Animal models
All experiments were conducted in accordance with the ethical regulations for animal research of the University of Malaga (UMA), the French Ministry of Higher Education and Research and of the EU and were approved by the Ethics Committee of UMA. Most experiments were conducted on mice with a CD1 background, including the single-cell analysis of the PE, spatial transcriptomics, RNA in situ hybridisation experiments and in vitro cell/tissue cultures. The Wt1-CreERT2 mouse strain expresses the CreERT2 fusion protein in the presence of tamoxifen or its derivatives under the control of the Wt1 gene promoter37. The RosaR26R-EYFP line58 was used to tag Wt1-lineage cells. All embryos were staged according to vaginal plug observation time point (E0.5 stage). Specific mouse strains were genotyped via PCR as documented in the original reports.
To induce recombination, pregnant Wt1-CreERT2;R26R-EYFP females received a single intraperitoneal administration of 4-hydroxytamoxifen (4OHT) (Sigma, H6278) dissolved in Cremophor® (Sigma, C5135)59 at E8.75 stage. Doses of 1 mg of 4OHT per mouse were used (300 µL total administration volume).
Ex vivo E11.5 heart culture
E11.5 embryos were collected from either CD1 or Wt1-CreERT2;R26R-EYFP mouse lines, washed in sterile PBS, and their pericardium removed. Hearts were then excised and placed individually in 96-round bottom well plates (Deltalab). Hearts were incubated at 37ºC and 5% CO2 in a medium comprising DMEM (Gibco, 11965) supplemented with 10% FBS (Pansera, P30-2602), 1% L-glutamine (Sigma, G7513), 1% penicillin/streptomycin (Gibco, 15140130), and 2 ng/mL bFGF (R&D Systems, 233-FB) as previously described 60. To arrest the cell cycle, the experimental growth medium was supplemented with 500 ng/mL nocodazole (Sigma, M1404). Hearts were cultured for a total of 24h.
In vitro culture of epicardial explants and EPICs
To grow primary epicardial cells, hearts from E11.5 embryos were excised and cultured in 4-well plates (Nunc) on glass coverslips coated with 0.1% gelatine (Panreac). Cells were incubated at 37ºC and 5% CO2 in the standard growth medium described above. Initially, a minimal volume of growth medium was used to prevent the displacement of the beating heart. After one day of culture, some epicardial cells had extended beyond the heart-gelatine interface. Subsequently, the heart was removed, leaving epicardial cells only. Then, the explants were starved overnight in a culture medium containing 1% FBS (starving medium). Finally, epicardial explants were incubated in the starving medium conditioned with 100 ng/mL WNT5A (Sigma, A2547).
EPICs are an in vitro model for epicardial-derived mesenchymal cells. EPICs were originally derived from E11.5 embryonic epicardium38. These cells were cultured under the same conditions previously described, including incubation in WNT5A conditioned medium. Cells were passaged using 0.05% trypsin-EDTA (Gibco, 25300-054) when they reached 70% confluence.
Proepicardial differentiation in cardiac gastruloids
R1 (R1 (R1/E #1036) (ECACC - cat. Number 07072001) mouse embryonic stem cells (mESC) were cultured at 37ºC and 5% CO2 in GMEM-BHK21 medium (Gibco, 21710-025) supplemented with 8% embryonic stem cell qualified FBS (Pansera, P30-2602), 1% non-essential amino acids (Gibco, 11140-035), 1% sodium pyruvate (Gibco, 11360-039), 1X penicillin/streptomycin (Gibco, 15070-063) and 0.1 mM β-mercaptoethanol (Sigma, M3148). This basal growth medium was completed with 3 µM CHIR99021 (Sigma SML1046), 1 µM PD0325901 (Sigma PZ0162) and 5 ng/mL LIF (Sigma LIF2050). Cells were cultured in 0.1% gelatin-coated labware. For passaging, cells were washed with DPBS (Ca-, Mg-, Gibco, 14190-094) and detached with 0.05% trypsin-EDTA. Cells were passaged no more than 10 times before the differentiation protocols were started.
Gastruloids were generated as previously described61,62. 400 R1 mESCs were plated in 40 µL N2B27 medium in 96-well Clear Round Bottom Ultra-Low Attachment Microplates (Corning, 0707). N2B27 medium included Neurobasal™ medium (Gibco, 21103049), DMEM/F12 (Gibco, 11320074), 0.5% B27 supplement (Gibco, 17504-044), 0.5% N2 supplement (Gibco, 17502-048), 1X GlutaMax™ (Gibco, 35050-038) and 0.1 mM β-mercaptoethanol (Sigma, M3148). Plates were centrifuged at 100g for 5 min and placed in a 5% CO2 incubator for 48 hours (day 2). Then, 150 µL of N2B27 containing 3 µM CHIR99021 (Sigma SML1046) was added to each well to induce gastrulation. At day 3 (72 h), the culture medium was replaced by N2B27 without CHIR90921. At day 4 (96 h), the medium was replaced by N2B27 supplemented with 30 ng/µL FGF2 (PMG0034 Gibco), 5 ng/µL VEGF (PHC9394 Gibco), and 0.5 mM ascorbic acid (Sigma, 255564) (Cardiogenic medium, N2B27+++). Gastruloids were then cultured at 37ºC and 5% CO2 on an orbital shaker (100 rpm). From day 5 (120 h) onward, the medium was changed daily with N2B27+++ medium until day 7 (included). After day 8, N2B27 medium was used and changed daily until day 10 of incubation.
Digestion of E9.5 cardiac inflow region and single-cell RNA sequencing
25 E9.5 embryos were harvested from 3 CD1 pregnant females. Once extracted, embryos were washed in cold HBSS (Ca2+-/Mg2+-, Sigma, 14170112) and membranes were excised using microdissection tools. Subsequently, embryos were transferred to a plate containing cold DMEM (Gibco, 11965) supplemented with 5% FBS. Using tungsten needles and microsurgery forceps, the posterior part of cardiac inflow was excised and placed in 1.5 mL LoBind® Eppendorf tubes (Eppendorf, 0030108051). Tissue explants were pooled and maintained in a small DMEM 5% FBS drop. Then, the tissue samples were washed in HBSS using the minimum centrifuge velocity (400g). Single-cell dissociation was carried out by incubating tissue explants in 60 µL 0.05% trypsin-EDTA at 37ºC for 6 min (gently rocking the tube by hand every 2 min). Tissue dissociation was blocked using HBSS-10% FBS. The cell suspension was transferred to a new LoBind® Eppendorf tube using a 40-µm cell strainer (pluriSelect, 43-10040-40). Cells were washed twice in HBSS-5% FBS and finally diluted in HBSS-5% FBS until 900 single-cells/µL was reached according to the user guide for Chromium Next GEM Single Cell 3’ Reagent Kits v3.1 (10X Genomics, CG000204 Rev D). Viability and cell number were estimated using trypan blue in both the Countess™ 3 Automated Cell Counter (Invitrogen) and the KOVA™ Glasstic™ slide system (Fisher Scientific, 87144). All the experimental procedures before library construction took less than 2 hours. scRNA-seq libraries were constructed from 10,000 individual cells using 10X Genomics Chromium Next GEM Single Cell 3’ kit v3.1 (PN-1000268) and the Chromium controller. Libraries were then sequenced on an Illumina NovaSeq 6000.
scRNA-seq analysis
Raw data were initially analysed using 10X Genomics Cell Ranger 6.0.1. First, output data was converted to fastq and aligned on the Mus musculus reference genome GRCm38/mm10. Finally, Cell Ranger count analysis provided various parameters such as estimated number of cells, mean reads per cell and median number of genes per cell. As a result, 3 files were created: one for each technical replicate and a third by merging the two technical replicates. Then, each dataset was analysed with the R package Seurat v4.3.0, which is the R toolkit for single-cell genomics63.
First, we evaluated the data quality through a linear correlation between the number of genes (features) and reads (counts) detected. After this analysis, we decided to keep cells with a range of 200 to 7,500 genes per cell, and genes detected in at least 10 cells. Then, we set cutoffs on cells with more than 5% mitochondrial gene expression, more than 10% haemoglobin genes and less than 10% of ribosomal genes. Cell doublets were predicted and removed using the R package DoubletFinder v2.0.364.
After completing the quality control steps, expression values across cells were normalised and scaled using the sctransform framework65. Then, principal component analysis (PCA) reduced the dimensionality of the data by identifying principal components that captured the most significant variation in gene expression. Subsequently, a graph-based clustering approach was applied to cells and non-linear dimensional reduction UMAP was set up for data visualisation. Differential expression analysis was also conducted applying the findMarkers function providing the gene markers that defined each cluster. The analysis was restricted to genes with an FDR < 0.05 and a minimum fold change of 0.2 in 75% of the clusters to recover genes that were markers shared by closely related cells. Cell identities were manually annotated using previous studies as references31–33.
The Harmony algorithm66 was used to integrate our scRNA-seq dataset of the posterior cardiac inflow with the published scRNA-seq dataset of E12.5 Wt1-lineage cells25. The RunHarmony function was used with a 2.5 value for the theta parameter and 30 dimensions of the Harmony reduction were used for calculating the UMAP.
Total RNA isolation and sequencing
E9.5 stage embryos from more than 3 CD1 mouse females were collected and washed in sterile PBS. Using tungsten needles and microsurgery forceps, two tissue samples were excised: the proepicardium (PE), corresponding to the ventral part of the STM, and part of the heart (HEART). Microdissected samples were pooled (n = 40) and put into TRIzol™ (Invitrogen, 15596026). RNA from three sample replicates were obtained. Total RNA was isolated using the phenol-chloroform method and stored at -80ºC. RNA integrity and concentration of all samples were measured using Agilent 2100 Bioanalyzer (Agilent Technologies). The RNA Integrity Number (RIN) for each sample was > 9, except for the first PE sample. Before RNA sequencing, gene expression of distinct marker genes for each anatomical region (Wt1 and Tbx18 for PE and Tnnt2 for HEART) was measured by qPCR (data not shown). The same RNA samples that were previously analysed by qPCR were submitted to sequencing. First, cDNA was prepared with at least 50 ng of total RNA using the standard Illumina TrueSeq RNASeq library preparation kit. Then, libraries were sequenced by Novogene in a NovaSeq 6000 system using the 150 bp paired-end sequencing strategy.
RNA-seq analysis
For quality control and preprocessing, SeqtrimBBsoftware was used with specific configurations67. The plugins used included PluginAdapters for adapter removal, PluginPolyAt for trimming poly-A tails, PluginContaminants with a ribosomal contaminants database and a minimum ratio threshold, PluginQuality for quality filtering, and PluginLowComplexity for removing reads with low complexity regions. The minimum read length was set to 40 bases with a minimum quality PHRED score of 20. GRCm39 was used as the reference genome for alignment with sequences from Mus musculus. Gene expression quantification was performed using the featureCounts software. The expression levels were normalised using the counts per million (CPM) method to account for library size differences between samples. Data analysis was performed with iDEP 1.1 (integrated Differential Expression and Pathway analysis)68. Briefly, after uploading the read count data, iDEP filtered out genes expressed at extremely low levels (in three samples, a gene must have more than 0.5 CPM) using the cpm function in edgRand conserved gene IDs according to Ensembl gene ID. Then, iDEP applied the regularised log transformation from the DESeq2package. Genes with an adjusted p-value (false discovery rate, FDR) less than 0.05 and a fold change greater than 2 were considered significantly, differentially expressed.
Immunohistochemistry
Tissues were fixed for 1–4 h in 4% PFA (depending on their size) and then washed three times in PBS. Samples were then incubated for 15–30 min in an ethanol series (50º, 70º, 80º, 90º, 96º and 100º) followed by a double 15-min incubation in butanol before paraffin embedding. Samples for cryo-sectioning were initially cryoprotected in 15% and 30% sucrose-in-PBS solutions. Then, samples were embedded in pure OCT medium (Tissue-Tek®, 4583) and flash-frozen in liquid N2-cooled 2-methylbutane (Sigma, 277258). Paraffin and cryosections of 8 µm were cut and mounted in poly-L-lysine- or AAS (3-aminopropyltriethoxysilane)-treated slides, respectively.
Paraffin-embedded samples were de-waxed in xylene and re-hydrated in an ethanol series (100º, 96º, 90º, 80º, 70º, 50º), finishing in water and 1X PBS incubation steps. Antigen retrieval was performed using a 0.1 M citrate buffer (0.0825M sodium citrate and 0.0175M citric acid (pH 6) in a pressure cooker for 10 min. Non-specific binding sites were blocked in SBT buffer for 1h at room-temperature (RT) (SBT: 10% goat serum, 1% bovine serum albumin, and 0.5% Triton X-100 in Tris-PBS). For frozen samples, sections were initially rehydrated in PBS, followed by a non-specific binding site blocking step in SBT buffer at RT for 1h in SBT. Fixed cell culture samples were directly immersed in SBT after washing in 1X PBS. Then, samples were incubated overnight (O/N) at 4ºC with primary antibodies diluted in SBT. Primary antibodies used were Wt1 (1:100; Abcam, ab89901), Troponin I (1:100; Santa Cruz, sc133117), GFP (1:500; Abcam, ab13970), PHH3 (1:100; Millipore, 06570), Sox9 (1:100; Millipore, ab5535), and Tbx5 (1:100; Santa Cruz, sc515536). After that, samples were washed in TBST (0.1% Triton X-100 in PBS) and incubated with the corresponding secondary antibodies (anti-chicken AF488, Jackson 703-545-155; anti-rabbit AF647, Jackson 715605-151; anti-mouse AF546, Invitrogen A11030; anti-mouse Cy5, Jackson 715-175-150). Nuclei were counterstained with DAPI (Sigma, D9542) and cell membranes were stained with wheat germ agglutinin (WGA). Samples were mounted with PBS:Glycerol (1:1). All samples were analysed using the Leica SP5 HyD Confocal Microscope and images were processed using ImageJ 1.6 software.
RNA in situ hybridisation (RNAScope®)
Embryos were harvested at different developmental stages, washed in 1X PBS and fixed in 4% PFA O/N at 4ºC. After that, samples were washed in PBS, dehydrated using 30-min ethanol series (50º, 70º, 80º, 90º and 96º), and incubated overnight in 100º ethanol. After incubation in xylene for 15 min samples were embedded in paraffin and stored at 4ºC. Gastruloids were collected, washed in 1X PBS, and fixed in 4% PFA O/N at 4ºC. After that, samples were washed with 1X PBS and embedded in heat-liquified HistoGel™ (Epredia, HG-4000-012). Solidified HistoGel blocks were post-fixed with 4% PFA for 1h at RT and embedded in paraffin as described above.
Tissue sections of 8 µm were cut and mounted in Superfrost® Plus slides (Epredia™J1800AMNZ, 10149870) following the protocol provided by the RNAscope™ Multiplex Fluorescent v2 kit (ACD, 323110). The HybEZ™ Hybridisation System (ACD, 310010) was used for hybridization and incubation steps. Briefly, tissue sections were pre-treated – including deparaffinising and rehydrating – and submitted to treatment with an antigen retrieval reactive and protease solution. Then, RNA-specific probes designed for different fluorescent detection channels were hybridised to RNA targets (ACD™ Bio Wt1, ref. 432711; Itga4, ref. 542901; Twist1, ref. 1266861; Lhx2, ref 485791; Tcf21, ref. 508668; Ror2, ref. 430041; Wnt5a, ref. 316791; Gfp, ref. 400281; Postn, ref. 418581). Different Opal™ fluorescent reagents were used to detect gene probes (Akoya Opal™ 520, ref. FP1487001KT; Opal™ 570, ref. FP1488001KT; Opal™ 570, ref. FP1496001KT). After three specific signal amplifications, single RNA transcripts in distinctly fluorescent channels were imaged using the SP5 Confocal Microscope (Leica).
qPCR
Total RNA was isolated using the phenol-chloroform method and measured in a NanoDrop™ 2000/2000c Spectrophotometer (Thermo Scientific). Samples with a 1.8-2 260/280 ratio were considered for the cDNA synthesis. Prior to cDNA preparation, genomic DNA was removed using DNase I (Invitrogen, 18068015). Then, 500 ng of total RNA was converted into cDNA using the AffinityScript Multiple Temperature cDNA Synthesis Kit (Agilent, 200436). qPCR was run in QuantStudio 5 Real-Time PCR System according to PowerUp SYBR Green instructions (Thermo Scientific, A25742). The forward and reverse primers were used at 0.8 µM and in each qPCR reaction the amount of cDNA equivalent to 2.5-5 ng RNA was used. The amplification protocol consisted of 5 min at 95ºC followed by 45 cycles of 10 sec at 95ºC, 20 sec at 60ºC, and 20 sec at 72ºC. qPCR primers were designed using Primer-BLAST (NCBI). The melting curve analysis and size fractionation by agarose gel electrophoresis were used to confirm amplification of the expected products69. Primers are listed in Supplementary Table S1. Relative gene expression (N0) was normalised with Tbp as a reference gene.
Cell proliferation analysis
Both EdU (5′-ethynyl-2′-deoxyuridine, Sigma, 900584) and BrdU (5-Bromo-2′-desoxiuridina, Sigma, B5002) were used to measure cell proliferation as they are nucleoside analogues of the thymidine that incorporate into the cell nuclei during the DNA synthesis phase of the cell division. Adult pregnant females were intraperitoneally injected with 50 mg EdU/kg body weight (diluted in 0.9% NaCl solution) 2h before embryo harvesting. Ex vivo cultured hearts were incubated with 10 µM EdU for 1h. Primary cultures of epicardial cells were incubated with 5 µg/mL BrdU for 1h. EdU detection on tissue sections was based on a Click-iT™ reaction between the EdU molecule and the fluorescent dye azide-fluoride 545 (Sigma, 760757). BrdU detection required a pre-treatment of 4% PFA-fixed cells (0.2N HCl incubation for 20 min followed by sodium tetraborate incubation for 10 min) and immunocytochemistry using a mouse anti-BrdU antibody (DSHB, G3G4) and a secondary anti-mouse antibody.
Cell cycle analysis by flow cytometry
Following ex vivo culture as described above, E11.5 hearts were collected and washed in PBS. Single-cell dissociation was carried out by incubating the hearts in 0.125 µg/mL Liberase TH (Roche, 5401135001) at 37ºC while rocking for a total of 10 min. Samples were filtered through 40-µm cell strainers and washed in cold PBS. Then, cells were fixed in cold 70% ethanol and stored at -20ºC for at least 24 h. Finally, samples were washed twice in cold PBS and incubated with Propidium Iodide (PI) solution (1 mg/mL PI, 0.1% Triton X-100, 200 µg/mL RNase I). The ratio between the number of cells and the volume of PI solution was maintained in all samples. Samples were analysed using the FACSCanto II flow cytometer (BD Biosciences). Downstream analysis of FCS files was performed using Kaluza v2.2 and the Michael H. Fox algorithm (included in the software) was used to analyse the cell cycle.
Protein extraction and quantification
EPIC cytoplasmic and nuclear protein fractions were isolated following the instructions in the Pierce™ NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo Scientific, 78833). Protein extracts were stored at -80ºC. The Pierce™ BCA Protein Assay Kit (Thermo Scientific, 23225) was used to determine the protein concentration. BSA and protein sample absorbance were measured at 595 nm in 96-well microplates (Deltalab, 900015) in the Biochrom Asys Expert Plus spectrophotometer (Biochrom Ltd, microplate reader G0 20150), and the protein concentration was determined using a linear regression from multiple BSA dilutions.
Western blot
A total of 30 µg of proteins was prepared in 1X Laemmli buffer (Thermo Scientific, J60015 AD), denatured at 95ºC for 5 min and separated in 12% Bis/Tris polyacrylamide gels. The proteins were transferred onto PVDF membranes (0.45 µm Immobilon-P®, Millipore, IPVH00010) using a Trans-Blot Turbo Transfer System (Bio-Rad). Then, membranes were blocked using 5% (m/v) non-fat dry milk diluted in TBS-T (10 mM Tris-HCl, 150 mM NaCl, 0.005% Tween-20) for 1h at RT, and incubated overnight with agitation at 4ºC (primary antibodies were prepared in blocking buffer). The proteins were incubated with rabbit anti-c-Jun (1;1000, Cell Signalling, 9165), mouse anti-Lamin B1 (2 ug/mL; Invitrogen, 33-2000) or rabbit anti-Gapdh (1:1000; Cell Signalling, 2118S). After three washes with TBS-T, HRP-conjugated anti-rabbit IgG (1:10.000; Sigma A0545) or anti-mouse IgG antibodies (1:10.000; Sigma A5278) were used in the following incubation step. Membranes were revealed using the SuperSignal™ West Pico PLUS (Thermo Scientific, 34580) or the SuperSignal™ West Femto kit (Thermo Scientific, 34095), according to the estimated levels of protein, and revealed in the ChemiDoc™ MP Imaging System (Bio-Rad).
Cell quantification
Itga4 + cells were quantified on the posterior part of the cardiac inflow from tissue sections from E9.5 Wt1-CreERT2;R26R-EYFP embryos (n = 6) in co-localisation with Wt1 and Yfp gene expression. In sections of E12.5 Wt1-CreERT2;R26R-EYFP embryos (n = 5), Itga4+/Yfp+ cells were quantified in the epicardium and subepicardium cardiac compartments using WGA staining to delimit cell membranes and basal lamina. In the in vivo proliferation analysis, Wt1+/Itga4+ (n = 9) and Itga4+/Ror2+ cells (n = 8) were quantified in the above-mentioned cell compartments in co-localisation with EdU staining. A minimum of 100 epicardial and subepicardial cells were analysed in each heart section. WT1+ and YFP+ cells were quantified in sections from wild-type and Wt1-CreERT2;R26R-EYFP E11.5 hearts cultured ex vivo for 24h in vehicle and nocodazole conditions (n = 5) using troponin I staining to delimit the epicardium-myocardium compartments and WGA to mark cell membranes. C-JUN+ cells were quantified in sections from E12.5 embryos (n = 6) in co-localisation with EdU and using troponin I staining to delimit the epicardium-myocardium boundaries. Between 30–100 epicardial and subepicardial cells were analysed in each heart. BrdU+ cells were quantified in primary culture of epicardial cells (n = 8). In all cases, Dapi was used to stain cell nuclei and quantification was performed using ImageJ software.
Statistical analysis
Data were expressed as mean ± SEM for bar graph data presented and statistically significant differences among samples were measured by unpaired T-test assuming both populations have the same SD. P values < 0.05 were considered statistically significant. Bar graph data analysis was performed using GraphPad Prism (version 8).
Reporting summary
Further information on research design is available in the Nature Research Reporting Summary linked to this article.