Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

TBX1 Represses Vegfr2 Gene Expression and Enhances the Cardiac Fate of VEGFR2+ Cells

Abstract

The T-box transcription factor TBX1 has critical roles in maintaining proliferation and inhibiting differentiation of cardiac progenitor cells of the second heart field (SHF). Haploinsufficiency of the gene that encodes it is a cause of congenital heart disease. Here, we developed an embryonic stem (ES) cell-based model in which Tbx1 expression can be modulated by tetracycline. Using this model, we found that TBX1 down regulates the expression of VEGFR2, and we confirmed this finding in vivo during embryonic development. In addition, we found a Vegfr2 domain of expression, not previously described, in the posterior SHF and this expression is extended by loss of Tbx1. VEGFR2 has been previously described as a marker of a subpopulation of cardiac progenitors. Clonal analysis of ES-derived VEGFR2+ cells indicated that 12.5% of clones expressed three markers of cardiac lineage (cardiomyocyte, smooth muscle and endothelium). However, a pulse of Tbx1 expression was sufficient to increase the percentage to 20.8%. In addition, the percentage of clones expressing markers of multiple cardiac lineages increased from 41.6% to 79.1% after Tbx1 pulse. These results suggest that TBX1 plays a role in maintaining a progenitor state in VEGFR2+ cells.

Introduction

The majority of the cardiac cells derives from two separate populations of progenitors named “heart fields” [1]. In the mouse, the two heart fields are specified during gastrulation [2,3]. The first heart field (FHF) contributes to the left ventricle and part of the atria, while the second heart field (SHF) contributes to the outflow tract (OFT), the right ventricle (RV) and part of the atria [4]. TBX1 is a transcription factor expressed in several tissues but its early expression in mesodermal tissue is particularly important for normal development of SHF-derived heart segments, especially the OFT [57]. Mesodermal-specific deletion of Tbx1 down regulates cell proliferation in the SHF [8] revealing its role in the expansion of cardiac progenitors [9]. In addition, Cre recombination-based cell tracing has shown that descendants of Tbx1-expressing cells populate myocardial and endocardial layers of the OFT and RV and, to a lesser extent the right atrium [5,10].

Chen and co-workers [9] showed that Tbx1 is expressed in and regulates proliferation of multipotent cardiac progenitors. In addition, they showed premature differentiation in the SHF of Tbx1-/- embryos. Furthermore, TBX1 is required for the addition of progenitor cells from the anterior SHF (an anteriorly located sub-population of SHF) to the OFT, and is also important for differentiation at the inflow tract of the heart (IFT) [11]. How TBX1 functions within multipotent cardiac progenitors and how it interacts with other factors important for multipotency are still open questions. Among the factors shown to be expressed in multipotent cardiac progenitors there is VEGFR2, also known as KDR or FLK1 [12]. VEGFR2 is required for hematopoietic and endothelial cell (EC) lineage development [13]. Conditional deletion of the Vegfr2 gene causes heart defects and loss of endocardial cells [14]. Clonal analysis of embryonic stem (ES) cell-derived VEGFR2+ cells showed that they are able to generate cardiomyocyte, vascular smooth muscle and endothelial cells in culture [15]. VEGFR2+ cells isolated from mouse embryos at E7.5 showed a gene expression profile with characteristics of both first and second heart fields [12]. In addition, in vitro differentiation of ES cells produced a population of multipotent cardiac progenitor cells, which expresses Vegfr2, Isl1, and Nkx2.5 as well as Tbx1 [16].

Here, we provide evidence that TBX1 is a negative regulator of Vegfr2 gene and protein expression in differentiating ES cells and in vivo. Loss of Tbx1 in mouse embryos causes expansion of a previously not described VEGFR2 expression domain. Furthermore, analysis of ES-derived clones showed that VEGFR2+ cells are prevalently EC progenitors but, upon induction of Tbx1 expression, they also expressed marker of cadiomyocyte and smooth muscle lineage. These data suggest that TBX1 supports multipotency of a subpopulation of VEGFR2+ cells.

Materials and Methods

Generation of the mES-Tbx1TetOFF Cell Line and Tissue Culture

Mouse ES EBRTc cells [17] were cultured in DMEM high glucose (Invitrogen Ltd, Paisley, UK, catalogue no. 11995–065) supplemented with 15% fetal bovine serum (HyClone "defined", Thermo Scientific, Logan, UT, USA, catalogue no. SH30070.03), 0.1 mM nonessential amino acids (Gibco-Brl, Invitrogen Ltd, Paisley, UK, catalogue no. 11140–050), 0.1 mM 2-mercaptoethanol (Sigma-Aldrich, St. Louis, MO, USA, catalogue no. M6250), and 1,000 U/ml ESGRO Leukemia Inhibitory Factor (LIF) (Millipore, Billerica, MA, USA, catalogue no. ESG1107), on gelatin-coated dishes. 2 × 106 EBRTc cells were transfected with the PTHC-Tbx1-myc plasmid and with the pCAGGS-Cre vector, as described [17]. Thirty puromycin-resistant clones were selected and eleven of them were screened by Southern blotting. One positive clone was selected for further analysis and named mES-Tbx1TetOFF. For embryoid body (EB)-based differentiation, cells were seeded in hanging drops (400 cells/drop) placed on the lids of tissue culture dishes and incubated for 2 days, and then cultured in suspension for 3 more days in Petri dishes. All steps were performed in complete DMEM medium, supplemented with 15% FBS, 7.5μg/ml tetracycline (Sigma, catalogue no. T7660) without LIF. 5-day-old EBs were plated on gelatin-coated plates for further analysis with or without tetracycline. For clonal analysis EBs were dissociated at day 4.75 using a kit (Miltenyi Biotec GmbH catalogue. n. 130-096-348) according to manufacturer’s instructions, VEGFR2+ cells were isolated using CD309 Miltenyi microbeads kit (Miltenyi Biotec GmbH catalogue. n. 130-090-858). VEGFR2+ cells were plated at clonal density (104 cells/60mm plate) in media with LIF for 7 days. Individually isolated colonies were then picked, trypsinized and plated in individual 96 wells, grown for 3 days and then divided in two plates, Tet+ and Tet-. After 3 days in culture, RNA was extracted and processed for RT-PCR.

P19Cl6 cells [18] were cultured in alpha-MEM, 10% FBS and 1% Glutamine. For differentiation 5x105 cells were plated on a 35-mm tissue culture dish. After one day, the medium was replaced with growth media containing 10μM 5-Azacytidine for 24 h. From the next day, cells were incubated in growth media containing 1.0% DMSO that was changed daily. Day 0 of differentiation is defined as the day of 5-Aza addition to the media.

For siRNA-mediated knockdown of Tbx1, cells were cultured in 35 mm dishes and transfected at day 0 with siRNA (LifeTecnology n. 4390771 s-74767; s-74769) using Lipofectamine-RNAi-MAX transfection reagent (LifeTechnology) according to the manufacturer protocol. Non-targeting siRNA was used for control transfections. Cells were harvested at day 1 for RNA extraction and reverse transcription.

RNA Isolation

Total RNA was isolated with TRIZOL (Invitrogen) and reverse-transcribed using the High Capacity cDNA reverse transcription kit (Applied Biosystem catalogue. n. 4368814). Quantitative real-time PCR (qRT-PCR) was performed using SYBR Green PCR master mix (Applied Biosystem). Relative gene expression was evaluated using the 2–ΔΔCt method, and Gapdh expression as normalizer. Primers are listed in S1 Table. Expression data are shown as the mean ± SEM. Statistical analysis was performed using the Student’s t-test for expression data, and chi-square test for clonal analysis.

Immunohistochemistry and In Situ Hybridization

Animal research was conducted according to EU and Italian regulations. The animal protocol has been approved by the animal welfare committee of the Institute of Genetics and Biophysics (Organismo per il Benessere Animale or OPBA-IGB), protocol n.: 0002183 (June 4, 2013). Tbx1+/- mice [19] were crossed to obtain Tbx1-/- embryos. Genotyping was performed as in the original report. Embryos were fixed in 4% paraformaldeyde (PFA)/PBS at 4°C overnight and embedded in OCT. 10 μm sagittal and transverse sections were cut using a cryostat and were post-fixed in 4% PFA at room temperature for 5 min prior to immunostaining. Sections were briefly rinsed with PBS, treated with 0.5% H2O2 in ethanol to block endogenous peroxidase activity and blocked in 20% Goat Serum (GS) in PBS with 0.05% Tween for at least 30’ at room temperature, incubated in biotin anti mouse CD309 (VEGFR2)(Biolegend) overnight at 4°C in 5% GS in PBS, 0.05%Tween; the signal of biotin anti mouse CD309 antibody was enhanced using Vectastain Elite-ABC kit reaction (PK 6200, Vector Laboratories, Burlingame, CA) and visualized using DAB Peroxidase (HRP) Substrate Kit (SK-4100 Vector Laboratories, Burlingame, CA). For in situ hybridization, Vegfr2 or Tbx1 anti-sense RNA probes were labeled using the DIG-RNA labeling kit (Roche) and hybridized to cryosections following published methods [20]. Digital photographs were taken with a Leica DM6000 microscope and acquisition software LAS V4.1.

Flow Cytometry and Immunofluorescence

EBs were dissociated into single cell suspension using Trypsin-EDTA solution. 1.5-2x106/ml cells were collected and fixed for 30 min at 4°C in 70% cold EtOH (anti-NKX2.5 and anti-GATA4 antibodies), or 10 min at RT in 4%PFA (anti-P-H3 antibodies). Samples were permeabilized in 0.5%NP-40/PBS /1%BSA buffer for 10 min with slow shaking at RT, and incubated with the primary antibody (1:50 for the anti-NKX2.5 and anti-GATA4; 1:100 for the anti-P-H3) for 30 min at RT in 0.1%NP-40/PBS/1%BSA. After washing in 0.1%NP-40/PBS/1%BSA, samples were incubated with 1:500 anti-goat FITC or anti-mouse Texas Red for 15 min at RT. All washings were performed in PBS/1%BSA at RT. Flow cytometry with anti VEGFR2 antibodies was performed according to manufacturer's protocol (Biolegend). Immunofluorescence was performed on cells plated on gelatin-treated chamber-slides (Permanox, ThermoScientific) at day 5 of differentiation, and fixed at day 6 for anti-P-H3 and day 8 for VEGFR2. Antibodies used are listed in S1 Table.

Results

Generation of a Dynamic Model of Tbx1 Gene Expression

We have inserted a Tbx1 cDNA into the Rosa 26 locus of mouse embryonic stem cells EBRTc using a docking system that includes a TetOff regulation construct [17]. The strategy is illustrated in Fig 1A. The resulting engineered ES cell line is hereafter referred to as mES-Tbx1TetOff. This cell line expresses Tbx1 upon removal of tetracycline (Fig 1B; average of 3 different experiments). Tbx1 RNA was detectable in undifferentiated cells 24 hrs after tetracycline removal from ES media (Fig 1B). Transgene Tbx1 expression was also confirmed by using specific primers in the 3’-tagged region (S1 Fig). Both induced and not induced cells were able to differentiate into beating cells (S1 Video and S2 Video).

thumbnail
Fig 1. Generation of the ES cell line inducible system mES-Tbx1TetOFF.

A: top, docking site at the Rosa-Tet locus of the EBRTc cell line [17]; middle: exchange vector (PTHC-Tbx1-myc) carrying a Tbx1 cDNA; bottom: recombinant allele after Cre-induced recombination. B: qRT-PCR analysis of Tbx1 expression in undifferentiated mES-Tbx1TetOFF cells. Values are relative to that at tetracycline removal time (0h) and are the average of three different experiments. Error bar: S.E.M.

https://doi.org/10.1371/journal.pone.0138525.g001

TBX1 Enhances Cell Proliferation and Modulates the Expression of Cardiac Differentiation Markers

To test the effect of Tbx1 expression in our ES cell system, we have used mES-Tbx1TetOff cells in the EB-based differentiation protocol illustrated in Fig 2A. In this system, endogenous Tbx1 expression was detected at day 5 of differentiation, reached a peak at day 6 and then it decreased (Fig 2B). To enhance Tbx1 expression, we removed tetracycline from the tissue culture media at day 5 for 24 hrs and then continued the differentiation protocol in the presence of tetracycline (Fig 2C). Under these conditions, Tbx1 expression was enhanced by approximately 3 folds at day 6 (Fig 2D). The number of mitotic cells was quantified at day 6 using an antibody anti-P-H3 by immunofluorescence (Fig 2E) and by flow cytometry. Fig 2F shows the average of three experiments. The increased mitotic activity in cells subjected to Tbx1 expression pulse is consistent with our previous data [9] and indicates that the protein encoded by the transgene is biologically active.

thumbnail
Fig 2. Differentiation assay of the mES-Tbx1TetOFF cell line.

A: Scheme of experimental protocol. B: qRT-PCR assay of Tbx1 expression in uninduced (+Tet) mES-Tbx1Tet OFF cells. A peak of expression is evident at day 6. C: Schematic representation of the experimental strategy used for pulse Tbx1 expression. D: qRT-PCR analysis of Tbx1 expression at day 6 after 24hrs without tetracycline. E: Immunofluorescence with an anti-P-H3 antibody on two colonies of mES-Tbx1Tet OFF cells with and without tetracycline at day 6. F: graphic representation of quantitative evaluation of mitotic activity using flow cytometry.

https://doi.org/10.1371/journal.pone.0138525.g002

Next, we investigated the effects of increased Tbx1 expression on cell differentiation. To this end, we tested the expression of cardiac and endothelial lineage differentiation markers Nkx2.5, Isl1, Gata4, Vegfr2, Mef2c and Mlc2v by quantitative real time reverse-transcription PCR (qRT-PCR) at days 6, 8, and 11, of differentiation using the scheme indicated in Fig 2C, with and without a 24hrs-pulse at day 5. At day 6 Gata4 expression was down regulated in cells subjected to Tbx1 pulse, while the other markers did not change significantly (Fig 3A). At day 8, Nkx2.5 was up regulated while Mef2c, Vegfr2 and Isl1 were down regulated. At day 11, Nkx2.5 and Mlc2v were up regulated while Vegfr2 and Isl1 were down regulated compared to controls without pulse (Fig 3A). At day 8, flow cytometry, confirmed and quantified the reduction of VEGFR2+ cells, and showed reduction of GATA4+ cells and increase of NKX2.5+ cells (Fig 3B and 3C). The reduction of VEGFR2 expression was also visualised by immunofluorescence, at the same day (Fig 3D). Thus, after a Tbx1 expression pulse, differentiating ES cells have an "early" response (detected soon after the pulse at day 6) characterized by down-regulation of Gata4, and a "late" response (detectable after 48 hrs from the pulse, day 8) characterized by the expansion of the NKX2.5+ cell population and shrinkage of the VEGFR2+ and GATA4+ cell populations. We were intrigued by the suppressive effect of Tbx1 over expression on Vegfr2 expression. Vegfr2 is a marker of EC progenitors and hematopoietic cells, but it also identifies a sub-population of cardiac progenitors [13,21,22]. Thus, we analysed the impact of Tbx1 loss of function (by siRNA knock down) on Vegfr2 expression in an independent cell differentiation model. S2A Fig shows the expression of Tbx1 and Vegfr2 during differentiation of P19Cl6 cells [18], note the nearly complementary pattern. We knocked down Tbx1 expression at day 0 and found that at day 1 the expression of Vegfr2 was significantly increased in 3 repeated experiments (S2B' Fig), confirming that Tbx1 is a negative regulator of Vegfr2.

thumbnail
Fig 3. Tbx1 expression pulse at day 5 produces persistent changes of expression of cardiac markers.

A: qRT-PCR assay at Day 6, Day 8 and Day 11 of mES-Tbx1Tet OFF cells with (red) and without (blue) pulse at Day 5. The data are represented as mean of fold change relative to Day 5 value (not shown) of four different experiments. Error bars indicate SEM. (* = p-value<0.05; ** = p-value <0.005; *** = p-value <0.0005). B: Flow cytometric evaluation of the effects of Tbx1 pulse on VEGFR2, NKX2.5 and GATA4 cell population at Day 8. Flow cytometry at Day 8 shows a decrease of the numbers of VEGFR2+ and GATA4+ cells (B, B”) and an increase of the number of NKX2.5+ cells (B’) after Tbx1 pulse. C: The histograms show the mean percentage of positive cells in three different experiments. Error bar indicate SEM. D: Immunofluorescence detection of VEGFR2 with (Tet-) or without (Tet+) Tbx1 pulse. FSC: Forward Scatter.

https://doi.org/10.1371/journal.pone.0138525.g003

VEGFR2+ Cells Express the Cardiac Marker Smarcd3 in Response to Tbx1 Expression

We have followed up the relationship between Tbx1 and Vegfr2 and explored the destiny of VEGFR2+ cells following transient expression of Tbx1, we have subjected mES-Tbx1TetOff cells to the differentiation protocol shown in Fig 2C, but cells were harvested at day 4.75 (as in [12]). At this time of differentiation, the percentage of VEGFR2+ cells was about 15% of total as assayed by flow cytometry (S3 Fig). Then we isolated VEGFR2+ cells using magnetic beads, we tested markers of cardiac and endothelial lineages in the VEGFR2-enriched cell population, and compared their expression with that of the unbound cell population (Fig 4A and 4B). VEGFR2+ cells were positive for all the markers tested (including Tbx1), with the exception of Smarcd3, a muscle differentiation marker (Fig 4B). In contrast, the unbound population expressed Smarcd3 but did not express Tbx20 (Fig 4B). Next, we performed another experiment using the same scheme, but the VEGFR2-enriched cell population was split in two samples, one of which was subjected to Tbx1 induction by withdrawing tetracycline for 24 hrs. After 24hrs of induction, we extracted RNA and performed marker analyses. Results revealed changes of Vegfr2 and Smarcd3 expression (Fig 4D and 4E) but the other markers showed a similar level of expression as tested by standard RT-PCR, (Fig 4F). These results suggest that Tbx1 expression in VEGFR2+ cells from differentiating ESCs may confer or reinforce a muscle differentiation potential.

thumbnail
Fig 4. Expression profile of VEGFR2+ cells isolated at Day 4.75 of differentiation from mES-Tbx1TetOff cells and subjected to additional 48hrs in culture, with or without tetracycline.

A: qRT-PCR assay of Vegfr2 expression of cells bound to magnetic beads (VEGFR2+ cells) and unbound (UN). B: RT-PCR analysis of VEGFR2+ and unbound cells. C: qRT-PCR of Tbx1 expression in VEGFR2+ cells. D,E: qRT-PCR evaluation of Vegfr2 and Smarcd3 expression in VEGFR2+ cells with and without Tet.

https://doi.org/10.1371/journal.pone.0138525.g004

Clonal Analysis: VEGFR2+ Cells Are Heterogeneous, Plastic, and Their Fate Can Be Modulated by Tbx1 Expression in Tissue Culture

To explore further the ability of TBX1 to “push”VEGFR2+ cells toward a cardiac fate, we performed clonal analysis. The experimental procedure is outlined in Fig 5A. Briefly, at day 5 of differentiation, VEGFR2+ cells were isolated and plated at clonal density. They were grown in the presence of LIF and after 7 days we picked individual, well isolated colonies, and seeded them in 96-well plates. After one day, plates were duplicated and cultured with and without tetracycline for 24 hrs. After additional 3 days of culture in the absence of LIF we harvested cells and extracted RNA to assay expression by RT-PCR. Vegfr2 expression was retained in 58.3% (n = 28) of control clones tested (n = 48) and in 12.5% (n = 6) of clones subjected to Tbx1 pulse. Next, we tested the expression of cardiac lineage markers Nkx2.5, Pecam1, and Acta2 indicative of cardiomyocyte precursors, EC and smooth muscle cells, respectively (examples of raw data are shown in Fig 5B). A summary of results is shown schematically in Fig 5C, which includes only the clones positive for at least one of those markers (n = 48). In the absence of Tbx1 pulse, 41.6% of clones (n = 20) expressed only the endothelial marker Pecam1. 12.5% of clones (n = 6) expressed two markers, Nkx2.5 and Pecam1 and 12.5% of clones (n = 6) expressed all three marker. These results indicate that under the conditions tested, the most prevalent profile is endothelial, while a relatively low percentage of clones shows a cardiac progenitor profile (triple positive clones).

thumbnail
Fig 5. Clonal analysis of VEGFR2+ cells.

A: schematic representation of experimental strategy. B: Examples of RT-PCR analysis on VEGFR2+ cells in absence and in presence of Tbx1 pulse. C: Color map panels of marker positivity of individual clones with or without Tbx1 pulse. D: Graphic representation of clone multiplicity, based on the number of positive markers with and without tetracycline. Blue represents the percentage of clones that were positive for all three markers; Red represents the percentage of clones positive for two markers; Green represents the percentage of clones that are positive for one marker.

https://doi.org/10.1371/journal.pone.0138525.g005

In contrast, the same clones, after Tbx1 pulse, exhibited a more diverse pattern (Fig 5C, right panel). In particular, only 16.6% of clones (n = 8) expressed Pecam1 alone, and about 33% of clones (n = 16) were double positive for Nkx2.5 and Pecam1, and 20.8% of clones (n = 10) was triple positive (Fig 5C). The overall number of Pecam1+ clones (independently from the expression of other markers) did not change significantly with the Tbx1 pulse (40 vs. 44), thus Tbx1 is not a Pecam1 gene suppressor. The most evident change is the multiplicity of markers expressed by clones that have undergone Tbx1 pulse (Fig 5D). Indeed, the percentage of clones positive for at least two markers was 41.6% (n = 20) without Tbx1 pulse, but increased to 79.1% (n = 38) with induction (p = 0.000172; chi square-test). The increased multiplicity is due to an increase of the number of clones positive for Nkx2.5 (from 18 to 30; p = 0.0143) and to a reduction of the number of clones expressing only Pecam1 (from 20 to 8; p = 0.0070).

These data suggest that forced Tbx1 expression increases the likelihood of expression of multiple cardiac markers in clones derived from VEGFR2+ cells.

Vegfr2 Is Expressed in the Posterior Second Heart Field (pSHF) and Its Expression Domain Is Expanded in Tbx1-/- Mouse Mutants

To validate our in vitro findings, we tested whether TBX1 has an effect on Vegfr2 expression in vivo. At E9.5, Vegfr2 is mostly expressed in EC but we found that it is also expressed in the mesodermal/epithelial-like tissue of the pSHF (arrows in Fig 6A and 6B) in the proximity of the IFT and in the dorsal mesenchyme protrusion region (DMP) (arrowheads in Fig 6A and 6B). In Tbx1-/- embryos, the Vegfr2 gene or protein expression domains of the pSHF and DMP region expanded more dorsally and anteriorly (Fig 6A, 6A', 6B and 6B', examples from a total of 3 embryos analyzed). Moreover, we observed that in transverse sections, VEGFR2 immunostaining was asymmetric in WT embryos, but was symmetric in Tbx1-/- embryos (Fig 6C-C'). The expression of Vegfr2 in the pSHF has not been reported to date. Tbx1 is not expressed in the pSHF in the inflow region (S4 Fig), while Vegfr2 is detected in this region at E8.5–9.0 (Fig 6). Therefore, we have analysed the expression patterns of Vegfr2 and Tbx1 at an earlier embryonic stage. At E8.5 (8 somites) Tbx1 was strongly expressed in the pharyngeal mesoderm while Vegfr2 was also expressed in the pharyngeal mesoderm but to a lesser extent (arrows in Fig 7A' and 7D'). Immunohistochemistry at the same developmental stage confirmed the presence of the VEGFR2 protein in the pharyngeal mesoderm (Fig 7E and 7F) and showed the expansion of this expression domain in Tbx1-/- embryos (Fig 7E and 7F'). Immunostaining also revealed that at E9.5, VEGFR2 expression in the Ist pharyngeal arch is expanded in Tbx1-/- embryos, compared to wild type (S5 Fig).

thumbnail
Fig 6. Vegfr2 expression in wild type and Tbx1-/- mouse embryos E9.5 and E9.0.

A, A’: In situ hybridization of wild type and Tbx1-/- embryos. B-C': Immunohistochemisty with an anti-VEGFR2 antibody on sagittal and transverse sections. The arrows indicate the expression domain in the pSHF. The arrowheads indicate the DMP region (n = number of embryos examined). Scale bars: 200 μm in A, A’, B, B’. 50 μm in C, C’. s: number of somites.

https://doi.org/10.1371/journal.pone.0138525.g006

thumbnail
Fig 7. Vegfr2 and Tbx1 expression in E8.5 mouse embryos.

A-D': In situ hybridization of Tbx1 (A-D) or Vegfr2 (A'-D') shown in a rostro-caudal series of transverse sections of wild type embryos. Arrows indicate Vegfr2 expression in the pharyngeal mesenchyme. E-F: VEGFR2 immunohistochemistry in similar sections as in A and B. G-H: VEGFR2 immunohistochemistry in corresponding sections of a Tbx1-/- embryo. Arrows indicate VEGFR2+ cells in the pharyngeal mesenchyme. OFT: cardiac outflow tract. IFT: cardiac inflow tract. Scale bars: 200 μm in A-D, A’-D’. 50 μm in E-H. s: number of somites.

https://doi.org/10.1371/journal.pone.0138525.g007

Overall, our data show that TBX1 is a negative regulator of VEGFR2 expression during mouse embryogenesis in specific tissues.

Discussion

TBX1 plays a key role in the control of proliferation and differentiation of cardiac progenitor cells of the SHF, and in tissue culture it marks cells with cardiomyocyte, smooth muscle and endothelial differentiation potential [9]. In this work we provide the first evidence that TBX1 suppresses (directly or indirectly) the expression of the Vegfr2 gene in differentiating ES cells and during embryonic development. How does TBX1 regulate Vegfr2 gene expression? TBX1 may directly bind the Vegfr2 gene. Indeed, computational analyses using the TRANSFAC database identified over 30 potential T-box binding sites within a 50Kb DNA segment centered on the gene (data not shown). It is also possible that the regulation is indirect. Indeed, our time-course expression analyses after Tbx1 induction (Fig 3) showed that the expression of Gata4 is down regulated earlier than that of Vegfr2, and it has been shown that Gata4 regulates positively the Vegfr2 gene [23], thus it is possible that the regulation of Vegfr2 is mediated by Gata4 regulation. It has been shown that Gata4 expression is up regulated in Tbx1-/- embryos [24] and is down regulated in Tbx1 overexpressing embryos [25]. Vegfr2 is expressed in hematopoietic, endothelial and cardiac progenitors [12,26]. Conditional deletion of mouse Vegfr2 in the endothelium (using an endothelial-specific Cre driver) causes embryonic lethality at E9.5-E10.5 associated with loss of the endocardium and hypoplasia of the OFT and RV [14]. This suggests that VEGFR2 (or the endocardium) has a non-autonomous role in the development of SHF-derived structures. A more recent study [27] has demonstrated that endothelial cells signal to cardiac neural crest-derived cells to support their migration. However, we found expression of Vegfr2 in the pSHF (but not in the anterior SHF) as late as E9.5 and in the pharyngeal mesoderm at earlier stages. In addition, we found Vegfr2 expression in the DMP, an SHF derivative that gives rise to the atrioventricular mesenchymal complex [28]. These expression data suggest that VEGFR2 may have a role in the SHF development independent from its role in the endocardium. Indeed, conditional deletion of Vegfr2 in the Isl1 expression domain, which includes the SHF, causes embryo lethality at E14.5 associated with cardiac defects but with preserved endocardium [14].

It has been shown that the pSHF and DMP of Tbx1-/- embryos have altered expression profile, and the DMP is hypoplastic, which may be the cause of the atrioventricular septal defects (AVSDs) found in these mutants [11]. Whether the expansion of the VEGFR2 domain in the pSHF and DMP of Tbx1-/- embryos could lead to AVSD is unknown because there is no information as to the phenotypic consequences of excessive VEGFR2 expression. However, in quail, increased VEGF signalling causes endocardial cell hyperplasia in the dorsal mesocardium region and affects heart development [29]. Future studies using transgenic mice should address whether or not VEGFR2 expansion contributes to the Tbx1 mutant phenotype.

Is there a specific role of TBX1 in the fate of VEGFR2-expressing cells? Our tissue culture experiments using differentiating ES cells suggest that TBX1 works as a suppressor of Vegfr2 expression. Overexpression of Tbx1 in isolated VEGFR2+ cells also caused up regulation of Smarcd3 expression, a marker of cardiac and skeletal muscle differentiation. Together these two findings suggest that TBX1 may favour cardiac fate at the expense of endothelial fate. However, clonal analysis of VEGFR2+ cells provided a more complex picture. Indeed, in basal conditions, clones derived from VEGFR2+ cells showed a prevalent endothelial fate. Transient induction of Tbx1 expression did not suppress the endothelial marker gene Pecam1 but was sufficient to activate, in many clones, Nkx2.5 gene expression. Although some of the transcriptional changes of clones might be secondary to genetic drift, the overall trend is significant. These results could be interpreted in at least two different ways: A) Tbx1 acts as an anti-differentiation factor and its overexpression tends to preserve a progenitor status; B) TBX1 acts as a “simple” activator of Nkx2.5 gene expression. The latter interpretation, however, is consistent with previous findings showing down regulation of Nkx2.5 in Tbx1-/- mutant [24]. The mechanism by which Tbx1 may maintain a progenitor state is not clear. One possibility is that TBX1 induces a permissive chromatin state by interacting with the BAF/ SWI-SNF chromatin remodelling complex [30] and H3 lysine 4 methyltrasferase enzymes [30,31]. Another possibility is that the BMP-suppressive activity of TBX1, mediated by interaction with SMAD1 [32], may result in an anti-differentiation activity, as suggested by a study of self renewal of skin stem cells [33]. Our work identifies VEGFR2+ cells as a model for studying the plasticity of cardiac progenitors and for the identification of molecular players that maintain their progenitor status. We propose that TBX1 is a candidate for this function.

Supporting Information

S1 Fig. Quantitative real time PCR to evaluate transgene expression after Tbx1 induction for 24 hrs, 48hrs and 72hrs.

https://doi.org/10.1371/journal.pone.0138525.s001

(TIF)

S2 Fig. Tbx1 knock down in P19Cl6 induces upregulation of Vegfr2 expression.

A-B: qRT-assay of Tbx1 and Vegfr2 expression during P19Cl6 differentiation. Note that Tbx1 expression peaks at day 1 (A) while Vegfr2 expression increases at day 2 (B). qRT-PCR assays of Tbx1 (A') and Vegfr2 (B') expression at day 1 after knock down of Tbx1 at day 0.

https://doi.org/10.1371/journal.pone.0138525.s002

(TIF)

S3 Fig. VEGFR2+ cell quantification in differentiating mES-Tbx1TetOFF cells. by FACS analysis at 4.75 day of differentiation.

https://doi.org/10.1371/journal.pone.0138525.s003

(TIF)

S4 Fig. Tbx1 expression pattern in wild type embryos.

In situ hybridizations of Tbx1 are shown in two consecutive transverse sections near the venous pole of the heart, the arrows indicate the pSHF region. Wild type, 16 somite embryo (approx. E9.0). Scale bars: 50 μm s: number of somites. IFT: inflow tract; Ph: pharynx.

https://doi.org/10.1371/journal.pone.0138525.s004

(TIF)

S5 Fig. VEGFR2 expression in wild type and Tbx1-/- embryos.

Immunoistochemistry of VEGFR2 shows expansion of expression in Tbx1 null embryos. The panels show rostral, medial and caudal transverse sections of the pharyngeal region of wild type and Tbx1-/- embryos at three different somite stages. Scale bars: 50 μm. s: number of somites.

https://doi.org/10.1371/journal.pone.0138525.s005

(TIF)

S1 Table. List of primers and antibodies used.

https://doi.org/10.1371/journal.pone.0138525.s006

(XLSX)

S1 Video. Differentiated mES-Tbx1TetOFF cell.

The colonies showing pulsing activity without TBX1 induction (+Tet).

https://doi.org/10.1371/journal.pone.0138525.s007

(AVI)

S2 Video. Differentiated mES-Tbx1TetOFF cell.

The colonies showing pulsing activity with TBX1 induction (-Tet).

https://doi.org/10.1371/journal.pone.0138525.s008

(AVI)

Acknowledgments

We thank Drs. Kary Alitalo for the Vegfr2 in situ hybridization probe, and Gilda Cobellis for help with the EBRTc cell line. We thank Drs. Cinzia Caprio for providing reagents. We thank the core facilities of the Institute of Genetics and Biophysics (FACS, Integrated Microscopy and Mouse transgenic). This project was funded by grants from the Telethon Foundation GGP14211, and from the Italian Ministry of Education and Research (MIUR) PON_01–02342 and FareBio (to AB).

Author Contributions

Conceived and designed the experiments: GL AB. Performed the experiments: GL RF. Analyzed the data: GL AB. Contributed reagents/materials/analysis tools: GL. Wrote the paper: GL AB.

References

  1. 1. Vincent SD, Buckingham ME. How to make a heart. The origin and regulation of cardiac progenitor cells. Curr Top Dev Biol. 2010;90: 1–41. pmid:20691846
  2. 2. Lescroart F, Chabab S, Lin X, Rulands S, Paulissen C, Rodolosse A, et al. Early lineage restriction in temporally distinct populations of Mesp1 progenitors during mammalian heart development. Nat Cell Biol. 2014;
  3. 3. Devine WP, Wythe JD, George M, Koshiba-Takeuchi K, Bruneau BG. Early patterning and specification of cardiac progenitors in gastrulating mesoderm. Elife. 2014;3: 1–23.
  4. 4. Watanabe Y, Buckingham M. The formation of the embryonic mouse heart: heart fields and myocardial cell lineages. Ann N Y Acad Sci. 2010;1188: 15–24. pmid:20201881
  5. 5. Xu H, Cerrato F, Baldini A. Timed mutation and cell-fate mapping reveal reiterated roles of Tbx1 during embryogenesis, and a crucial function during segmentation of the pharyngeal system via regulation of endoderm expansion. Development. 2005;132: 4387–4395. pmid:16141220
  6. 6. Xu H, Morishima M, Wylie JN, Schwartz RJ, Bruneau BG, Lindsay EA, et al. Tbx1 has a dual role in the morphogenesis of the cardiac outflow tract. Development. 2004;131: 3217–3227. pmid:15175244
  7. 7. Vitelli F, Morishima M, Taddei I, Lindsay EA, Baldini A. Tbx1 mutation causes multiple cardiovascular defects and disrupts neural crest and cranial nerve migratory pathways. Hum Mol Genet. 2002;11: 915–922. pmid:11971873
  8. 8. Zhang Z, Huynh T, Baldini A. Mesodermal expression of Tbx1 is necessary and sufficient for pharyngeal arch and cardiac outflow tract development. Development. 2006;133: 3587–3595. pmid:16914493
  9. 9. Chen L, Fulcoli FG, Tang S, Baldini A. Tbx1 regulates proliferation and differentiation of multipotent heart progenitors. Circ Res. 2009;105: 842–851. pmid:19745164
  10. 10. Huynh T, Chen L, Terrell P, Baldini A. A fate map of Tbx1 expressing cells reveals heterogeneity in the second cardiac field. Genesis. 2007;45: 470–475. pmid:17610275
  11. 11. Rana MS, Theveniau-Ruissy M, De Bono C, Mesbah K, Francou A., Rammah M, et al. Tbx1 Coordinates Addition of Posterior Second Heart Field Progenitor Cells to the Arterial and Venous Poles of the Heart. Circ Res. 2014;115: 790–799. pmid:25190705
  12. 12. Kattman SJ, Huber TL, Keller GM. Multipotent Flk-1+ Cardiovascular Progenitor CellsGive Rise to the Cardiomyocyte, Endothelial, and Vascular Smooth Muscle Lineages. Dev Cell. 2006;11: 723–732. pmid:17084363
  13. 13. Ema M, Takahashi S, Rossant J. Deletion of the selection cassette, but not cis-acting elements, in targeted Flk1-lacZ allele reveals Flk1 expression in multipotent mesodermal progenitors. Blood. 2006;107: 111–117. pmid:16166582
  14. 14. Milgrom-Hoffman M, Harrelson Z, Ferrara N, Zelzer E, Evans SM, Tzahor E. The heart endocardium is derived from vascular endothelial progenitors. Development. 2011;138: 4777–4787. pmid:21989917
  15. 15. Yamashita J, Itoh H, Hirashima M, Ogawa M, Nishikawa S, Yurugi T, et al. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature. 2000;408: 92–96. pmid:11081514
  16. 16. Moretti A, Caron L, Nakano A, Lam JT, Bernshausen A, Chen Y, et al. Multipotent Embryonic Isl1+ Progenitor Cells Lead to Cardiac, Smooth Muscle, and Endothelial Cell Diversification. Cell. 2006;127: 1151–1165. pmid:17123592
  17. 17. Masui S, Shimosato D, Toyooka Y, Yagi R, Takahashi K, Niwa H. An efficient system to establish multiple embryonic stem cell lines carrying an inducible expression unit. Nucleic Acids Res. 2005;33: 1–8.
  18. 18. Ogawa K, Mueller I, Kobayashi R, Nakajima T, Ishii M. Effective and steady differentiation of a clonal derivative of P19CL6 embryonal carcinoma cell line into beating cardiomyocytes. J Biomed Biotechnol. 2010;2010.
  19. 19. Lindsay EA, Vitelli F, Su H, Morishima M, Huynh T, Pramparo T, et al. Tbx1 haploinsufficieny in the DiGeorge syndrome region causes aortic arch defects in mice. Nature. 2001;410: 97–101. pmid:11242049
  20. 20. Tiveron MC, Hirsch MR, Brunet JF. The expression pattern of the transcription factor Phox2 delineates synaptic pathways of the autonomic nervous system. J Neurosci. 1996;16: 7649–7660. pmid:8922421
  21. 21. Motoike T, Markham DW, Rossant J, Sato TN. Evidence for novel fate of Flk1+ progenitor: Contribution to muscle lineage. Genesis. 2003;35: 153–159. pmid:12640619
  22. 22. Martin-Puig S, Wang Z, Chien KR. Lives of a Heart Cell: Tracing the Origins of Cardiac Progenitors. Cell Stem Cell. 2008;2: 320–331. pmid:18397752
  23. 23. Ishitobi H, Wakamatsu A, Liu F, Azami T, Hamada M, Matsumoto K, et al. Molecular basis for Flk1 expression in hemato-cardiovascular progenitors in the mouse. Development. 2011. pp. 5357–5368. https://doi.org/10.1242/dev.065565 pmid:22071109
  24. 24. Liao J, Aggarwal VS, Nowotschin S, Bondarev A, Lipner S, Morrow BE. Identification of downstream genetic pathways of Tbx1 in the second heart field. Dev Biol. 2008;316: 524–537. pmid:18328475
  25. 25. Pane LS, Zhang Z, Ferrentino R, Huynh T, Cutillo L, Baldini A. Tbx1 is a negative modulator of Mef2c. Hum Mol Genet. 2012;21: 2485–2496. pmid:22367967
  26. 26. Lacaud G, Keller G, Kouskoff V. Tracking mesoderm formation and specification to the hemangioblast in vitro. Trends in Cardiovascular Medicine. 2004. pp. 314–317. pmid:15596108
  27. 27. Milgrom-Hoffman M, Michailovici I, Ferrara N, Zelzer E, Tzahor E. Endothelial cells regulate neural crest and second heart field morphogenesis. Biol Open. 2014; 1–10.
  28. 28. Briggs LE, Kakarla J, Wessels A. The pathogenesis of atrial and atrioventricular septal defects with special emphasis on the role of the dorsal mesenchymal protrusion. Differentiation. 2012. pp. 117–130. https://doi.org/10.1016/j.diff.2012.05.006 pmid:22709652
  29. 29. Drake CJ, Wessels A, Trusk T, Little CD. Elevated vascular endothelial cell growth factor affects mesocardial morphogenesis and inhibits normal heart bending. Dev Dyn. 2006;235: 10–18. pmid:16170779
  30. 30. Chen L, Fulcoli FG, Ferrentino R, Martucciello S, Illingworth EA, Baldini A. Transcriptional control in cardiac progenitors: Tbx1 interacts with the BAF chromatin remodeling complex and regulates Wnt5a. PLoS Genet. 2012;8.
  31. 31. Stoller JZ, Huang L, Tan CC, Huang F, Zhou DD, Yang J, et al. Ash2l interacts with Tbx1 and is required during early embryogenesis. Exp Biol Med (Maywood). 2010;235: 569–576.
  32. 32. Fulcoli FG, Huynh T, Scambler PJ, Baldini A. Tbx1 regulates the BMP-smad1 pathway in a transcription independent manner. PLoS One. 2009;4.
  33. 33. Chen T, Heller E, Beronja S, Oshimori N, Stokes N, Fuchs E. An RNA interference screen uncovers a new molecule in stem cell self-renewal and long-term regeneration. Nature. 2012;485: 104–108. pmid:22495305