tbx1 represses mef2c gene expression and is correlated ... · research article tbx1 represses mef2c...

7
RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior heart field enhancer Luna Simona Pane 1, * , , Filomena Gabriella Fulcoli 1, , Andrea Cirino 2 , Alessandra Altomonte 1 , Rosa Ferrentino 1 , Marchesa Bilio 1 and Antonio Baldini 1,2,§ ABSTRACT The TBX1 gene is haploinsufficient in 22q11.2 deletion syndrome (22q11.2DS), and genetic evidence from human patients and mouse models points to a major role of this gene in the pathogenesis of this syndrome. Tbx1 can activate and repress transcription, and previous work has shown that one of its functions is to negatively modulate cardiomyocyte differentiation. Tbx1 occupies the anterior heart field (AHF) enhancer of the Mef2c gene, which encodes a key cardiac differentiation transcription factor. Here, we show that increased dosage of Tbx1 correlates with downregulation of Mef2c expression and reduced acetylation of its AHF enhancer in cultured mouse myoblasts. Consistently, 22q11.2DS-derived and in vitro- differentiated human induced pluripotent stem cells (hiPSCs) expressed higher levels of MEF2C and showed increased AHF acetylation, compared with hiPSCs from a healthy donor. Most importantly, we show that in mouse embryos, loss of Tbx1 enhances the expression of the Mef2c-AHF-Cre transgene in a specific region of the splanchnic mesoderm, and in a dosage-dependent manner, providing an in vivo correlate of our cell culture data. These results indicate that Tbx1 regulates the Mef2c AHF enhancer by inducing histone deacetylation. KEY WORDS: Anterior heart field enhancer, Mef2c, Tbx1 INTRODUCTION During development, cardiac progenitors of the second heart field (SHF) are recruited or incorporated into the cardiac outflow tract and right ventricle (Kelly et al., 2001; Mjaatvedt et al., 2001; Waldo et al., 2001). During this process, cells activate a differentiation program that leads to expression of specialized proteins, such as contractile proteins necessary for cardiomyocyte function. This process, still to be dissected in detail, should be tightly regulated so that a sufficient number of progenitors are allowed to proliferate and are prevented from differentiating prematurely. These two basic functions (pro-proliferative and anti-differentiative) are likely effected by a combination of signals and transcription factors, among which Tbx1 is a major candidate (Jerome and Papaioannou, 2001; Lindsay et al., 2001; Merscher et al., 2001; Rana et al., 2014). Indeed, Tbx1 is expressed in cardiac progenitors of the SHF, is shut down as progenitors differentiate, and positively regulates pro- proliferative signals and negatively regulates pro-differentiative factors, such as Gata4 (Liao et al., 2008; Pane et al., 2012), Mef2c (Pane et al., 2012), Srf (Chen et al., 2009) and Smad1 (Fulcoli et al., 2009). Downregulation of Srf and Smad1 signaling is effected through nontranscriptional mechanisms, but Mef2c gene expression appears to be transcriptionally repressed by Tbx1. Tbx1 regulates its target genes by interacting with the SWI-SNF- like BAF complex and with histone methyltransferases (Stoller et al., 2010; Chen et al., 2012; Fulcoli et al., 2016; Baldini et al., 2017). However, there are a number of questions to be answered. In this work, we addressed the question as to how Tbx1 represses Mef2c expression. We used three different model systems: (1) a mouse myoblast cell line, (2) transgenic mouse embryos and (3) differentiating human induced pluripotent stem cells (hiPSCs) from a 22q11.2 deletion syndrome (22q11.2DS) patient and a healthy donor. Results indicate that transcriptional repression is associated with reduced histone 3 acetylation in the region bound by Tbx1 in cultured cells, which is the previously defined anterior heart field (AHF) enhancer of the Mef2c gene (Dodou et al., 2004). In addition, we show that, in vivo, Tbx1 regulation of the AHF enhancer is dosage dependent and regionally restricted, suggesting that there are crucial interactions with other transcription factors in regulating this enhancer. RESULTS Increased Tbx1 expression is associated with reduced histone 3 acetylation of the Mef2c AHF enhancer sequence To understand the mechanisms by which Tbx1 represses Mef2c, we used mouse myoblast C2C12 cells undergoing muscle differentiation. We transfected a Tbx1 expression vector in these cells at day 0 and let them differentiate (Fig. 1A). The Mef2c gene was strongly upregulated at day 3 of differentiation in control cells, but in Tbx1-transfected cells this upregulation was strongly reduced (Fig. 1B). To exclude that this effect might be due to a delayed differentiation caused by early Tbx1 transfection, we repeated the experiment by transfecting Tbx1 after the initiation of differentiation, at day 2, and assayed Mef2c expression at day 3. Also in this case, we observed a strong reduction of Mef2c expression (Fig. 1C). Tbx1 chromatin immunoprecipitation (ChIP) with DNA sequencing (ChIP-seq) data obtained on P19Cl6 cells (Fulcoli et al., 2016) and previously published quantitative ChIP (q-ChIP) data on C2C12 cells (Pane et al., 2012) revealed enrichment at the Mef2c AHF enhancer, as previously defined (Dodou et al., 2004) (Fig. 2A). In particular, by comparing Tbx1 ChIP-seq data on P19Cl6 cells with histone modification data during cardiac Received 16 March 2017; Accepted 9 July 2018 1 CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso, Via Pietro Castellino 111, 80131 Napoli, Italy. 2 Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Napoli, Italy. *Present address: Discovery Sciences, Innovative Medicines, AstraZeneca R&D Gothenburg, Pepparedsleden 1, SE-431 83 Mo ̈ lndal, Sweden. These authors contributed equally to this work § Author for correspondence ([email protected]) A.B., 0000-0002-5330-0256 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 1 © 2018. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967 Disease Models & Mechanisms

Upload: others

Post on 30-Apr-2020

9 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Tbx1 represses Mef2c gene expression and is correlated ... · RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior

RESEARCH ARTICLE

Tbx1 represses Mef2c gene expression and is correlated withhistone 3 deacetylation of the anterior heart field enhancerLuna Simona Pane1,*,‡, Filomena Gabriella Fulcoli1,‡, Andrea Cirino2, Alessandra Altomonte1,Rosa Ferrentino1, Marchesa Bilio1 and Antonio Baldini1,2,§

ABSTRACTThe TBX1 gene is haploinsufficient in 22q11.2 deletion syndrome(22q11.2DS), and genetic evidence from human patients and mousemodels points to a major role of this gene in the pathogenesis of thissyndrome. Tbx1 can activate and repress transcription, and previouswork has shown that one of its functions is to negatively modulatecardiomyocyte differentiation. Tbx1 occupies the anterior heartfield (AHF) enhancer of the Mef2c gene, which encodes a keycardiac differentiation transcription factor. Here, we show thatincreased dosage of Tbx1 correlates with downregulation of Mef2cexpression and reduced acetylation of its AHF enhancer in culturedmouse myoblasts. Consistently, 22q11.2DS-derived and in vitro-differentiated human induced pluripotent stem cells (hiPSCs)expressed higher levels of MEF2C and showed increased AHFacetylation, compared with hiPSCs from a healthy donor. Mostimportantly, we show that in mouse embryos, loss of Tbx1 enhancesthe expression of theMef2c-AHF-Cre transgene in a specific region ofthe splanchnic mesoderm, and in a dosage-dependent manner,providing an in vivo correlate of our cell culture data. These resultsindicate that Tbx1 regulates the Mef2c AHF enhancer by inducinghistone deacetylation.

KEY WORDS: Anterior heart field enhancer, Mef2c, Tbx1

INTRODUCTIONDuring development, cardiac progenitors of the second heart field(SHF) are recruited or incorporated into the cardiac outflow tract andright ventricle (Kelly et al., 2001; Mjaatvedt et al., 2001; Waldoet al., 2001). During this process, cells activate a differentiationprogram that leads to expression of specialized proteins, such ascontractile proteins necessary for cardiomyocyte function. Thisprocess, still to be dissected in detail, should be tightly regulated sothat a sufficient number of progenitors are allowed to proliferate andare prevented from differentiating prematurely. These two basicfunctions (pro-proliferative and anti-differentiative) are likely

effected by a combination of signals and transcription factors,among which Tbx1 is a major candidate (Jerome and Papaioannou,2001; Lindsay et al., 2001; Merscher et al., 2001; Rana et al., 2014).Indeed, Tbx1 is expressed in cardiac progenitors of the SHF, is shutdown as progenitors differentiate, and positively regulates pro-proliferative signals and negatively regulates pro-differentiativefactors, such as Gata4 (Liao et al., 2008; Pane et al., 2012), Mef2c(Pane et al., 2012), Srf (Chen et al., 2009) and Smad1 (Fulcoli et al.,2009). Downregulation of Srf and Smad1 signaling is effectedthrough nontranscriptional mechanisms, butMef2c gene expressionappears to be transcriptionally repressed by Tbx1.

Tbx1 regulates its target genes by interacting with the SWI-SNF-like BAF complex and with histone methyltransferases (Stolleret al., 2010; Chen et al., 2012; Fulcoli et al., 2016; Baldini et al.,2017). However, there are a number of questions to be answered. Inthis work, we addressed the question as to how Tbx1 repressesMef2c expression. We used three different model systems: (1) amouse myoblast cell line, (2) transgenic mouse embryos and (3)differentiating human induced pluripotent stem cells (hiPSCs) froma 22q11.2 deletion syndrome (22q11.2DS) patient and a healthydonor. Results indicate that transcriptional repression is associatedwith reduced histone 3 acetylation in the region bound by Tbx1 incultured cells, which is the previously defined anterior heart field(AHF) enhancer of theMef2c gene (Dodou et al., 2004). In addition,we show that, in vivo, Tbx1 regulation of the AHF enhancer isdosage dependent and regionally restricted, suggesting that thereare crucial interactions with other transcription factors in regulatingthis enhancer.

RESULTSIncreased Tbx1 expression is associated with reducedhistone 3 acetylation of the Mef2c AHF enhancer sequenceTo understand the mechanisms by which Tbx1 represses Mef2c,we used mouse myoblast C2C12 cells undergoing muscledifferentiation. We transfected a Tbx1 expression vector in thesecells at day 0 and let them differentiate (Fig. 1A). The Mef2c genewas strongly upregulated at day 3 of differentiation in control cells,but in Tbx1-transfected cells this upregulation was strongly reduced(Fig. 1B). To exclude that this effect might be due to a delayeddifferentiation caused by early Tbx1 transfection, we repeated theexperiment by transfectingTbx1 after the initiation of differentiation,at day 2, and assayedMef2c expression at day 3. Also in this case, weobserved a strong reduction of Mef2c expression (Fig. 1C).

Tbx1 chromatin immunoprecipitation (ChIP) with DNAsequencing (ChIP-seq) data obtained on P19Cl6 cells (Fulcoliet al., 2016) and previously published quantitative ChIP (q-ChIP)data on C2C12 cells (Pane et al., 2012) revealed enrichment at theMef2c AHF enhancer, as previously defined (Dodou et al., 2004)(Fig. 2A). In particular, by comparing Tbx1 ChIP-seq data onP19Cl6 cells with histone modification data during cardiacReceived 16 March 2017; Accepted 9 July 2018

1CNR Institute of Genetics and Biophysics Adriano Buzzati Traverso,Via Pietro Castellino 111, 80131 Napoli, Italy. 2Department of Molecular Medicineand Medical Biotechnology, University of Naples Federico II, 80131 Napoli, Italy.*Present address: Discovery Sciences, Innovative Medicines, AstraZeneca R&DGothenburg, Pepparedsleden 1, SE-431 83 Molndal, Sweden.‡These authors contributed equally to this work

§Author for correspondence ([email protected])

A.B., 0000-0002-5330-0256

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

1

© 2018. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967

Disea

seModels&Mechan

isms

Page 2: Tbx1 represses Mef2c gene expression and is correlated ... · RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior

differentiation of mouse embryonic stem cells (ESCs) (Wamstadet al., 2012), we noted that the Tbx1-enriched region has relativelylow H3K27Ac expression in ESCs but progressively becomesH3K27Ac-rich during cardiac differentiation. Using available datafrom ENCODE, we also noted that the same region is acetylatedat embryonic day (E) 14.5 in the heart, where Tbx1 is not expressed(Fig. 2A). These observations suggest that, in this region, H3acetylation increases as differentiation progresses. Thus, wereasoned that Tbx1, directly or indirectly, might negativelyregulate acetylation in this region. To test this possibility, weperformed q-ChIP with an antibody against H3 acetylation (anti-H3-Ac) in C2C12 cells. Results showed that Tbx1 overexpressionsubstantially reduced H3-Ac enrichment at the Mef2c AHFenhancer (Fig. 2B). Consistent results were obtained usingantibodies against H3K27Ac on C2C12 cells after Tbx1knockdown, which increased H3K27Ac enrichment (Fig. S1). Weaddressed the question as to whether the H3 deacetylating effect ofTbx1 might be due to a direct interaction with HDAC1 or HDAC2.However, repeated co-immunoprecipitation (IP) experiments failedto demonstrate HDAC1-Tbx1 or HDAC2-Tbx1 co-IP, suggesting

that deacetylation is an indirect effect or that Tbx1 interacts withother HDACs (Fig. S2).

These results suggest that Tbx1 contrasts Mef2c AHF enhanceractivity by negatively regulating its H3 acetylation.

Loss of Tbx1 is associated with expansion of transgenicMef2c-AHF-Cre expression in vivoTo determine whether Tbx1 represses the activity of theMef2c AHFenhancer in vivo, we used the transgenic Mef2c-AHF-Cre line(Verzi et al., 2005), which carries the AHF enhancer that drives theexpression of Cre recombinase. The activity of the enhancer wasdetected using immunofluorescence with an anti-Cre antibody onembryo sections. We tested Mef2c-AHF-Cre;Tbx1+/+ (control),Mef2c-AHF-Cre;Tbx1+/− (heterozygous) and Mef2c-AHF-Cre;Tbx1flox/− embryos (homozygous null in the Mef2c-AHF-Creexpression domain, and heterozygous elsewhere). We found that atE9.5 (20 somites), Mef2c-AHF-Cre was expressed in the coremesoderm of the first and second pharyngeal arches (PAs), in thecardiac outflow tract (OFT), in the SHF (mostly anterior) and in aposterior-lateral cell population of the splanchnic mesoderm at thelevel of the cardiac inflow tract inlet, in continuity with the SHF(Fig. 3). Cre expression in the more anterior domains (PAs, OFTand SHF) was comparable between control and mutant embryos

Fig. 1. Tbx1 negatively regulates Mef2c expression in C2C12 at day 3.(A,B) Time course of Tbx1 (A) andMef2c (B) expression during in vitro C2C12myoblast differentiation, with or without transient transfection of Tbx1. Note thatTbx1 negatively regulates Mef2c expression at day 3 of differentiation (B).(C) Mef2c mRNA levels are affected by Tbx1 overexpression 24 h aftertransfection (performed at day 2). Data are from three experiments(mean±s.d.); *P<0.05. The arrows in A and B indicate the time of transfection(24 h before induction of differentiation, which is on day 0).

Fig. 2. Acetylation of histone 3 in the AHF enhancer region of Mef2c isreduced by Tbx1 overexpression. (A) Examples of ChIP-seq data profiles asshown using the UCSC genome browser (https://genome.ucsc.edu) in thegenomic region containing the AHF enhancer ofMef2c (GenBank AY324098):H3K27Ac profiles at four stages of cardiomyocyte differentiation of mouseESCs: undifferentiated ESCs (ESC), cells expressing mesodermal markers(MES), cardiac precursors (CP), cardiomyocytes (CM); H3K27Ac ChIP-seqdata profiles in E14.5 heart tissue; Tbx1 ChIP-seq data profiles. Arrowsindicate the primers used for real-time PCR amplification. (B) q-ChIP assay ofAcH3 on differentiating C2C12 cells (day 3) transfected with an empty vector(EV) or with a vector overexpressing Tbx1 (Tbx1), followed by quantitativereal-time PCR. Data are from three experiments (mean±s.d.); *P<0.05.

2

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967

Disea

seModels&Mechan

isms

Page 3: Tbx1 represses Mef2c gene expression and is correlated ... · RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior

(Fig. 3). However, more posteriorly, Cre expression was expandedin both heterozygous and homozygous mutants, albeit more evidentin the latter (Fig. 3). The region of expanded Mef2c-AHF-Creexpression is well within the Tbx1 expression domain at this stage(Fig. 4). Thus it is possible that Tbx1 regulates the enhancer directly.

Increased expression of MEF2C in 22q11.2DS-derivedhiPSCs upon cardiac differentiationTo determine whether the above observations are also relevant forhuman cells, we have generated induced pluripotent stem cells(hiPSCs) from a 22q11.2DS patient and from an unrelated healthydonor (Fig. 5). hiPSC lines were characterized as shown in Figs S3and S4. Two hiPSC clones were differentiated into cardiacprogenitors using a previously described protocol (Leschik et al.,2008; Moretti et al., 2010). We measured TBX1 expression andfound that it peaked on day 4 of differentiation (Fig. 7A), when cellsalso expressed other markers of cardiac progenitors (Fig. S5b). We

observed that 22q11.2DS-derived cell lines expressed a lower levelof TBX1, as the deletion removes one copy of the gene (Fig. S5A).

We measured the expression of MEF2C and GATA4, which isanother target of Tbx1 (Liao et al., 2008; Pane et al., 2012), byquantitative reverse transcription PCR (qRT-PCR) on days 4, 8 and12 of differentiation, and found that both genes are upregulated inpatient-derived cells at all differentiation points tested, comparedwith control cells (Fig. 6A,B).

Next, we performed q-ChIP assays on the AHF enhancerhomologous region of the MEF2C gene and on the GATA4promoter. Results demonstrated that H3-Ac enrichment issignificantly higher in 22q11.2DS cells compared with controlsfor both genes (Fig. 7).

DISCUSSIONOverall, our data indicate that Tbx1 functions as a transcriptionalrepressor of the Mef2c AHF enhancer in mouse and human cells

Fig. 3. Tbx1 regulates theMef2c AHF enhancer in vivo.(A-E) Immunofluorescence oftransverse sections of E9.5 (20 somites)embryos with the genotype indicated,using anti-Cre (green) and anti-Isl1 (red)antibodies. The first row of sections (A)corresponds to a level immediatelybelow the outflow tract. The second row(B) is just anteriorly to the inflow tract,while the third fourth and fifth rows (C-E)correspond to the inflow tract. Notethat the Cre signals in A and B arecomparable in the three genotypes,while in C-E, reduced dosage of Tbx1is associated with expansion of Creexpression (dashed line arrows). Weexamined three embryos per genotype.Fig. S3 shows Tbx1 gene expression insimilar sections. A, atrium; DPW, dorsalpericardial wall (line arrowhead); IFT,inflow tract; NT, neural tube; OFT,outflow tract; Ph, pharynx. Scale bar:100 μm.

3

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967

Disea

seModels&Mechan

isms

Page 4: Tbx1 represses Mef2c gene expression and is correlated ... · RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior

and, in vivo, in mouse embryos. In embryos, we noted that theresponse of the enhancer is Tbx1 dosage dependent and is restrictedto the splanchnic mesoderm, in a posterior-lateral domain,indicating context-dependent regulation. The AHF enhancerintegrates the actions of several critical transcription factors(Dodou et al., 2004), e.g. Isl1, Nkx2-5, Gata4 and Tbx1.Interpreting the combinatorial code governing its regulation is ofinterest for the understanding of SHF development. We havepreviously shown that in cultured cells, Tbx1 inhibits Gata4-mediated activation of the AHF enhancer, but not Isl1-mediatedactivation (Pane et al., 2012). Interestingly, here we noted that theMef2c-AHF-Cre expanded expression domain in mutant embryos isnot stained with our Isl1 immunofluorescence, whereas most of theother Mef2c-AHF-Cre expression domains are Isl1 positive. In thefuture, it would be of interest to establish whether Isl1 represents a‘protective factor’ against Tbx1-dependent suppression.Ripply3 binds Tbx1 and inhibits its transcriptional activity in

cell culture experiments (Okubo et al., 2011). In addition, andconsistently, Ripply3 turns Tbx1 into a transcriptional repressor inXenopus (Janesick et al., 2012). Ripply proteins can represstranscription through indirect interaction with HDAC1 (Brantjeset al., 2001; Jennings and Ish-Horowicz, 2008). However, in mouse,Ripply3 expression is restricted to the pharyngeal endoderm

(Okubo et al., 2011), so it is unlikely to play a role in theobserved expansion of the Mef2c-AHF-Cre domain in thesplanchnic mesoderm. Groucho/TLE proteins can also mediaterepression activity of Tbx15, Tbx18 and Tbx20 (Farin et al., 2007;Kaltenbrun et al., 2013), but Tbx1 lacks the Groucho interactiondomain (Farin et al., 2007). Our data showed that Tbx1 does notinteract directly with HDAC1 or HDAC2, suggesting the existenceof molecular intermediates responsible for driving histonedeacetylation. Alternatively, Tbx1 occupation of the enhancermight result in displacement of a transcription factor recruitinghistone acetyl transferases.

Of particular interest is the embryonic region in which Tbx1effects this function as it might be related to sorting anterior versusposterior heart field cells, and/or affect cardiomyocyte progenitorcontribution to the OFT. Future studies should be directed tothe identification of molecular intermediate(s) operating in thesplanchnic mesoderm and affecting the transcriptional activity ofTbx1 in this region.

MATERIALS AND METHODSGeneration of hiPSCsNeonatal skin fibroblasts from a 22q11.2DS/DiGeorge syndrome patientwere obtained from the Coriell Institute (GM07215); control skin fibroblastswere obtained from an unrelated age- and sex-matched anonymous, healthydonor. Informed consent was reviewed by the Institutional Review Board ofBaylor College of Medicine.

For hiPSC induction, retroviruses encoding the human OCT3 (alsoknown as SLC22A3), OCT4 (also known as POU5F1), SOX2, KLF4 andc-MYC (also known as MYC) factors were independently produced bytransfecting HEK293T cells with pMXs vectors (Addgene plasmids 17217,17218, 17219 and 17220) and the combination of moloney gag-pol plasmidpUMVC (Addgene plasmid 8449) and VSV envelope plasmid (Addgeneplasmid 8454) in Dulbecco’s modified eagle medium (DMEM) containing10% fetal bovine serum (FBS) using Fugene HD (Roche, Penzberg,Germany). Viral supernatants were harvested after 48 h and 72 h, filteredthrough a 0.45-μm low-protein-binding cellulose acetate filter, concentratedby a spin column (Millipore, Billerica, MA, USA), and used directly toinfect twice (24 h apart) 1.5×105 human primary skin fibroblasts (PSFs) inthe presence of 8 μg/ml polybrene. After 6 days, cells were seeded on mouse

Fig. 4. Transverse sections of a Tbx1+/lacZ embryo at E9.5 (22 somites)after staining with the β-gal chromogenic substrate salmon-gal.(A-E) Sections are at similar levels as in Fig. 3A-E. Scale bars: 100 µm.(F,G) Left and right sides of the whole-mount stained embryo from whichsections in A-E were derived. (H) Wild-type littermate with negative staining.DPW, dorsal pericardial wall; IFT, inflow tract; Ph, pharynx.

Fig. 5. Experimental design for the generation and cardiac differentiationof control and 22q11.2DS hiPSCs. hiPSCs were generated byoverexpression of retroviral transgenes forOCT3/4,SOX2 andKLF4 in primaryskin fibroblasts from a 22q11.2DS (DGS) patient and from an unrelated healthydonor. Two hiPSCs clones from each individual were differentiated in cardiacprogenitors and analyzed by qRT-PCR.

4

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967

Disea

seModels&Mechan

isms

Page 5: Tbx1 represses Mef2c gene expression and is correlated ... · RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior

embryonic fibroblast (MEF) feeders at a density of 5×104 cells/10-cm dishand cultured for 4 additional weeks in human ESC medium, consistingof DMEM/F12 supplemented with 20% knockout serum replacement(KSR, Invitrogen, Woltham, MA, USA), 2 mM L-glutamine, 0.1 mMnonessential amino acids, 0.1 mM β-mercaptoethanol, 50 U/ml penicillin,50 mg/ml streptomycin and 10 ng/ml human b-FGF (R&D Systems,Minneapolis, WI, USA), before hiPSC colonies were manually picked.Karyotyping of the hiPSC lines was performed at the Cell Culture Facility ofthe Telethon Institute of Genetics and Medicine in Naples, Italy, usingstandard methods.

Cardiac differentiation of hiPSCsTo generate hiPSC embryoid bodies (EBs), hiPSC colonies were dissociatedinto clumps using phosphate-buffered saline (PBS) containing 2.5 mg/mltrypsin (USB, Staufen, Germany), 1 mg/ml collagenase IV (Invitrogen),20% KSR and 1 mM CaCl2 (10 min at 37°C) and maintained for 3 days inMEF-conditioned human ESC medium in low-attachment plates. Forspontaneous differentiation, medium was then replaced with DMEM/F12supplemented with 20% FBS, 2 mM L-glutamine, 0.1 mM nonessentialamino acids, 0.1 mM β-mercaptoethanol, 50 U/ml penicillin and 50 μg/mlstreptomycin, and EBs were analyzed at day 15 for expression of markergenes of the three different germ layers. To improve cardiac differentiation,ascorbic acid (50 μg/ml) was added to the medium, and EBs were plated atday 7 on gelatin-coated dishes for better detection of beating foci.

For induction of human cardiac progenitors, hiPSCs were seeded onMEFfeeders and, 1 day later, treated for 4 days with 10 ng/ml human BMP2(R&D Systems) and 1 μM SU5402 FGF receptor inhibitor (Calbiochem,Darmstadt, Germany) in RPMI 1640 medium supplemented with 2 mML-glutamine and 2% B27 supplement without vitamin A (Invitrogen), asdescribed previously (Leschik et al., 2008; Moretti et al., 2010). Conversion

of human cardiac progenitors into myocytes and vascular cells (endothelialand smooth muscle cells) was induced by supplementing the culturemedium with 50 μg/ml ascorbic acid and 10 ng/ml human VEGF(R&D Systems).

Immunofluorescence and alkaline phosphatase activity assayhiPSCs (undifferentiated or differentiated) were fixed in 3.7% (vol/vol)formaldehyde and subjected to immunostaining by using the followingprimary antibodies: anti-human NANOG (rabbit polyclonal, Abcam,Cambridge, UK; 1:500), Alexa Fluor 488-conjugated anti-human TRA1-81 (mouse monoclonal, BD Pharmingen, Franklin Lakes, NJ, USA; 1:20).Alexa Fluor 488-, 594- and 647-conjugated secondary antibodies specific tothe appropriate species were used (Life Technologies, Carlsbad, CA, USA;1:500). Nuclei were detected with 1 µg/ml Hoechst 33528. Direct alkalinephosphatase activity was analyzed using the NBT/BCIP alkalinephosphatase blue substrate (Roche), according to the manufacturer’sguidelines. Microscopy was performed using the imaging systems(DMI6000-AF6000), filter cubes and software from Leica microsystems.Images were pseudo-colored using Adobe Photoshop.

Mouse lines and immunofluorescenceMef2c-AHF-Cre mice (Verzi et al., 2005) were crossed with Tbx1+/lacZ mice(Lindsay et al., 2001) to obtain Mef2c-AHF-Cre;Tbx1+/lacZ animals, whichwere then crossed with Tbx1flox/flox (Xu et al., 2004) or Tbx1+/lacZ mice.

Fig. 6. Analysis of MEF2C and GATA4 expression in control and22q11.2DS hiPSC-derived cardiac cells. (A,B) qRT-PCR-basedMEF2C (A)andGATA4 (B) expression in two hiPSC clones from a control (CTR) individualand two from a 22q11.2DS (DGS) patient at days 4, 8 and 12 of cardiacdifferentiation. n=3, *P<0.05 vs day 0.

Fig. 7. Acetylation of histone 3 in MEF2C and GATA4 loci is enhanced indifferentiating 22q11.2DShiPSCs. (A,B) Localization of primer pairs (arrows)used for ChIP experiments with human cells on MEF2C and GATA4 genes.‘AHF Enhancer’ indicates a region of high similarity with the murine AHFenhancer. For reference, we report ENCODE ChIP-seq data for H3K27Ac fortwo cell types, undifferentiated human embryonic stem cells (H1-hESC) andhuman skeletal muscle myoblasts (HSMM). TheGATA4 promoter region is notacetylated in these two cell types. (C) ChIP assays with an anti-AcH3 antibodyin differentiating control and 22q11.2DS hiPSCs, followed by quantitativereal-time PCR.

5

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967

Disea

seModels&Mechan

isms

Page 6: Tbx1 represses Mef2c gene expression and is correlated ... · RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior

Tbx1+/lacZ (also referred to as Tbx1+/−) mice were also crossed withwild-type mice to harvest embryos to be used for Tbx1 gene expression byβ-galactosidase assay. Animal studies were carried out under the auspices ofthe animal protocol 257/2015-PR (licensed to the A.B. laboratory)reviewed, according to Italian regulations, by the Italian Istituto Superioredi Sanità and approved by the Italian Ministero della Salute. Pregnantfemales (mostly 3-6 months old) at E9.5 were sacrificed using CO2

inhalation. The laboratory applies the ‘3Rs’ principles to minimize the use ofanimals and to limit or eliminate suffering.

Immunofluorescence on paraffin-embedded E9.5 embryo sections wasperformed using an anti-Cre antibody (69050-3, Novagen, Madison, WI,USA; 1:1000), and an anti-Isl1 antibody (39.4D5-s, Developmental StudiesHybridoma Bank, Iowa City, IA, USA; 1:50). β-galactosidase assays wereperformed on whole-mount embryos using salmon-gal (6-chloro-3-indolyl-beta-D-galactopyranoside, B21131, Alfa Aesar, Haverhill, MA, USA).

qRT-PCRTotal mRNA was isolated from PSFs, hiPSCs, EBs and cardiac cells usingthe Stratagene Absolutely RNA kit and 1 µg was used to synthesize cDNAusing the High-Capacity cDNA Reverse Transcription kit (AppliedBiosystems, Foster City, CA, USA). Gene expression was quantified byqRT-PCR using 1 µl of the reverse transcription reaction and the PowerSYBR Green PCR Master Mix (Applied Biosystems). Gene expressionlevels were normalized toGAPDH. A list of primers is provided in Table S1.

Cell lines, plasmids and transfectionsMouse C2C12 myoblasts (CRL-1772, American Type Culture Collection)were cultured in DMEM supplemented with 10% FBS. Cultures were testedfor mycoplasma infection and were negative. Authentication has beenperformed using differentiation marker analyses. For differentiation andtransient transfection, cells were plated at a density of 1.5×105 cells/well ona 35-mm tissue culture dish and incubated at 37°C in 5% CO2. After 24 h,the medium was replaced with a differentiation medium containing 2%horse serum (Hyclone, Marlborough, MA, USA). Transfection of 1 µg ofTbx1-3HA (Chen et al., 2012) was performed with X-tremeGENE (Roche)according to the manufacturer’s instructions.

ChIPC2C12 cells and hiPSCs were crosslinked with 1% formaldehyde for15 min at room temperature and glycine was added to stop the reaction to afinal concentration of 0.125 M for 5 min. The cell pellet was suspended in6× volumes of cell lysis buffer (10 mMHEPES, 60 mMKCl, 1 mMEDTA,0.075% v/v NP40, 1 mM DTT and 1× protease inhibitors, adjusted to pH7.6) in a 1.5 ml tube incubating on ice for 15 min. Isolated nuclei weresuspended in Buffer B (LowCell ChIP Kit reagent) and chromatin wassheared into 200-500-bp long fragments using the Covaris S2 SamplePreparation System (duty cycle, 5%; cycles, 5; intensity, 3; temperature,4°C; cycles per burst, 200; power mode, frequency sweeping; cycle time,60 s; degassing mode, continuous). Soluble chromatin was incubated with6 μg anti-acetyl-histone H3 (06-599, Millipore), anti-H3K27Ac (ab4729,Abcam) or normal rabbit/mouse IgG (sc-2027, Santa Cruz Biotechnology,Dallas, TX 75220, USA). Subsequent steps included extensive washesand reverse crosslinking following the LowCell ChIP Kit instructions. Forquantitative ChIP, we performed real-time PCR of the immunoprecipitatedDNA and inputs, using the FastStart Universal SYBR Green Master kit(Roche) and the 7900HT Fast Real-Time PCR System or StepOnePlus(Applied Biosystems) using primers specific for the AHF-enhancer regionof Mef2c (Table S1).

Tbx1 knockdown in C2C12 cells was obtained by transfection withnontargeted or Tbx1-targeted small interfering RNA (pool of s74767 ands74769, Life Technologies; 50 nmol/l) using Lipofectamine RNA iMAX(Life Technologies). Cells were crosslinked and processed for ChIP 48 hafter transfection, as described above.

Co-IPC2C12 cells were transfected with aTbx1:3xHA expression vector (1 µg)and harvested after 24 h for protein extraction. Then, 100 μl protein-G

Dynabeads (10004D, Life Technologies) were crosslinked to 20 μg anti-HAantibodies (11583816001, clone 12CA5, Roche) or control IgG (ab46540,Abcam). Nuclear extracts were incubated overnight at 4°C with crosslinkedantibodies and extensively washed with PBS-0.02% Tween 20. Twoconsecutive elutions were performed with 0.1 M glycine (pH2.8) andimmediately neutralized with Tris-HCl (pH 8.0). Samples were subjected tosodium dodecyl sulfate–polyacrylamide gel electrophoresis and proteinswere transferred into polyvinylidene fluoride membrane for western blotanalyses with anti-HDAC1 antibody (Ab7028, Abcam), anti-HDAC2(sc-7899, Santa Cruz Biotechnology), anti-HA antibody (11583816001,clone 12CA5, Roche) and VeriBlot for IP secondary antibody (HRP)(ab131366, Abcam; 1:200).

Statistical analysisAll data were expressed as means±s.e.m. (or s.d., as indicated in the figurelegends) from independent experiments. Differences between groups wereexamined for statistical analysis using two-tailed Student’s t-test. P<0.05was considered significant.

AcknowledgementsWe thank Dr Thomas Zwaka and his laboratory members for invaluable help inestablishing methods for hiPSC generation and characterization. We acknowledgethe support of the Integrated Microscopy Facility of the Institute of Genetics andBiophysics and the Cell Culture Facility of TIGEM, Pozzuoli, Italy.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsConceptualization: L.S.P., F.G.F., A.B.; Methodology: L.S.P., F.G.F., A.C., A.A.,R.F., M.B.; Validation: F.G.F., A.A.; Formal analysis: L.S.P., F.G.F., R.F., M.B., A.B.;Investigation: L.S.P., F.G.F., A.C., A.A., A.B.; Data curation: L.S.P., F.G.F., A.B.;Writing - original draft: A.B.; Writing - review & editing: L.S.P., F.G.F.; Visualization:M.B.; Supervision: A.B.; Project administration: A.B.; Funding acquisition: A.B.

FundingThis work was supported by Fondazione Telethon (GGP14211), Fondation Leducq(TNE 15CVD01) and Ministero dell’Istruzione, dell’Universita e della Ricerca/FAREBIO (to A.B.).

Supplementary informationSupplementary information available online athttp://dmm.biologists.org/lookup/doi/10.1242/dmm.029967.supplemental

ReferencesBaldini, A., Fulcoli, F. G. and Illingworth, E. (2017). Tbx1: transcriptional and

developmental functions. Curr. Top. Dev. Biol. 122, 223-243.Brantjes, H., Roose, J., van DeWetering, M. and Clevers, H. (2001). All Tcf HMG

box transcription factors interact with Groucho-related co-repressors. NucleicAcids Res. 29, 1410-1419.

Chen, L., Fulcoli, F. G., Tang, S. and Baldini, A. (2009). Tbx1 regulatesproliferation and differentiation of multipotent heart progenitors. Circ. Res. 105,842-851.

Chen, L., Fulcoli, F. G., Ferrentino, R., Martucciello, S., Illingworth, E. A. andBaldini, A. (2012). Transcriptional control in cardiac progenitors: Tbx1 interactswith the BAF chromatin remodeling complex and regulates Wnt5a. PLoS Genet.8, e1002571.

Dodou, E., Verzi, M. P., Anderson, J. P., Xu, S. M. and Black, B. L. (2004).Mef2c is a direct transcriptional target of ISL1 and GATA factors in theanterior heart field during mouse embryonic development. Development 131,3931-3942.

Farin, H. F., Bussen, M., Schmidt, M. K., Singh, M. K., Schuster-Gossler, K.and Kispert, A. (2007). Transcriptional repression by the T-box proteinsTbx18 and Tbx15 depends on Groucho corepressors. J. Biol. Chem. 282,25748-25759.

Fulcoli, F. G., Huynh, T., Scambler, P. J. and Baldini, A. (2009). Tbx1 regulatesthe BMP-Smad1 pathway in a transcription independent manner. PLoS ONE4, e6049.

Fulcoli, F. G., Franzese,M., Liu, X., Zhang, Z., Angelini, C. andBaldini, A. (2016).Rebalancing gene haploinsufficiency in vivo by targeting chromatin. Nat.Commun. 7, 11688.

Janesick, A., Shiotsugu, J., Taketani, M. and Blumberg, B. (2012). RIPPLY3 is aretinoic acid-inducible repressor required for setting the borders of thepre-placodal ectoderm. Development 139, 1213-1224.

6

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967

Disea

seModels&Mechan

isms

Page 7: Tbx1 represses Mef2c gene expression and is correlated ... · RESEARCH ARTICLE Tbx1 represses Mef2c gene expression and is correlated with histone 3 deacetylation of the anterior

Jennings, B. H. and Ish-Horowicz, D. (2008). The Groucho/TLE/Grg family oftranscriptional co-repressors. Genome Biol. 9, 205.

Jerome, L. A. and Papaioannou, V. E. (2001). DiGeorge syndrome phenotype inmice mutant for the T-box gene, Tbx1. Nat. Genet. 27, 286-291.

Kaltenbrun, E., Greco, T. M., Slagle, C. E., Kennedy, L. M., Li, T., Cristea, I. M.and Conlon, F. L. (2013). A Gro/TLE-NuRD corepressor complex facilitatesTbx20-dependent transcriptional repression. J. Proteome Res. 12, 5395-5409.

Kelly, R. G., Brown, N. A. and Buckingham, M. E. (2001). The arterial pole ofthe mouse heart forms from Fgf10-expressing cells in pharyngeal mesoderm.Dev. Cell 1, 435-440.

Leschik, J., Stefanovic, S., Brinon, B. and Puceat, M. (2008). Cardiaccommitment of primate embryonic stem cells. Nat. Protoc. 3, 1381-1387.

Liao, J., Aggarwal, V. S., Nowotschin, S., Bondarev, A., Lipner, S. and Morrow,B. E. (2008). Identification of downstream genetic pathways of Tbx1 in the secondheart field. Dev. Biol. 316, 524-537.

Lindsay, E. A., Vitelli, F., Su, H., Morishima, M., Huynh, T., Pramparo, T.,Jurecic, V., Ogunrinu, G., Sutherland, H. F., Scambler, P. J. et al. (2001). Tbx1haploinsufficieny in the DiGeorge syndrome region causes aortic arch defects inmice. Nature 410, 97-101.

Merscher, S., Funke, B., Epstein, J. A., Heyer, J., Puech, A., Lu, M. M., Xavier,R. J., Demay, M. B., Russell, R. G., Factor, S. et al. (2001). TBX1 is responsiblefor cardiovascular defects in velo-cardio-facial/digeorge syndrome. Cell 104,619-629.

Mjaatvedt, C. H., Nakaoka, T., Moreno-Rodriguez, R., Norris, R. A., Kern, M. J.,Eisenberg, C. A., Turner, D. andMarkwald, R. R. (2001). The outflow tract of theheart is recruited from a novel heart-forming field. Dev. Biol. 238, 97-109.

Moretti, A., Bellin, M., Jung, C. B., Thies, T.-M., Takashima, Y., Bernshausen, A.,Schiemann, M., Fischer, S., Moosmang, S., Smith, A. G. et al. (2010). Mouse

and human induced pluripotent stem cells as a source for multipotent Isl1+cardiovascular progenitors. FASEB J. 24, 700-711.

Okubo, T., Kawamura, A., Takahashi, J., Yagi, H., Morishima, M., Matsuoka, R.and Takada, S. (2011). Ripply3, a Tbx1 repressor, is required for development ofthe pharyngeal apparatus and its derivatives in mice. Development 138, 339-348.

Pane, L. S., Zhang, Z., Ferrentino, R., Huynh, T., Cutillo, L. and Baldini, A.(2012). Tbx1 is a negative modulator of Mef2c. Hum. Mol. Genet. 21, 2485-2496.

Rana, M. S., Theveniau-Ruissy, M., De Bono, C., Mesbah, K., Francou, A.,Rammah, M., Domınguez, J. N., Roux, M., Laforest, B., Anderson, R. H. et al.(2014). Tbx1 coordinates addition of posterior second heart field progenitor cellsto the arterial and venous poles of the heart. Circ. Res. 115, 790-799.

Stoller, J. Z., Huang, L., Tan, C. C., Huang, F., Zhou, D. D., Yang, J., Gelb, B. D.and Epstein, J. A. (2010). Ash2l interacts with Tbx1 and is required during earlyembryogenesis. Exp. Biol. Med. (Maywood) 235, 569-576.

Verzi, M. P., Mcculley, D. J., De Val, S., Dodou, E. and Black, B. L. (2005). Theright ventricle, outflow tract, and ventricular septum comprise a restrictedexpression domain within the secondary/anterior heart field. Dev. Biol. 287,134-145.

Waldo, K. L., Kumiski, D. H., Wallis, K. T., Stadt, H. A., Hutson, M. R., Platt, D. H.and Kirby, M. L. (2001). Conotruncal myocardium arises from a secondary heartfield. Development 128, 3179-3188.

Wamstad, J. A., Alexander, J. M., Truty, R. M., Shrikumar, A., Li, F., Eilertson,K. E., Ding, H., Wylie, J. N., Pico, A. R., Capra, J. A. et al. (2012). Dynamic andcoordinated epigenetic regulation of developmental transitions in the cardiaclineage. Cell 151, 206-220.

Xu, H., Morishima, M., Wylie, J. N., Schwartz, R. J., Bruneau, B. G., Lindsay,E. A. and Baldini, A. (2004). Tbx1 has a dual role in the morphogenesis of thecardiac outflow tract. Development 131, 3217-3227.

7

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm029967. doi:10.1242/dmm.029967

Disea

seModels&Mechan

isms