Click here to close
Hello! We notice that you are using Internet Explorer, which is not supported by Xenbase and may cause the site to display incorrectly.
We suggest using a current version of Chrome,
FireFox, or Safari.
Mech Dev
2014 Nov 01;134:31-41. doi: 10.1016/j.mod.2014.09.001.
Show Gene links
Show Anatomy links
Carboxy terminus of GATA4 transcription factor is required for its cardiogenic activity and interaction with CDK4.
Gallagher JM
,
Yamak A
,
Kirilenko P
,
Black S
,
Bochtler M
,
Lefebvre C
,
Nemer M
,
Latinkić BV
.
???displayArticle.abstract??? GATA4-6 transcription factors regulate numerous aspects of development and homeostasis in multiple tissues of mesodermal and endodermal origin. In the heart, the best studied of these factors, GATA4, has multiple distinct roles in cardiac specification, differentiation, morphogenesis, hypertrophy and survival. To improve understanding of how GATA4 achieves its numerous roles in the heart, here we have focused on the carboxy-terminal domain and the residues required for interaction with cofactors FOG2 and Tbx5. We present evidence that the carboxy terminal region composed of amino acids 362-400 is essential for mediating cardiogenesis in Xenopus pluripotent explants and embryos. In contrast, the same region is not required for endoderm-inducing activity of GATA4. Further evidence is presented that the carboxy terminal cardiogenic region of GATA4 does not operate as a generic transcriptional activator. Potential mechanism of action of the carboxy terminal end of GATA4 is provided by the results showing physical and functional interaction with CDK4, including the enhancement of cardiogenic activity of GATA4 by CDK4. These results establish CDK4 as a GATA4 partner in cardiogenesis. The interactions of GATA4 with its other well described cofactors Tbx5 and FOG2 are known to be involved in heart morphogenesis, but their requirement for cardiac differentiation is unknown. We report that the mutations that disrupt interactions of GATA4 with Tbx5 and FOG2, G295S and V217G, respectively, do not impair cardiogenic activity of GATA4. These findings add support to the view that distinct roles of GATA4 in the heart are mediated by different determinants of the protein. Finally, we show that the rat GATA4 likely induces cardiogenesis cell autonomously or directly as it does not require activity of endodermal transcription factor Sox17, a GATA4 target gene that induces cardiogenesis non-cell autonomously.
???displayArticle.pubmedLink???
25241353
???displayArticle.pmcLink???PMC4259525 ???displayArticle.link???Mech Dev ???displayArticle.grants???[+]
Fig. 1 â Carboxy-terminal domain (362â440) of GATA4 is required for cardiogenesis. (A) Schematic representation of GATA4
structure showing transcriptional activation domains (TAD), Zn fingers (Zn), nuclear localisation signal (nls) and the
position of mutations tested in this study. Right â sequence of the C-terminal region of GATA4 (rat, NP_653331; Xenopus
NP_001085355) and GATA5 (rat NP_001019487; Xenopus NP_001081962), showing poor conservation in these cardiogenic
factors. (B) Deletion of C-terminal 78 amino acids abolishes cardiogenic activity of GATA4. Animal caps injected with wt or
1â361 mutant rGATA4 were tested for expression of cardiomyocyte-specific genes myl7 and myh6 (synonyms: Myosin Light
Chain 2 and Myosin Heavy Chain α, respectively) at st.34 and for levels of GATA4 protein at st. 10 (lower panel). Note that
the effect of this deletion is negligible on endothelial marker aplnr (apelin receptor; synonym- msr) and is less pronounced
on hba3 (haemoglobin alpha 3; synonym- αglobin), a blood marker. odc1- loading control (expression of ornithine
decarboxylase 1). (C) Nuclear localisation of 1â361 rGATA4 was determined by immunohistochemistry at st. 7â8. (D) Lack of
activity of 1â361 in vivo. Embryos injected with 1â361 rGATA4 at 8â16 cell-stage in anterior ectodermal precursors were
analysed for myl7 expression by whole mount hybridisation. myl7 expression was only observed in the heart (h) in all
embryos examined (n = 50, from 2 independent experiments). In contrast, cardiogenic GATA4 versions readily induce
ectopic myl7 expression (Figs 3 and 5). (E) Non-cardiogenic 1â361 GATA4 mutant retains gene expression-inducing activity
in animal cap explants. At st. 10 the mutant 1â361 induces endogenous gata4 and (F) at st. 34 pan-endodermal marker
alpha-2 macroglobulin (a2m; synonym â endodermin) is induced (as well as by N-terminal deletions 153â440 and 201â440
non-cardiogenic GATA4 (Gallagher et al., 2012)).
Fig. 2 â CDK4 interacts with C-terminal GATA4 in vitro and enhances cardiogenic activity of GATA4 (A) In vitro translated
35S radio-labelled CDK4 protein was incubated with GST-N-terminal GATA4, GST-C-terminal GATA4 or GST proteins on
glutathione sepharose beads. The bound proteins were resolved by SDS/PAGE and revealed by autoradiography. Note that
CDK4 binds C-terminal GATA4 (1st lane). The experiment is one representative of two. (B) Fold synergy of wild type GATA4
(1â440) or C-terminal deletion (1â361) with CDK4 on GATA-dependent promoter. The C-terminal region is required for
synergy. 10 ng of GATA4 and 100 ng of CDK4 expression vectors were used. The experiment was performed twice.
* p < 0.005 of GATA4 1â440/CDK4 fold synergy vs GATA4 1â440 using Studentâs t test. (C) NIH3T3 cells were transiently
transfected with GATA-luc and increasing doses of the indicated GATA4 expression vector (5, 25, 50 and 100 ng) with our
without treatment with 3 M CDK4 inhibitor. The cells were treated the following day after transfection and kept for 18 hrs.
Note that 1â361 GATA4 activation is not inhibited by CDK4. Experiment was performed twice. # p < 0.001 of GATA4 1â440
with CDK4 inhibitor vs. GATA4 1â440 using two-way ANOVA. (D) CDK4 enhances cardiogenic activity of GATA4. Animal cap
explants injected with indicated capped mRNAs (400 and 100 pg/embryo of CDK4 and CyclinD2 (CycD2), respectively;
300 pg/embryo of rGATA4 (suboptimal dose)) were analysed for expression of cardiomyocyte marker myl7 at st. 34.
Fig. 3 â Deletion of 15 or 40 C-terminal amino acids does not block cardiogenic activity of GATA4. (A) Expression of myh6
and odc1 in st. 34 animal caps expressing wt, 1â400, 1â425 and 1â425 mPKC (S419A; S420A) versions of rGATA4.Western
blot analysis (lower panel) of protein levels at st. 10. (B) Nuclear localisation of 1â400, 1â425 and 1â425 rGATA4. (C) Embryos
injected in anterior ectodermal blastomeres at 8â16 cell stage with 1â400, 1â425 and 1â425 were analysed for myl7
expression at st. 34. myl7 was detected in the heart (h) and in ectopic locations (arrows) of representative embryos (out of
40, 4â7 embryos showed ectopic cardiac expression).
Fig. 4 â Non-cardiogenic GATA4 mutants lacking N- or
C-terminus can be partially rescued by a heterologous
transcriptional activator. (A) 1â361 and 201â440 VP fusions
are stronger transcriptional activators than the wt rGATA4.
Embryos were injected at 1- or 2-cell stage with a firefly
luciferase reporter under the control of two GATA sites,
Renilla luciferase plasmid driven by the thymidine kinase
(TK) promoter and indicated mRNAs. Control-DNA alone
sample. Animal caps were excised at st. 8.5 and collected
for analysis 3 hours later. GATA-luc activity represents
relative fold activation (firefly luciferase activity normalised
by Renilla luciferase activity, with control (no mRNA)
sample set at 1). In three separate experiments 201â
440 + VP and 1â361 + VP were found to be considerably
more active than the wt rGATA4, with variable fold
differences but consistent trend of 201â440 + VP and
1â361 + VP showing 5â10 times greater activity than the wt
rGATA4. (B) VP16 minimal activation domain (VP) weakly
rescues cardiogenic activity of 1â361 and 201â440, as
assessed by expression of myl7 and myh6. (C) In contrast,
the ability of 1â361 and 201â440 VP fusions to induce the
expression of endothelial marker aplnr and a blood marker
hba3 is comparable to the wt rGATA4.
Fig. 5 â Cardiogenic activity of GATA4 does not require residues which are essential for interaction with FOG2 or Tbx5.
(A) E215D, V217G and G295S mutations do not affect cardiogenesis induced by GATA4 in animal pole explants, as assessed
by expression of myh6 at st. 34. Bottom panel âWestern blot analysis of rGATA4 variants in animal caps collected at st. 10.
(B) Nuclear localisation of E215, V217G and G295S GATA4 mutants. (C) Ectopic myl7 expression (arrows) induced by
indicated mutants in representative embryos (out of 50, 4â10 embryos showed ectopic myl7 expression). (D) In addition to
being able to efficiently induce cardiac differentiation markers myh6 and myl7, V217G and G295S mutants are
indistinguishable from the wt rGATA4 in their ability to induce liver (nr1h5; synonym-for1), endothelium (aplnr) and blood
(hba3) markers. (E) Model of the C-terminal Zn finger of GATA4 with G295S mutation. The key side chains, Ser295 and
Arg282, are indicated. The model shows the clash that would occur with the mutation in the absence of backbone or side
chain adaptations to resolve it.
Fig. 6 â Non-cell autonomous induction of cardiogenesis in animal cap explants by Sox17. Animal caps derived from
cardiac actinâGFP (CA-GFP) embryos injected with 200 pg of Sox17 mRNA or uninjected (AC) were conjugated and cultured
until st. 34. Different animal caps in each conjugate were distinguished by the presence of rhodamine-dextran in one of
them. (AâD) Bright field images of representative examples of indicated types of explants. (AââDâ) Corresponding red and
green fluorescence images (merged), showing contribution of rhodamine-dextran injected animal caps and the presence or
absence of striated muscles (CA-GFP; Latinkic et al., 2003). CA-GFP activity was only detected in naïve, uninjected part of
Sox17+AC conjugates, indicating non-cell autonomous cardiac induction. The number of conjugates showing the
represented phenotype and the total number of conjugates are shown in the lower right corner (AâD). (E) Activity of Sox17
is shown by induction of endoderm marker a2m in st. 10 animal cap explants. (F) Inhibition of Sox17 function by
Sox17::EnR does not block cardiogenesis by rGATA4. rGATA4 (400 pg/embryo) and Sox17::EnR (200 pg per embryo) were
uniformly injected at 1- or 2-cell stage and RT-PCR was performed at st. 34 for myl7 and odc1 loading control.
Aries,
Essential role of GATA-4 in cell survival and drug-induced cardiotoxicity.
2004, Pubmed
Aries,
Essential role of GATA-4 in cell survival and drug-induced cardiotoxicity.
2004,
Pubmed
Arnold,
The SWISS-MODEL workspace: a web-based environment for protein structure homology modelling.
2006,
Pubmed
Brickman,
Hex is a transcriptional repressor that contributes to anterior identity and suppresses Spemann organiser function.
2000,
Pubmed
,
Xenbase
Burgess,
Inhibition of S/G2 phase CDK4 reduces mitotic fidelity.
2006,
Pubmed
Chambers,
The RSRF/MEF2 protein SL1 regulates cardiac muscle-specific transcription of a myosin light-chain gene in Xenopus embryos.
1994,
Pubmed
,
Xenbase
Charron,
Tissue-specific GATA factors are transcriptional effectors of the small GTPase RhoA.
2001,
Pubmed
Chomczynski,
Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction.
1987,
Pubmed
Crispino,
Proper coronary vascular development and heart morphogenesis depend on interaction of GATA-4 with FOG cofactors.
2001,
Pubmed
Frey,
Cardiac hypertrophy: the good, the bad, and the ugly.
2003,
Pubmed
Fu,
Direct reprogramming of human fibroblasts toward a cardiomyocyte-like state.
2013,
Pubmed
Gallagher,
Dissociation of cardiogenic and postnatal myocardial activities of GATA4.
2012,
Pubmed
,
Xenbase
Garg,
GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5.
2003,
Pubmed
,
Xenbase
Gilchrist,
Defining a large set of full-length clones from a Xenopus tropicalis EST project.
2004,
Pubmed
,
Xenbase
Grépin,
A hormone-encoding gene identifies a pathway for cardiac but not skeletal muscle gene transcription.
1994,
Pubmed
,
Xenbase
Grépin,
Enhanced cardiogenesis in embryonic stem cells overexpressing the GATA-4 transcription factor.
1997,
Pubmed
Haworth,
GATA4 and GATA5 are essential for heart and liver development in Xenopus embryos.
2008,
Pubmed
,
Xenbase
Holtzinger,
Gata4 regulates the formation of multiple organs.
2005,
Pubmed
Hudson,
Xsox17alpha and -beta mediate endoderm formation in Xenopus.
1997,
Pubmed
,
Xenbase
Ieda,
Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors.
2010,
Pubmed
Kikuchi,
Primary contribution to zebrafish heart regeneration by gata4(+) cardiomyocytes.
2010,
Pubmed
Latinkić,
Induction of cardiomyocytes by GATA4 in Xenopus ectodermal explants.
2003,
Pubmed
,
Xenbase
Latinkić,
Distinct enhancers regulate skeletal and cardiac muscle-specific expression programs of the cardiac alpha-actin gene in Xenopus embryos.
2002,
Pubmed
,
Xenbase
Liang,
The transcription factors GATA4 and GATA6 regulate cardiomyocyte hypertrophy in vitro and in vivo.
2001,
Pubmed
Liang,
The transcription factor GATA4 is activated by extracellular signal-regulated kinase 1- and 2-mediated phosphorylation of serine 105 in cardiomyocytes.
2001,
Pubmed
Liu,
Sox17 is essential for the specification of cardiac mesoderm in embryonic stem cells.
2007,
Pubmed
Misra,
Congenital heart disease-causing Gata4 mutation displays functional deficits in vivo.
2012,
Pubmed
Molkentin,
Cytoplasmic signaling pathways that regulate cardiac hypertrophy.
2001,
Pubmed
Morrisey,
GATA-4 activates transcription via two novel domains that are conserved within the GATA-4/5/6 subfamily.
1997,
Pubmed
,
Xenbase
Movassagh,
Cardiac differentiation in Xenopus requires the cyclin-dependent kinase inhibitor, p27Xic1.
2008,
Pubmed
,
Xenbase
Nam,
Reprogramming of human fibroblasts toward a cardiac fate.
2013,
Pubmed
Nemer,
A novel mutation in the GATA4 gene in patients with Tetralogy of Fallot.
2006,
Pubmed
Omichinski,
NMR structure of a specific DNA complex of Zn-containing DNA binding domain of GATA-1.
1993,
Pubmed
Pu,
GATA4 is a dosage-sensitive regulator of cardiac morphogenesis.
2004,
Pubmed
Qian,
In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes.
2012,
Pubmed
Samuel,
Early activation of FGF and nodal pathways mediates cardiac specification independently of Wnt/beta-catenin signaling.
2009,
Pubmed
,
Xenbase
Takeuchi,
Directed transdifferentiation of mouse mesoderm to heart tissue by defined factors.
2009,
Pubmed
Tevosian,
FOG-2, a cofactor for GATA transcription factors, is essential for heart morphogenesis and development of coronary vessels from epicardium.
2000,
Pubmed
van Berlo,
Serine 105 phosphorylation of transcription factor GATA4 is necessary for stress-induced cardiac hypertrophy in vivo.
2011,
Pubmed
Wang,
Convergence of protein kinase C and JAK-STAT signaling on transcription factor GATA-4.
2005,
Pubmed
Watt,
GATA4 is essential for formation of the proepicardium and regulates cardiogenesis.
2004,
Pubmed
Yamak,
Cyclin D2 is a GATA4 cofactor in cardiogenesis.
2014,
Pubmed
,
Xenbase
Zhou,
Regulation of GATA4 transcriptional activity in cardiovascular development and disease.
2012,
Pubmed