethylation at ERα binding sites, suggesting that GATA4 is a pioneer factor for ERα. As such, knockdown of GATA4 reduced recruitment of ERα to DNA. Our study illustrates that GATA4 is a pioneer factor for ERα recruitment to osteoblast-specific enhancers.
Collapse
Affiliation(s)
- Gustavo A Miranda-Carboni
- Department of Obstetrics and Gynecology, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Kaltenbrun E, Tandon P, Amin NM, Waldron L, Showell C, Conlon FL. Xenopus: An emerging model for studying congenital heart disease. ACTA ACUST UNITED AC 2011; 91:495-510. [PMID: 21538812 DOI: 10.1002/bdra.20793] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 01/18/2011] [Accepted: 01/28/2011] [Indexed: 02/02/2023]
Abstract
Congenital heart defects affect nearly 1% of all newborns and are a significant cause of infant death. Clinical studies have identified a number of congenital heart syndromes associated with mutations in genes that are involved in the complex process of cardiogenesis. The African clawed frog, Xenopus, has been instrumental in studies of vertebrate heart development and provides a valuable tool to investigate the molecular mechanisms underlying human congenital heart diseases. In this review, we discuss the methodologies that make Xenopus an ideal model system to investigate heart development and disease. We also outline congenital heart conditions linked to cardiac genes that have been well studied in Xenopus and describe some emerging technologies that will further aid in the study of these complex syndromes.
Collapse
Affiliation(s)
- Erin Kaltenbrun
- University of North Carolina McAllister Heart Institute, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
50
|
Co-occupancy by multiple cardiac transcription factors identifies transcriptional enhancers active in heart. Proc Natl Acad Sci U S A 2011; 108:5632-7. [PMID: 21415370 DOI: 10.1073/pnas.1016959108] [Citation(s) in RCA: 273] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Identification of genomic regions that control tissue-specific gene expression is currently problematic. ChIP and high-throughput sequencing (ChIP-seq) of enhancer-associated proteins such as p300 identifies some but not all enhancers active in a tissue. Here we show that co-occupancy of a chromatin region by multiple transcription factors (TFs) identifies a distinct set of enhancers. GATA-binding protein 4 (GATA4), NK2 transcription factor-related, locus 5 (NKX2-5), T-box 5 (TBX5), serum response factor (SRF), and myocyte-enhancer factor 2A (MEF2A), here referred to as "cardiac TFs," have been hypothesized to collaborate to direct cardiac gene expression. Using a modified ChIP-seq procedure, we defined chromatin occupancy by these TFs and p300 genome wide and provided unbiased support for this hypothesis. We used this principle to show that co-occupancy of a chromatin region by multiple TFs can be used to identify cardiac enhancers. Of 13 such regions tested in transient transgenic embryos, seven (54%) drove cardiac gene expression. Among these regions were three cardiac-specific enhancers of Gata4, Srf, and swItch/sucrose nonfermentable-related, matrix-associated, actin-dependent regulator of chromatin, subfamily d, member 3 (Smarcd3), an epigenetic regulator of cardiac gene expression. Multiple cardiac TFs and p300-bound regions were associated with cardiac-enriched genes and with functional annotations related to heart development. Importantly, the large majority (1,375/1,715) of loci bound by multiple cardiac TFs did not overlap loci bound by p300. Our data identify thousands of prospective cardiac regulatory sequences and indicate that multiple TF co-occupancy of a genomic region identifies developmentally relevant enhancers that are largely distinct from p300-associated enhancers.
Collapse
|