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

RESEARCH ARTICLE Tbx1 represses Mef2c expression and is correlated with histone 3 deacetylation of the anterior heart field enhancer Luna Simona Pane1,*,‡, Filomena Gabriella Fulcoli1,‡, Andrea Cirino2, Alessandra Altomonte1, Rosa Ferrentino1, Marchesa Bilio1 and Antonio Baldini1,2,§

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

Received 16 March 2017; Accepted 9 July 2018 P19Cl6 cells with histone modification data during cardiac Disease Models & Mechanisms

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

Fig. 2. Acetylation of histone 3 in the AHF enhancer region of Mef2c is reduced by Tbx1 overexpression. (A) Examples of ChIP-seq data profiles as shown using the UCSC genome browser (https://genome.ucsc.edu) in the genomic region containing the AHF enhancer of Mef2c (GenBank AY324098): H3K27Ac profiles at four stages of cardiomyocyte differentiation of mouse ESCs: undifferentiated ESCs (ESC), cells expressing mesodermal markers (MES), cardiac precursors (CP), cardiomyocytes (CM); H3K27Ac ChIP-seq data profiles in E14.5 heart tissue; Tbx1 ChIP-seq data profiles. Arrows Fig. 1. Tbx1 negatively regulates Mef2c expression in C2C12 at day 3. indicate the primers used for real-time PCR amplification. (B) q-ChIP assay of (A,B) Time course of Tbx1 (A) and Mef2c (B) expression during in vitro C2C12 AcH3 on differentiating C2C12 cells (day 3) transfected with an empty vector myoblast differentiation, with or without transient transfection of Tbx1. Note that (EV) or with a vector overexpressing Tbx1 (Tbx1), followed by quantitative Tbx1 negatively regulates Mef2c expression at day 3 of differentiation (B). real-time PCR. Data are from three experiments (mean±s.d.); *P<0.05. (C) Mef2c mRNA levels are affected by Tbx1 overexpression 24 h after transfection (performed at day 2). Data are from three experiments P (mean±s.d.); * <0.05. The arrows in A and B indicate the time of transfection that deacetylation is an indirect effect or that Tbx1 interacts with (24 h before induction of differentiation, which is on day 0). other HDACs (Fig. S2). These results suggest that Tbx1 contrasts Mef2c AHF enhancer differentiation of mouse embryonic stem cells (ESCs) (Wamstad activity by negatively regulating its H3 acetylation. et al., 2012), we noted that the Tbx1-enriched region has relatively low H3K27Ac expression in ESCs but progressively becomes Loss of Tbx1 is associated with expansion of transgenic H3K27Ac-rich during cardiac differentiation. Using available data Mef2c-AHF-Cre expression in vivo from ENCODE, we also noted that the same region is acetylated To determine whether Tbx1 represses the activity of the Mef2c AHF at embryonic day (E) 14.5 in the heart, where Tbx1 is not expressed enhancer in vivo, we used the transgenic Mef2c-AHF-Cre line (Fig. 2A). These observations suggest that, in this region, H3 (Verzi et al., 2005), which carries the AHF enhancer that drives the acetylation increases as differentiation progresses. Thus, we expression of Cre recombinase. The activity of the enhancer was reasoned that Tbx1, directly or indirectly, might negatively detected using immunofluorescence with an anti-Cre antibody on regulate acetylation in this region. To test this possibility, we embryo sections. We tested Mef2c-AHF-Cre;Tbx1+/+ (control), performed q-ChIP with an antibody against H3 acetylation (anti- Mef2c-AHF-Cre;Tbx1+/− (heterozygous) and Mef2c-AHF-Cre; H3-Ac) in C2C12 cells. Results showed that Tbx1 overexpression Tbx1flox/− embryos (homozygous null in the Mef2c-AHF-Cre substantially reduced H3-Ac enrichment at the Mef2c AHF expression domain, and heterozygous elsewhere). We found that at enhancer (Fig. 2B). Consistent results were obtained using E9.5 (20 somites), Mef2c-AHF-Cre was expressed in the core antibodies against H3K27Ac on C2C12 cells after Tbx1 mesoderm of the first and second pharyngeal arches (PAs), in the knockdown, which increased H3K27Ac enrichment (Fig. S1). We cardiac outflow tract (OFT), in the SHF (mostly anterior) and in a addressed the question as to whether the H3 deacetylating effect of posterior-lateral cell population of the splanchnic mesoderm at the Tbx1 might be due to a direct interaction with HDAC1 or HDAC2. level of the cardiac inflow tract inlet, in continuity with the SHF However, repeated co-immunoprecipitation (IP) experiments failed (Fig. 3). Cre expression in the more anterior domains (PAs, OFT to demonstrate HDAC1-Tbx1 or HDAC2-Tbx1 co-IP, suggesting and SHF) was comparable between control and mutant embryos Disease Models & Mechanisms

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

Fig. 3. Tbx1 regulates the Mef2c AHF enhancer in vivo. (A-E) Immunofluorescence of transverse 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 immediately below 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. Note that the Cre signals in A and B are comparable in the three genotypes, while in C-E, reduced dosage of Tbx1 is associated with expansion of Cre expression (dashed line arrows). We examined three embryos per genotype. Fig. S3 shows Tbx1 gene expression in similar sections. A, atrium; DPW, dorsal pericardial wall (line arrowhead); IFT, inflow tract; NT, neural tube; OFT, outflow tract; Ph, pharynx. Scale bar: 100 μm.

(Fig. 3). However, more posteriorly, Cre expression was expanded observed that 22q11.2DS-derived cell lines expressed a lower level in both heterozygous and homozygous mutants, albeit more evident of TBX1, as the deletion removes one copy of the gene (Fig. S5A). in the latter (Fig. 3). The region of expanded Mef2c-AHF-Cre We measured the expression of MEF2C and GATA4, which is expression is well within the Tbx1 expression domain at this stage another target of Tbx1 (Liao et al., 2008; Pane et al., 2012), by (Fig. 4). Thus it is possible that Tbx1 regulates the enhancer directly. quantitative reverse transcription PCR (qRT-PCR) on days 4, 8 and 12 of differentiation, and found that both genes are upregulated in Increased expression of MEF2C in 22q11.2DS-derived patient-derived cells at all differentiation points tested, compared hiPSCs upon cardiac differentiation with control cells (Fig. 6A,B). To determine whether the above observations are also relevant for Next, we performed q-ChIP assays on the AHF enhancer human cells, we have generated induced pluripotent stem cells homologous region of the MEF2C gene and on the GATA4 (hiPSCs) from a 22q11.2DS patient and from an unrelated healthy promoter. Results demonstrated that H3-Ac enrichment is donor (Fig. 5). hiPSC lines were characterized as shown in Figs S3 significantly higher in 22q11.2DS cells compared with controls and S4. Two hiPSC clones were differentiated into cardiac for both genes (Fig. 7). progenitors using a previously described protocol (Leschik et al., 2008; Moretti et al., 2010). We measured TBX1 expression and DISCUSSION found that it peaked on day 4 of differentiation (Fig. 7A), when cells Overall, our data indicate that Tbx1 functions as a transcriptional also expressed other markers of cardiac progenitors (Fig. S5b). We repressor of the Mef2c AHF enhancer in mouse and human cells Disease Models & Mechanisms

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

Fig. 5. Experimental design for the generation and cardiac differentiation of control and 22q11.2DS hiPSCs. hiPSCs were generated by overexpression of retroviral transgenes for OCT3/4, and in primary skin fibroblasts from a 22q11.2DS (DGS) patient and from an unrelated healthy donor. Two hiPSCs clones from each individual were differentiated in cardiac progenitors and analyzed by qRT-PCR.

(Okubo et al., 2011), so it is unlikely to play a role in the observed expansion of the Mef2c-AHF-Cre domain in the splanchnic mesoderm. Groucho/TLE proteins can also mediate repression activity of Tbx15, Tbx18 and Tbx20 (Farin et al., 2007; Kaltenbrun et al., 2013), but Tbx1 lacks the Groucho interaction domain (Farin et al., 2007). Our data showed that Tbx1 does not interact directly with HDAC1 or HDAC2, suggesting the existence Fig. 4. Transverse sections of a Tbx1+/lacZ embryo at E9.5 (22 somites) of molecular intermediates responsible for driving histone after staining with the β-gal chromogenic substrate salmon-gal. deacetylation. Alternatively, Tbx1 occupation of the enhancer (A-E) Sections are at similar levels as in Fig. 3A-E. Scale bars: 100 µm. might result in displacement of a transcription factor recruiting (F,G) Left and right sides of the whole-mount stained embryo from which histone acetyl transferases. sections in A-E were derived. (H) Wild-type littermate with negative staining. DPW, dorsal pericardial wall; IFT, inflow tract; Ph, pharynx. Of particular interest is the embryonic region in which Tbx1 effects this function as it might be related to sorting anterior versus posterior heart field cells, and/or affect cardiomyocyte progenitor and, in vivo, in mouse embryos. In embryos, we noted that the contribution to the OFT. Future studies should be directed to response of the enhancer is Tbx1 dosage dependent and is restricted the identification of molecular intermediate(s) operating in the to the splanchnic mesoderm, in a posterior-lateral domain, splanchnic mesoderm and affecting the transcriptional activity of indicating context-dependent regulation. The AHF enhancer Tbx1 in this region. integrates the actions of several critical transcription factors (Dodou et al., 2004), e.g. Isl1, Nkx2-5, Gata4 and Tbx1. MATERIALS AND METHODS Interpreting the combinatorial code governing its regulation is of Generation of hiPSCs interest for the understanding of SHF development. We have Neonatal skin fibroblasts from a 22q11.2DS/DiGeorge syndrome patient were obtained from the Coriell Institute (GM07215); control skin fibroblasts previously shown that in cultured cells, Tbx1 inhibits Gata4- were obtained from an unrelated age- and sex-matched anonymous, healthy mediated activation of the AHF enhancer, but not Isl1-mediated donor. Informed consent was reviewed by the Institutional Review Board of activation (Pane et al., 2012). Interestingly, here we noted that the Baylor College of Medicine. Mef2c-AHF-Cre expanded expression domain in mutant embryos is For hiPSC induction, retroviruses encoding the human OCT3 (also not stained with our Isl1 immunofluorescence, whereas most of the known as SLC22A3), OCT4 (also known as POU5F1), SOX2, KLF4 and other Mef2c-AHF-Cre expression domains are Isl1 positive. In the c- (also known as MYC) factors were independently produced by future, it would be of interest to establish whether Isl1 represents a transfecting HEK293T cells with pMXs vectors (Addgene plasmids 17217, ‘protective factor’ against Tbx1-dependent suppression. 17218, 17219 and 17220) and the combination of moloney gag-pol plasmid pUMVC (Addgene plasmid 8449) and VSV envelope plasmid (Addgene Ripply3 binds Tbx1 and inhibits its transcriptional activity in ’ cell culture experiments (Okubo et al., 2011). In addition, and plasmid 8454) in Dulbecco s modified eagle medium (DMEM) containing 10% fetal bovine serum (FBS) using Fugene HD (Roche, Penzberg, consistently, Ripply3 turns Tbx1 into a transcriptional repressor in Xenopus Germany). Viral supernatants were harvested after 48 h and 72 h, filtered (Janesick et al., 2012). Ripply proteins can repress through a 0.45-μm low--binding cellulose acetate filter, concentrated transcription through indirect interaction with HDAC1 (Brantjes by a spin column (Millipore, Billerica, MA, USA), and used directly to et al., 2001; Jennings and Ish-Horowicz, 2008). However, in mouse, infect twice (24 h apart) 1.5×105 human primary skin fibroblasts (PSFs) in

Ripply3 expression is restricted to the pharyngeal endoderm the presence of 8 μg/ml polybrene. After 6 days, cells were seeded on mouse Disease Models & Mechanisms

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

Fig. 6. Analysis of MEF2C and GATA4 expression in control and MEF2C 22q11.2DS hiPSC-derived cardiac cells. (A,B) qRT-PCR-based (A) Fig. 7. Acetylation of histone 3 in MEF2C and GATA4 loci is enhanced in GATA4 and (B) expression in two hiPSC clones from a control (CTR) individual differentiating 22q11.2DS hiPSCs. (A,B) Localization of primer pairs (arrows) and two from a 22q11.2DS (DGS) patient at days 4, 8 and 12 of cardiac used for ChIP experiments with human cells on MEF2C and GATA4 genes. n P differentiation. =3, * <0.05 vs day 0. ‘AHF Enhancer’ indicates a region of high similarity with the murine AHF enhancer. For reference, we report ENCODE ChIP-seq data for H3K27Ac for embryonic fibroblast (MEF) feeders at a density of 5×104 cells/10-cm dish two cell types, undifferentiated human embryonic stem cells (H1-hESC) and GATA4 and cultured for 4 additional weeks in human ESC medium, consisting human myoblasts (HSMM). The promoter region is not of DMEM/F12 supplemented with 20% knockout serum replacement acetylated in these two cell types. (C) ChIP assays with an anti-AcH3 antibody in differentiating control and 22q11.2DS hiPSCs, followed by quantitative (KSR, Invitrogen, Woltham, MA, USA), 2 mM L-glutamine, 0.1 mM real-time PCR. nonessential 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. of human cardiac progenitors into myocytes and vascular cells (endothelial Karyotyping of the hiPSC lines was performed at the Cell Culture Facility of and smooth muscle cells) was induced by supplementing the culture the Telethon Institute of Genetics and Medicine in Naples, Italy, using medium with 50 μg/ml ascorbic acid and 10 ng/ml human VEGF standard methods. (R&D Systems).

Cardiac differentiation of hiPSCs Immunofluorescence and alkaline phosphatase activity assay To generate hiPSC embryoid bodies (EBs), hiPSC colonies were dissociated hiPSCs (undifferentiated or differentiated) were fixed in 3.7% (vol/vol) into clumps using phosphate-buffered saline (PBS) containing 2.5 mg/ml formaldehyde and subjected to immunostaining by using the following trypsin (USB, Staufen, Germany), 1 mg/ml collagenase IV (Invitrogen), primary antibodies: anti-human NANOG (rabbit polyclonal, Abcam, 20% KSR and 1 mM CaCl2 (10 min at 37°C) and maintained for 3 days in Cambridge, UK; 1:500), Alexa Fluor 488-conjugated anti-human TRA1- MEF-conditioned human ESC medium in low-attachment plates. For 81 (mouse monoclonal, BD Pharmingen, Franklin Lakes, NJ, USA; 1:20). spontaneous differentiation, medium was then replaced with DMEM/F12 Alexa Fluor 488-, 594- and 647-conjugated secondary antibodies specific to supplemented with 20% FBS, 2 mM L-glutamine, 0.1 mM nonessential the appropriate species were used (Life Technologies, Carlsbad, CA, USA; amino acids, 0.1 mM β-mercaptoethanol, 50 U/ml penicillin and 50 μg/ml 1:500). Nuclei were detected with 1 µg/ml Hoechst 33528. Direct alkaline streptomycin, and EBs were analyzed at day 15 for expression of marker phosphatase activity was analyzed using the NBT/BCIP alkaline genes of the three different germ layers. To improve cardiac differentiation, phosphatase blue substrate (Roche), according to the manufacturer’s ascorbic acid (50 μg/ml) was added to the medium, and EBs were plated at guidelines. Microscopy was performed using the imaging systems day 7 on gelatin-coated dishes for better detection of beating foci. (DMI6000-AF6000), filter cubes and software from Leica microsystems. For induction of human cardiac progenitors, hiPSCs were seeded on MEF Images were pseudo-colored using Adobe Photoshop. feeders and, 1 day later, treated for 4 days with 10 ng/ml human BMP2 (R&D Systems) and 1 μM SU5402 FGF inhibitor (Calbiochem, Mouse lines and immunofluorescence Darmstadt, Germany) in RPMI 1640 medium supplemented with 2 mM Mef2c-AHF-Cre mice (Verzi et al., 2005) were crossed with Tbx1+/lacZ mice L-glutamine and 2% B27 supplement without vitamin A (Invitrogen), as (Lindsay et al., 2001) to obtain Mef2c-AHF-Cre;Tbx1+/lacZ animals, which flox described previously (Leschik et al., 2008; Moretti et al., 2010). Conversion were then crossed with Tbx1 /flox (Xu et al., 2004) or Tbx1+/lacZ mice. Disease Models & Mechanisms

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

Tbx1+/lacZ (also referred to as Tbx1+/−) mice were also crossed with Dynabeads (10004D, Life Technologies) were crosslinked to 20 μg anti-HA wild-type mice to harvest embryos to be used for Tbx1 gene expression by antibodies (11583816001, clone 12CA5, Roche) or control IgG (ab46540, β-galactosidase assay. Animal studies were carried out under the auspices of Abcam). Nuclear extracts were incubated overnight at 4°C with crosslinked the animal protocol 257/2015-PR (licensed to the A.B. laboratory) antibodies and extensively washed with PBS-0.02% Tween 20. Two reviewed, according to Italian regulations, by the Italian Istituto Superiore consecutive elutions were performed with 0.1 M glycine (pH2.8) and di Sanità and approved by the Italian Ministero della Salute. Pregnant immediately neutralized with Tris-HCl (pH 8.0). Samples were subjected to females (mostly 3-6 months old) at E9.5 were sacrificed using CO2 sodium dodecyl sulfate–polyacrylamide gel electrophoresis and proteins inhalation. The laboratory applies the ‘3Rs’ principles to minimize the use of were transferred into polyvinylidene fluoride membrane for western blot animals and to limit or eliminate suffering. analyses with anti-HDAC1 antibody (Ab7028, Abcam), anti-HDAC2 Immunofluorescence on paraffin-embedded E9.5 embryo sections was (sc-7899, Santa Cruz Biotechnology), anti-HA antibody (11583816001, performed using an anti-Cre antibody (69050-3, Novagen, Madison, WI, clone 12CA5, Roche) and VeriBlot for IP secondary antibody (HRP) USA; 1:1000), and an anti-Isl1 antibody (39.4D5-s, Developmental Studies (ab131366, Abcam; 1:200). Hybridoma Bank, Iowa City, IA, USA; 1:50). β-galactosidase assays were performed on whole-mount embryos using salmon-gal (6-chloro-3-indolyl- Statistical analysis beta-D-galactopyranoside, B21131, Alfa Aesar, Haverhill, MA, USA). All data were expressed as means±s.e.m. (or s.d., as indicated in the figure legends) from independent experiments. Differences between groups were qRT-PCR examined for statistical analysis using two-tailed Student’s t-test. P<0.05 Total mRNA was isolated from PSFs, hiPSCs, EBs and cardiac cells using was considered significant. the Stratagene Absolutely RNA kit and 1 µg was used to synthesize cDNA using the High-Capacity cDNA Reverse Transcription kit (Applied Acknowledgements Biosystems, Foster City, CA, USA). Gene expression was quantified by We thank Dr Thomas Zwaka and his laboratory members for invaluable help in qRT-PCR using 1 µl of the reverse transcription reaction and the Power establishing methods for hiPSC generation and characterization. We acknowledge the support of the Integrated Microscopy Facility of the Institute of Genetics and SYBR Green PCR Master Mix (Applied Biosystems). Gene expression Biophysics and the Cell Culture Facility of TIGEM, Pozzuoli, Italy. levels were normalized to GAPDH. A list of primers is provided in Table S1. Competing interests Cell lines, plasmids and transfections The authors declare no competing or financial interests. Mouse C2C12 myoblasts (CRL-1772, American Type Culture Collection) were cultured in DMEM supplemented with 10% FBS. Cultures were tested Author contributions for mycoplasma infection and were negative. Authentication has been Conceptualization: L.S.P., F.G.F., A.B.; Methodology: L.S.P., F.G.F., A.C., A.A., performed using differentiation marker analyses. For differentiation and 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.; transient transfection, cells were plated at a density of 1.5×105 cells/well on Writing - original draft: A.B.; Writing - review & editing: L.S.P., F.G.F.; Visualization: a 35-mm tissue culture dish and incubated at 37°C in 5% CO2. After 24 h, M.B.; Supervision: A.B.; Project administration: A.B.; Funding acquisition: A.B. the medium was replaced with a differentiation medium containing 2% horse serum (Hyclone, Marlborough, MA, USA). Transfection of 1 µg of Funding Tbx1-3HA (Chen et al., 2012) was performed with X-tremeGENE (Roche) This work was supported by Fondazione Telethon (GGP14211), Fondation Leducq according to the manufacturer’s instructions. (TNE 15CVD01) and Ministero dell’Istruzione, dell’Universitàe della Ricerca/ FAREBIO (to A.B.). ChIP C2C12 cells and hiPSCs were crosslinked with 1% formaldehyde for Supplementary information Supplementary information available online at 15 min at room temperature and glycine was added to stop the reaction to a http://dmm.biologists.org/lookup/doi/10.1242/dmm.029967.supplemental final concentration of 0.125 M for 5 min. The cell pellet was suspended in 6× volumes of cell lysis buffer (10 mM HEPES, 60 mM KCl, 1 mM EDTA, References 0.075% v/v NP40, 1 mM DTT and 1× protease inhibitors, adjusted to pH Baldini, A., Fulcoli, F. G. and Illingworth, E. (2017). Tbx1: transcriptional and 7.6) in a 1.5 ml tube incubating on ice for 15 min. Isolated nuclei were developmental functions. Curr. Top. Dev. Biol. 122, 223-243. suspended in Buffer B (LowCell ChIP Kit reagent) and chromatin was Brantjes, H., Roose, J., van De Wetering, M. and Clevers, H. (2001). All Tcf HMG sheared into 200-500-bp long fragments using the Covaris S2 Sample box transcription factors interact with Groucho-related co-repressors. Nucleic Acids Res. Preparation System (duty cycle, 5%; cycles, 5; intensity, 3; temperature, 29, 1410-1419. Chen, L., Fulcoli, F. G., Tang, S. and Baldini, A. (2009). Tbx1 regulates 4°C; cycles per burst, 200; power mode, frequency sweeping; cycle time, proliferation and differentiation of multipotent heart progenitors. Circ. Res. 105, 60 s; degassing mode, continuous). Soluble chromatin was incubated with 842-851. 6 μg anti-acetyl-histone H3 (06-599, Millipore), anti-H3K27Ac (ab4729, Chen, L., Fulcoli, F. G., Ferrentino, R., Martucciello, S., Illingworth, E. A. and Abcam) or normal rabbit/mouse IgG (sc-2027, Santa Cruz Biotechnology, Baldini, A. (2012). Transcriptional control in cardiac progenitors: Tbx1 interacts Dallas, TX 75220, USA). Subsequent steps included extensive washes with the BAF chromatin remodeling complex and regulates Wnt5a. PLoS Genet. and reverse crosslinking following the LowCell ChIP Kit instructions. For 8, e1002571. Dodou, E., Verzi, M. P., Anderson, J. P., Xu, S. M. and Black, B. L. (2004). quantitative ChIP, we performed real-time PCR of the immunoprecipitated Mef2c is a direct transcriptional target of ISL1 and GATA factors in the DNA and inputs, using the FastStart Universal SYBR Green Master kit anterior heart field during mouse embryonic development. Development 131, (Roche) and the 7900HT Fast Real-Time PCR System or StepOnePlus 3931-3942. (Applied Biosystems) using primers specific for the AHF-enhancer region Farin, H. F., Bussen, M., Schmidt, M. K., Singh, M. K., Schuster-Gossler, K. of Mef2c (Table S1). and Kispert, A. (2007). Transcriptional repression by the T-box proteins J. Biol. Chem. Tbx1 knockdown in C2C12 cells was obtained by transfection with Tbx18 and Tbx15 depends on Groucho corepressors. 282, Tbx1 25748-25759. nontargeted or -targeted small interfering RNA (pool of s74767 and Fulcoli, F. G., Huynh, T., Scambler, P. J. and Baldini, A. (2009). Tbx1 regulates s74769, Life Technologies; 50 nmol/l) using Lipofectamine RNA iMAX the BMP-Smad1 pathway in a transcription independent manner. PLoS ONE (Life Technologies). Cells were crosslinked and processed for ChIP 48 h 4, e6049. after transfection, as described above. Fulcoli, F. G., Franzese, M., Liu, X., Zhang, Z., Angelini, C. and Baldini, A. (2016). Rebalancing gene haploinsufficiency in vivo by targeting chromatin. Nat. Commun. 7, 11688. Co-IP Janesick, A., Shiotsugu, J., Taketani, M. and Blumberg, B. (2012). RIPPLY3 is a C2C12 cells were transfected with aTbx1:3xHA expression vector (1 µg) retinoic acid-inducible repressor required for setting the borders of the and harvested after 24 h for protein extraction. Then, 100 μl protein-G pre-placodal ectoderm. Development 139, 1213-1224. Disease Models & Mechanisms

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

Jennings, B. H. and Ish-Horowicz, D. (2008). The Groucho/TLE/Grg family of and human induced pluripotent stem cells as a source for multipotent Isl1+ transcriptional co-repressors. Genome Biol. 9, 205. cardiovascular progenitors. FASEB J. 24, 700-711. Jerome, L. A. and Papaioannou, V. E. (2001). DiGeorge syndrome phenotype in Okubo, T., Kawamura, A., Takahashi, J., Yagi, H., Morishima, M., Matsuoka, R. mice mutant for the T-box gene, Tbx1. Nat. Genet. 27, 286-291. and Takada, S. (2011). Ripply3, a Tbx1 repressor, is required for development of Kaltenbrun, E., Greco, T. M., Slagle, C. E., Kennedy, L. M., Li, T., Cristea, I. M. the pharyngeal apparatus and its derivatives in mice. Development 138, 339-348. and Conlon, F. L. (2013). A Gro/TLE-NuRD corepressor complex facilitates Pane, L. S., Zhang, Z., Ferrentino, R., Huynh, T., Cutillo, L. and Baldini, A. Tbx20-dependent transcriptional repression. J. Proteome Res. 12, 5395-5409. (2012). Tbx1 is a negative modulator of Mef2c. Hum. Mol. Genet. 21, 2485-2496. Kelly, R. G., Brown, N. A. and Buckingham, M. E. (2001). The arterial pole of Rana, M. S., Théveniau-Ruissy, M., De Bono, C., Mesbah, K., Francou, A., the mouse heart forms from Fgf10-expressing cells in pharyngeal mesoderm. Rammah, M., Domınguez,́ J. N., Roux, M., Laforest, B., Anderson, R. H. et al. Dev. Cell 1, 435-440. (2014). Tbx1 coordinates addition of posterior second heart field progenitor cells Leschik, J., Stefanovic, S., Brinon, B. and Pucéat, M. (2008). Cardiac to the arterial and venous poles of the heart. Circ. Res. 115, 790-799. commitment of primate embryonic stem cells. Nat. Protoc. 3, 1381-1387. Stoller, J. Z., Huang, L., Tan, C. C., Huang, F., Zhou, D. D., Yang, J., Gelb, B. D. Liao, J., Aggarwal, V. S., Nowotschin, S., Bondarev, A., Lipner, S. and Morrow, and Epstein, J. A. (2010). Ash2l interacts with Tbx1 and is required during early B. E. (2008). Identification of downstream genetic pathways of Tbx1 in the second embryogenesis. Exp. Biol. Med. (Maywood) 235, 569-576. heart field. Dev. Biol. 316, 524-537. Verzi, M. P., Mcculley, D. J., De Val, S., Dodou, E. and Black, B. L. (2005). The Lindsay, E. A., Vitelli, F., Su, H., Morishima, M., Huynh, T., Pramparo, T., right ventricle, outflow tract, and ventricular septum comprise a restricted Jurecic, V., Ogunrinu, G., Sutherland, H. F., Scambler, P. J. et al. (2001). Tbx1 expression domain within the secondary/anterior heart field. Dev. Biol. 287, haploinsufficieny in the DiGeorge syndrome region causes aortic arch defects in 134-145. mice. Nature 410, 97-101. Waldo, K. L., Kumiski, D. H., Wallis, K. T., Stadt, H. A., Hutson, M. R., Platt, D. H. Merscher, S., Funke, B., Epstein, J. A., Heyer, J., Puech, A., Lu, M. M., Xavier, and Kirby, M. L. (2001). Conotruncal myocardium arises from a secondary heart R. J., Demay, M. B., Russell, R. G., Factor, S. et al. (2001). TBX1 is responsible field. Development 128, 3179-3188. for cardiovascular defects in velo-cardio-facial/. Cell 104, Wamstad, J. A., Alexander, J. M., Truty, R. M., Shrikumar, A., Li, F., Eilertson, 619-629. K. E., Ding, H., Wylie, J. N., Pico, A. R., Capra, J. A. et al. (2012). Dynamic and Mjaatvedt, C. H., Nakaoka, T., Moreno-Rodriguez, R., Norris, R. A., Kern, M. J., coordinated epigenetic regulation of developmental transitions in the cardiac Eisenberg, C. A., Turner, D. and Markwald, R. R. (2001). The outflow tract of the lineage. Cell 151, 206-220. heart is recruited from a novel heart-forming field. Dev. Biol. 238, 97-109. Xu, H., Morishima, M., Wylie, J. N., Schwartz, R. J., Bruneau, B. G., Lindsay, Moretti, A., Bellin, M., Jung, C. B., Thies, T.-M., Takashima, Y., Bernshausen, A., E. A. and Baldini, A. (2004). Tbx1 has a dual role in the morphogenesis of the Schiemann, M., Fischer, S., Moosmang, S., Smith, A. G. et al. (2010). Mouse cardiac outflow tract. Development 131, 3217-3227. Disease Models & Mechanisms

7