Skip to main content
Scientific Data logoLink to Scientific Data
. 2018 Aug 21;5:180170. doi: 10.1038/sdata.2018.170

Transcriptomic analyses of murine ventricular cardiomyocytes

Morgan Chevalier 1,*, Sarah H Vermij 1,*, Kurt Wyler 2, Ludovic Gillet 3,4, Irene Keller 5, Hugues Abriel 1,a
PMCID: PMC6103258  PMID: 30129933

Abstract

Mice are used universally as model organisms for studying heart physiology, and a plethora of genetically modified mouse models exist to study cardiac disease. Transcriptomic data for whole-heart tissue are available, but not yet for isolated ventricular cardiomyocytes. Our lab therefore collected comprehensive RNA-seq data from wildtype murine ventricular cardiomyocytes as well as from knockout models of the ion channel regulators CASK, dystrophin, and SAP97. We also elucidate ion channel expression from wild-type cells to help forward the debate about which ion channels are expressed in cardiomyocytes. Researchers studying the heart, and especially cardiac arrhythmias, may benefit from these cardiomyocyte-specific transcriptomic data to assess expression of genes of interest.

Subject terms: Gene expression, Mechanisms of disease

Background & Summary

In this study, we present next-generation RNA sequencing (RNA-seq) data of murine ventricular cardiomyocytes (CMC). To date, only whole-heart RNA-seq data have been published1–3, in which a variety of cell types, such as fibroblasts, endothelial cells, and atrial and ventricular cardiomyocytes, are pooled. We endeavoured to provide RNA-seq data of isolated CMCs for several reasons. Firstly, since the pump function of the heart relies on proper CMC function, CMCs are the most thoroughly studied cardiac cell type. Researchers studying CMCs may benefit from CMC-specific RNA-seq data from which expression of genes of interest can be extracted. Secondly, because of the crucial role of ion channels in cardiac electrical excitability and arrhythmogenesis, researchers that study cardiac arrhythmias have debated the question of which ion channels are expressed in CMCs. However, existing ion channel expression data are low-throughput, often contradictory4–6, fragmented7, or expression is assessed in the whole heart. The present work reveals the expression of the more than 350 ion channel family members, including pore-forming and auxiliary subunits, in CMCs (see Fig. 1 and Tables 1, Tables 2 and Tables 3 (available online only)). We therefore believe that these data will be valuable for ion channel researchers attempting to resolve the ongoing debate.

Figure 1. Gene expression of ion channels in murine ventricular cardiomyocytes.

Figure 1

(a) Expression levels of voltage-gated ion channel genes: voltage-gated sodium channels (Na+; purple), voltage-gated calcium channels (Ca2+; blue), transient receptor potential cation channels (TRP; light blue), CS, CatSper channels (aqua), two-pore channels (2P; green), cyclic-nucleotide-regulated channels (cN; light green), calcium-activated potassium channels (KCa; ochre), voltage-gated potassium channels (K+; orange), inwardly rectifying potassium channels (Kir; red) and two-pore potassium channels (2PK; burgundy). (b) Expression levels of the ligand-gated purinergic receptor gene (PR; purple) and of ion channel genes from the “other” category: aquaporins (Aqp; blue), voltage-sensitive chloride channels (Cl-; light blue), calcium-activated chloride channels (CaCl-; green) and inositol triphosphate receptors (IP3; light green). (c) Expression levels of more ion channel genes from the “other” category: ryanodine receptors (Ryr; orange), gap junction proteins (GJ; red) and chloride intracellular channels (icCl-; burgundy). All expression levels are average TPM values of WT samples (n=5). Shown are genes with more than 75 reads per gene (normalized for gene length, prior to conversion to TPM) from Tables 1, Table 2 and Table 3 (available online only).

Table 1. Expression of voltage-gated ion channels.

Gene Protein TPM
Voltage-gated ion channel genes, their respective proteins, and transcript per million (TPM) values averaged from five WT samples.    
Sodium channels, voltage-gated
   
 Scn5a Nav1.5 86.900
 Scn4a Nav1.4 12.163
 Scn7a Nav2.1 1.650
 Scn10a Nav1.8 0.180
 Scn3a Nav1.3 0.123
 Scn2a Nav1.2 0.025
 Scn11a Nav1.9 0.009
 Scn8a Nav1.6 0.004
 Scn1a Nav1.1 0.003
 Scn9a Nav1.7 0.000
 Scn4b β4 subunit 17.765
 Scn1b β1 subunit 6.524
 Scn3b β3 subunit 0.074
 Scn2b β2 subunit 0.041
Calcium channels voltage-gated
   
 Cacna1c Cav1.2 32.050
 Cacna1s Cav1.1 3.045
 Cacna1g Cav3.1 3.006
 Cacna1h Cav3.2 1.738
 Cacna1a Cav2.1 0.360
 Cacna1d Cav1.3 0.225
 Cacna1b Cav2.2 0.028
 Cacna1i Cav3.3 0.008
 Cacna1e Cav2.3 0.003
 Cacna1f Cav1.4 0.000
 Cacna2d1 α2 and δ1 subunit 30.799
 Cacna2d3 α2 and δ3 subunit 0.048
 Cacna2d4 α2 and δ4 subunit 0.000
 Cacnb2 β2 subunit 8.023
 Cacnb1 β1 subunit 1.714
 Cacnb3 β3 subunit 0.398
 Cacnb4 β4 subunit 0.108
 Cacng6 γ6 subunit 1.513
 Cacna2d2 γ1 subunit 0.455
 Cacng7 γ7 subunit 0.261
 Cacng1 γ7 subunit 0.024
 Cacng4 γ4 subunit 0.010
 Cacng5 γ5 subunit 0.008
 Cacng2 γ2 subunit 0.002
 Cacng3 γ3 subunit 0.000
 Cacng8 γ8 subunit 0.000
Transient receptor potential cation channels
   
 Trpm7 TRPM7 5.530
 Pkd1 TRPP1 5.248
 Pkd2l2 TRPP5 4.380
 Mcoln1 TRPML1 2.836
 Trpm4 TRPM4 2.431
 Pkd2 TRPP2 2.424
 Trpc1 TRPC1 1.836
 Trpc3 TRPC3 0.875
 Trpv2 TRPV2 0.391
 Trpv4 TRPV4 0.366
 Trpm3 TRPM3 0.287
 Trpc6 TRPC6 0.051
 Trpm6 TRPM6 0.037
 Trpc4 TRPC4 0.033
 Trpv1 TRPV1 0.026
 Trpa1 TRPA1 0.022
 Mcoln3 TRPML3 0.018
 Trpv3 TRPV3 0.016
 Mcoln2 TRPML2 0.011
 Trpm2 TRPM2 0.011
 Trpm5 TRPM5 0.005
 Trpv5 TRPV5 0.005
 Trpv6 TRPV6 0.005
 Trpc7 TRPC7 0.002
 Pkd2l1 TRPP3 0.000
 Trpc2 TRPC2 0.000
 Trpc5 TRPC5 0.000
 Trpm1 TRPM1 0.000
 Trpm8 TRPM8 0.000
CatSper channels
   
 Catsper2 CATSPER2 1.744
 Catsper3 CATSPER3, CACRC 0.127
 Catsper4 CATSPER4 0.060
 Catsperd δ subunit 0.058
 Catsperg1 γ1 subunit 0.031
 Catsperb β subunit 0.003
 Catsper1 CATSPER1 0.000
 Catsperg2 γ2 subunit 0.000
Two-pore channels
   
 Tpcn1 TPCN1 4.183
 Tpcn2 TPCN2 0.446
Cyclic nucleotide-regulated channels
   
 Hcn2 HCN2 9.736
 Hcn4 HCN4 3.661
 Cnga3 CNGA3 1.880
 Cngb3 CNGB3 1.051
 Hcn1 HCN1 0.229
 Cngb1 CNGB1 0.067
 Hcn3 HCN3 0.056
 Cnga1 CNGA1 0.000
 Cnga2 CNGA2 0.000
 Cnga4 CNGA4 0.000
Potassium channels, calcium-activated
   
 Kcnn1 KCa2.1 5.677
 Kcnn2 KCa2.2 3.107
 Kcnt2 Kna 0.287
 Kcnmb1 β1 subunit 0.127
 Kcnn3 KCa2.3 0.043
 Kcnt1 KCa4.1 0.019
 Kcnu1 KCa5.1 0.014
 Kcnn4 KCa3.1 0.008
 Kcnma1 KCa1.1 0.007
 Kcnmb2 β2 subunit 0.000
 Kcnmb3 β3 subunit 0.000
 Kcnmb4 β4 subunit 0.000
Potassium channels, voltage-gated
   
 Kcng2 Kv6.2 44.470
 Kcnh2 Kv11.1 17.558
 Kcnd2 Kv4.2 12.960
 Kcnq1 Kv7.1 12.079
 Kcnb1 Kv2.1 8.310
 Kcna5 Kv1.5 4.250
 Kcnv2 Kv8.2 3.502
 Kcnd3 Kv4.3 3.270
 Kcna7 Kv1.7 2.800
 Kcne1 KCNE1 1.917
 Kcnq4 Kv7.4 1.326
 Kcna4 Kv1.4 0.907
 Kcne4 KCNE4 0.833
 Kcnf1 Kv5.1 0.000
 Kcnc3 Kv3.3 0.566
 Kcnc1 Kv3.1 0.237
 Kcna1 Kv1.1 0.175
 Kcne2 KCNE2 0.005
 Kcna6 Kv1.6 0.119
 Kcnab3 KCAB3 0.113
 Kcna2 Kv1.2 0.105
 Kcnab2 KCAB2 0.088
 Kcnab1 KCAB1 0.056
 Kcns1 Kv9.1 0.051
 Kcnq5 Kv7.5 0.048
 Kcnc4 Kv3.4 0.041
 Kcnd1 Kv4.1 0.031
 Kcnc2 Kv3.2 0.029
 Kcnh1 Kv10.1 0.027
 Kcng4 Kv6.4 0.020
 Kcna3 Kv1.3 0.019
 Kcns3 Kv9.3 0.012
 Kcnh3 Kv12.2 0.008
 Kcnh6 Kv11.2 0.007
 Kcng3 Kv6.3 0.005
 Kcne3 KCNE3 0.060
 Kcnq3 Kv7.3 0.004
 Kcnb2 Kv2.2 0.003
 Kcnq2 Kv7.2 0.002
 Kcnh8 Kv12.1 0.002
 Kcna10 Kv1.8 0.000
 Kcng1 Kv6.1 0.000
 Kcnh4 Kv12.3 0.000
 Kcnh5 Kv10.2 0.000
 Kcnh7 Kv11.3 0.000
 Kcns2 Kv9.2 0.000
 Kcnv1 Kv8.1 0.000
Potassium channels, inwardly rectifying
   
 Abcc9 SUR2A,SUR2B 85.126
 Kcnj11 Kir6.2 51.960
 Kcnj3 Kir3.1 26.920
 Kcnj5 Kir3.4 24.795
 Kcnj2 Kir2.1 20.573
 Kcnj12 Kir2.2 12.050
 Kcnj8 Kir6.1 8.852
 Abcc8 SUR1 8.225
 Kcnj14 Kir2.4, Kir1.3 0.620
 Kcnj4 Kir2.3 0.322
 Kcnj15 Kir4.2 0.105
 Kcnj9 Kir3.3 0.016
 Kcnj1 Kir1.1 0.003
 Kcnj10 Kir4.1, Kir1.2 0.002
 Kcnj13 Kir7.1, Kir1.4 0.000
 Kcnj16 Kir5.1 0.000
 Kcnj6 Kir3.2 0.000
Potassium channels, two-P
   
 Kcnk3 K2P3.1, TASK-1 76.710
 Kcnk6 K2P6.1, TWIK-2 0.580
 Kcnk1 K2P1.1, TWIK-1 0.280
 Kcnk5 K2P5.1, TASK-2 0.104
 Kcnk2 K2P2.2, TREK-1 0.078
 zKcnk13 K2P13.1, THIK-1 0.066
 Kcnk7 K2P7.1 0.007
 Kcnk10 K2P10.1, TREK-2 0.006
 Kcnk12 K2P12.1, THIK-2 0.000
 Kcnk15 K2P15.1, TASK-5 0.000
 Kcnk16 K2P16.1, TASLK-1 0.000
 Kcnk18 K2P18.1, TRESK-2 0.000
 Kcnk4 K2P4.1, TRAAK 0.000
 Kcnk9 K2P9.1, TASK-3 0.000
Hydrogen voltage-gated ion channels
   
 Hvcn1 Hv1 0.138

Table 2. Expression of ligand-gated ion channels.

Gene Protein TPM
Ligand-gated ion channel genes, their respective proteins, and transcript per million (TPM) values averaged from five WT samples.    
5-HT (serotonin) receptors, ionotropic
   
 Htr3a 5-HT3A 0.023
 Htr3b 5-HT3B 0.000
Acetylcholine receptors, nicotinic
   
 Chrna2 ACHA2 1.073
 Chrnb1 ACHB 0.668
 Chrnb2 ACHB2 0.066
 Chrng ACHG 0.028
 Chrna10 ACH10 0.019
 Chrna1 ACHA 0.014
 Chrne ACHE 0.007
 Chrna5 ACHA5 0.003
 Chrna3 ACHA3 0.000
 Chrna4 ACHA4 0.000
 Chrna6 ACHA6 0.000
 Chrna7 ACHA7 0.000
 Chrna9 ACHA9 0.000
 Chrnb3 ACHB3 0.000
 Chrnb4 ACHB4 0.000
 Chrnd ACHD 0.000
GABA(A) receptors
   
 Gabrr2 GBRR2 0.855
 Gabra3 GBRA3 0.155
 Gabre GBRE 0.102
 Gabrb3 GBRB3 0.058
 Gabrq GBRT 0.022
 Gabrg3 GBRG3 0.021
 Gabrb2 GBRB2 0.019
 Gabra2 GBRA2 0.006
 Gabrd GBRD 0.004
 Gabra5 GBRA5 0.004
 Gabrg1 GBRG1 0.002
 Gabra4 GBRA4 0.002
 Gabra1 GBRA1 0.002
 Gabra6 GBRA6 0.000
 Gabrb1 GBRB1 0.000
 Gabrg2 GBRB2 0.000
 Gabrp GBRP 0.000
 Gabrr1 GBRR1 0.000
 Gabrr3 GBRR3 0.000
Glutamate receptors, ionotropic
   
 Grik5 GRIK5 0.830
 Grin2c NMDE3 0.407
 Grin3b NMD3B 0.151
 Gria3 GRIA3 0.148
 Grin2d NMDE4 0.106
 Gria1 GRIA1 0.022
 Grik4 GRIK4 0.018
 Grik3 GRIK3 0.016
 Grik2 GRIK2 0.010
 Grin3a NMD3A 0.008
 Grin2a NMDE1 0.006
 Gria4 GRIA4 0.005
 Grid2 GRID2 0.003
 Gria2 GRIA2 0.002
 Grid1 GRID1 0.001
 Grik1 GRIK1 0.001
 Grin1 NMDZ1 0.001
 Grin2b NMDE2 0.000
Glycine receptors
   
 Glra4 GLRA4 0.045
 Glra1 GLRA1 0.000
 Glra2 GLRA2 0.000
 Glra3 GLRA3 0.000
Purinergic receptors, ionotropic
   
 P2rx5 P2X5 8.390
 P2rx4 P2X4 2.570
 P2rx6 P2X6 1.069
 P2rx7 P2X7 0.349
 P2rx3 P2X3 0.191
 P2rx1 P2X1 0.065
 P2rx2 P2X2 0.010
Zinc-activated channels
   
not expressed in mice    

Table 3. Expression of other ion channels.

Gene Protein TPM
Other ion channel genes, their respective proteins, and transcript per million (TPM) values averaged from five WT samples.    
Acid-sensing (proton-gated) ion channels
   
 Asic3 ASIC3 0.068
 Asic1 ASIC1 0.053
 Asic4 ASIC4 0.009
 Asic2 ASIC2 0.000
Aquaporins
   
 Aqp1 AQP1 82.978
 Aqp7 AQP7 5.290
 Aqp8 AQP8 4.274
 Aqp4 AQP4 2.601
 Aqp11 AQP11 0.154
 Aqp6 AQP6 0.047
 Aqp2 AQP2 0.043
 Aqp5 AQP5 0.018
 Aqp9 AQP9 0.007
 Aqp12 AQP12 0.000
 Aqp3 AQP3 0.000
 Mip MIP 0.000
Chloride channels, voltage-sensitive
   
 Clcn4 CLCN4 15.442
 Clcn7 CLCN7 8.289
 Clcn3 CLCN3 5.871
 Clcn6 CLCN6 2.146
 Clcn1 CLCN1 1.737
 Clcn2 CLCN2 0.667
 Clcnkb CLCKB 0.550
 Clcn5 CLCN5 0.216
 Clcnka CLCKA 0.003
Cystic fibrosis transmembrane conductance regulators
   
 Cftr CFTR 0.006
Calcium-activated chloride channels
   
 Ano10 ANO10 12.374
 Ano8 ANO8 4.659
 Best3 BEST3 4.218
 Ano4 ANO4 1.961
 Ano1 ANO1 1.565
 Ano5 ANO5 1.432
 Ano6 ANO6 1.228
 Ano3 ANO3 0.033
 Best1 BEST1 0.016
 Ano9 ANO9 0.011
 Best2 BEST2 0.006
 Ano2 ANO2 0.000
 Ano7 ANO7 0.000
Chloride intracellular channels
   
 Clic4 CLIC4 72.409
 Clic5 CLIC5 65.201
 Clic1 CLIC1 10.695
 Clic3 CLIC3 0.398
 Clic6 CLIC6 0.039
Gap junction proteins
   
 Gja1 CXA1 82.934
 Gja3 CXA3 7.013
 Gjc1 CXG1 2.952
 Gja4 CXA4 1.946
 Gja5 CXA5 0.613
 Gja6 CXA6 0.147
 Gjc2 CXG2 0.095
 Gjd3 CXD3 0.069
 Gjb5 CXB5 0.019
 Gjc3 CXG3 0.015
 Gjb2 CXB2 0.005
 Gja10 CXA10 0.000
 Gja8 CXA8 0.000
 Gjb1 CXB1 0.000
 Gjb3 CXB3 0.000
 Gjb4 CXB4 0.000
 Gjb6 CXB6 0.000
 Gjd2 CXD2 0.000
 Gjd4 CXD4 0.000
 Gje1 GJE1 0.000
IP3 receptors
   
 Itpr1 ITPR1 4.382
 Itpr2 ITPR2 1.929
 Itpr3 ITPR3 0.798
Pannexins
   
 Panx2 PANX2 0.207
 Panx1 PANX1 0.137
 Panx3 PANX3 0.000
Ryanodine receptors
   
 Ryr2 RYR2 225.160
 Ryr3 RYR3 0.220
 Ryr1 RYR1 0.047
Sodium leak channels, non-selective
   
 Nalcn NALCN 0.049
Sodium channels, non-voltage-gated
   
 Scnn1a SCNNA 0.077
 Scnn1b SCNNB 0.000
 Scnn1g SCNNG 0.000

We have also included cardiac-specific knockout models of the ion channel regulators dystrophin, synapse-associated protein-97 (SAP97), and calmodulin-activated serine kinase (CASK). They interact with ion channels and modify their cell biological properties, such as membrane localization3,8–11. Notably, CASK provides a direct link between ion channel function and gene expression. It regulates transcription factors (TFs) in the nucleus, such as Tbr-1, and induces transcription of T-element-containing genes12. CASK also regulates TFs of the basic helix-loop-helix family, which bind E-box elements in promoter regions, by modulating the inhibitor of the DNA-binding-1 TF13. Additionally, CASK and SAP97 directly interact with each other11. For these reasons, we include CASK, SAP97, and dystrophin knockout mice to investigate whether these three proteins have a similar effect on gene expression, which may suggest their involvement in similar pathways. However, research beyond the scope of this paper would be needed to determine whether CASK-dependent TF regulation caused the differential expression that we observed.

To date, mutations in approximately 27 ion channel genes have been associated with cardiac arrhythmias, such as congenital short- and long-QT syndrome (SQTS and LQTS), Brugada syndrome (BrS), and conduction disorders (see http://omim.org)14–16. Notably, our ion channel expression data, as presented in Fig. 1 and Tables 1, Table 2 and Table 3 (available online only), reveal that several arrhythmia-associated ion channel genes are not or are scarcely expressed in murine ventricular CMCs (including Kcne2, Kcne3, Scn2b, and Scn3b). Although murine and human ion channel expression may differ, we are presently unaware of any available transcriptome of human CMCs17,18. We are also unable to either exclude or assess the effect of enzymatic isolation on the transcriptome. Finally, other cardiac cell types such as (myo)fibroblasts may express these ion channels and therefore may be important for arrhythmogenesis. Indeed, many ion channel genes that are not expressed in cardiomyocytes have been reported in murine whole-heart tissue2. These include Scn1a, Scn3b, 10 voltage-gated Ca2+ channels, 10 Kv channels, and four two-pore K+ channels. Conversely, all ion channel genes expressed in CMCs are also reported in whole-heart expression data.

In sum, this study presents RNA-seq data from wildtype murine ventricular CMCs, as well as from SAP97, CASK, and dystrophin knockouts and controls (see Fig. 2 for a schematic overview of study design). We performed differential gene expression analysis to compare the knockouts to their controls, and we extracted wildtype ion channel gene expression data (Tables 1, Table 2 and Table 3 (available online only), Fig. 1). We believe that these data will be valuable for researchers studying cardiomyocytes and ion channels to assess expression of genes of interest.

Figure 2. Experimental design and workflow.

Figure 2

(1) 22 mice with six different genetic backgrounds (CASK KO and control, SAP97 KO and control, and MDX and control) were used. fl+, first exon of gene is floxed; Cre+, Cre recombinase is expressed. (2) Cardiomyocytes were isolated on a Langendorff system and RNA was isolated with a FFPE Clear RNAready kit. (3) Libraries were constructed with 1 μg RNA per sample using a TrueSeq Stranded Total RNA protocol and (4) sequenced on an Illumina HiSeq3000 machine. (5) Quality of the reads was assessed with FastQC, and (6) reads were mapped to the Mus musculus reference genome (GRCm38.83) with Tophat. (7) To assess sample variation within each group, we performed principle component analyses (PCA) (see Fig. 3). (8) Lastly, ion channel expression was determined.

Methods

Mouse models

All animal experiments conformed to the Guide to the Care and Use of Laboratory Animals (US National Institutes of Health, publication No. 85-23, revised 1996); have been approved by the Cantonal Veterinary Administration, Bern, Switzerland; and have complied with the Swiss Federal Animal Protection Law. Mice were kept on a 12-hour light/dark cycle. Lights were on from 6:30 AM to 6:30 PM. To avoid the influence of circadian rhythm, mice were sacrificed between 10:00 AM and 1:00 PM. Mice were all male and were between the ages of 8 and 15 weeks.

MHC-Cre

The cardiac-specific murine alpha-myosin heavy chain (μMHC) promoter drives the expression of Cre recombinase, which, in turn, can recombine LoxP sequences. The μMHC-Cre strain was generated as previously described19 and acquired from the Jackson Laboratory (stock #011038).

CASK and SAP97 knockout mice

CASK KO and SAP97/Dlg1 KO mice were generated as previously described9,20. Both the CASK and SAP97 mouse lines were on mixed backgrounds. The appropriate control mice were selected in accordance with the publications that characterized both mouse lines9,20. CASK control mice express Cre while the first CASK exon is not floxed. SAP97 control mice are Cre-negative and the first SAP97 gene was floxed.

Dystrophin knockout (MDX-5CV) mice

The MDX-5CV strain demonstrates total deletion of the dystrophin protein. It was created as previously described21, and acquired from the Jackson laboratory (stock #002379). MDX mice were on pure Bl6/Ros backgrounds. Control mice were on pure Bl6/J background, except for MDX_Ct5 and MDX_5, which were Bl6/Ros mice backcrossed three times on Bl6/J.

Cardiomyocyte isolation

Mice (n=3–5 per genotype, male, age 10–15 weeks) were heparinized (intraperitoneal injection of 100 μL heparin (5000 U/mL; Biochrom AG)) and killed by cervical dislocation. Hearts were excised, and the aortas were cannulated in ice-cold phosphate-buffered saline (PBS). Subsequently, hearts were perfused on a Langendorff system in a retrograde manner at 37 °C with 5 mL perfusion buffer (1.5 mL/min; in mM: 135 NaCl, 4 KCl, 1.2 NaH2PO4, 1.2 MgCl2, 10 HEPES, 11 glucose), followed by the application of type II collagenase (Worthington CLS2; 25 mL of 1 mg/mL in perfusion buffer with 50 μM CaCl2). Left and right ventricles were triturated in PBS to dissociate individual ventricular cardiomyocytes and then filtered through a 100 μm filter.

RNA extraction and sequencing

RNA-seq was performed by the Next Generation Sequencing Platform at the University of Bern. Total RNA was isolated from freshly dissociated cardiomyocytes with an FFPE Clear RNAready kit (AmpTec, Germany), which included a DNase treatment step. RNA quality was assessed with Qubit and Bioanalyzer, and RNA quantity was checked with Qubit.

To allow sequencing of long non-coding RNA (lncRNA), libraries were constructed with 1 μg RNA using the TruSeq Stranded Total RNA kit after Ribo-Zero Gold (Illumina) treatment for rRNA depletion. Library molecules with inserts <300 base pairs (bp) were removed. Paired-end libraries (2x150 bp) were sequenced on an Illumina HiSeq3000 machine.

RNA-seq data analysis

Between 17.5 and 56.4 million read pairs were obtained per sample and the quality of the reads was assessed using FastQC v.0.11.2 (http://www.bioinformatics.babraham.ac.uk/projects/fastqc/). Ribosomal RNA (rRNA) was removed by mapping the reads with Bowtie2 v.2.2.1 (ref. 22) to a collection of rRNA sequences (NR_003279.1, NR_003278.3 and NR_003280.2) downloaded from NCBI (www.ncbi.nlm.nih.gov). No quality trimming was required.The remaining reads were mapped to the Mus musculus reference genome (GRCm38.83) with Tophat v.2.0.13 (ref. 23). We used htseq-count v.0.6.1 (ref. 24) to count the number of reads overlapping with each gene, as specified in the Ensembl annotation (GRCm38.83). Detailed information about the genes including the Entrez Gene ID, the MGI symbol and the description of the gene was obtained using the Bioconductor package BioMart v.2.26.1 (ref. 25).

Raw reads were corrected for gene length and TPM (transcripts per million) values were calculated to compare the expression levels among samples. Gene lengths for the latter step were retrieved from the Ensembl annotation (GRCm38.83) as the total sum of all exons.

Principal component analysis (PCA) plots were done in DESeq2 v.1.10.1 (ref. 26) (https://bioconductor.org/packages/release/bioc/html/DESeq2.html) using the 500 genes with the most variable expression across samples. A regularized log transformation was applied to the counts before performing the PCA.

Statistics

To assess differential gene expression between genotypes, a Wald test was performed with the Bioconductor package DESeq2 v.1.10.1 (ref. 26). We considered p values of up to 0.01, accounting for a Benjamini-Hochberg false discovery rate adjustment, to indicate significant difference. Statistical tools used included DESeq2, R-3.2.5 (https://cran.r-project.org), and Biomart_2.26.1 (www.biomart.org).

Data Records

The data were submitted to NCBI Gene Expression Omnibus (GEO) (Data Citation 1). This GEO project contains raw data and TPM values from all samples, and differential gene expression analysis between knockout and control samples.

Technical Validation

RNA metrics

RNA-seq yielded 1.0 billion read pairs in total, with an average of 44.5 million read pairs per sample (standard deviation 8.4 million). The number of read pairs (in millions) was 306 for CASK KO and Ctrl, 268 for SAP97 KO and Ctrl, and 404 for MDX and Ctrl (see Table 4 for an overview of RNA-seq metrics, including mapping rates). One sample (MDX_1) yielded few reads and was therefore excluded from further analyses. The proportion of reads mapping to annotated exons ranged from 65 to 77%. Mapping, no-feature (2–13%), and ambiguous (11–23%) read pairs together accounted for 89–97% of the total number of RNA reads (Table 4). Read pairs covered 49,671 genes of the Mus musculus reference genome (GRCm83.38).

Table 4. RNA-seq raw data and mapping metrics.

Sample ID Genotype # read pairs total # non-rRNA read pairs % of total Insert size # read pairs mapping to a gene % of total # no-feature read pairs % of total # ambiguous read pairs % of total
Total and non-ribosomal RNA read pairs, average RNA fragment size (bp), and mapping metrics, including absolute number and percentages of read pairs mapping to all annotated exons of the mouse reference genome, and no-feature and ambiguous reads, per sample. Note the low number of read pairs in MDX_1, which is therefore excluded from further analysis. CASK KO and Ctrl, SAP97 KO and Ctrl ns=3, MDX KO n=4, MDX Ctrl n=5.                      
CASK_Ct1 WT+Cre 47,543,799 47,343,548 99.58 492 34,076,980 71.67 2,721,942 5.73 8,287,647 17.43
CASK_Ct2 WT+Cre 45,437,500 45,287,356 99.67 476 33,988,592 74.8 1,440,229 3.17 7,578,641 16.68
CASK_Ct3 WT+Cre 55,117,414 54,944,721 99.69 479 40,469,641 73.42 1,790,829 3.25 11,381,005 20.65
CASK_KO1 CASK_fl+Cre 45,685,573 45,565,815 99.74 472 32,765,612 71.72 5,738,568 12.56 5,504,670 12.05
CASK_KO2 CASK_fl+Cre 55,895,769 55,607,105 99.48 511 39,344,558 70.39 2,372,238 4.24 12,476,403 22.32
CASK_KO3 CASK_fl+Cre 56,437,329 56,008,449 99.24 499 42,159,185 74.7 2,804,256 4.97 9,655,232 17.11
MDX_1 MDX 17,485,935 17,320,513 99.05 380 (sample exluded)          
MDX_2 MDX 39,536,744 39,037,330 98.74 447 27,475,113 69.49 3,258,092 8.24 6,543,268 16.55
MDX_3 MDX 39,626,959 39,432,254 99.51 455 27,841,146 70.26 2,169,924 5.48 7,327,584 18.49
MDX_4 MDX 42,406,246 40,919,896 96.49 488 29,805,158 70.28 2,905,415 6.85 6,497,990 15.32
MDX_5 MDX 50,934,677 47,518,076 93.29 484 34,233,480 67.21 1,864,210 3.66 10,028,518 19.69
MDX_Ct1 WT 48,311,563 46,288,106 95.81 380 32,827,800 67.95 4,353,181 9.01 7,779,264 16.1
MDX_Ct2 WT 47,283,192 46,988,962 99.38 446 32,237,279 68.18 2,304,142 4.87 10,939,883 23.14
MDX_Ct3 WT 35,275,617 34,938,284 99.04 427 24,235,276 68.7 3,631,537 10.29 4,922,208 13.95
MDX_Ct4 WT 33,977,175 32,900,815 96.83 515 25,298,933 74.46 1,713,065 5.04 4,619,558 13.6
MDX_Ct5 WT 49,379,536 45,499,492 92.14 485 32,227,570 65.27 1,210,708 2.45 10,698,976 21.67
SAP_Ct1 WT+Cre 47,930,112 47,715,719 99.55 461 34,192,649 71.34 1,965,652 4.1 9,896,590 20.65
SAP_Ct2 WT+Cre 44,934,245 44,566,395 99.18 444 30,350,071 67.54 4,879,732 10.86 7,491,483 16.67
SAP_Ct3 WT+Cre 43,586,968 43,382,766 99.53 451 29,836,839 68.45 1,332,267 3.06 8,751,881 20.08
SAP_KO1 SAP_fl+Cre 44,319,566 44,146,526 99.61 452 34,155,235 77.07 2,606,959 5.88 5,090,692 11.49
SAP_KO2 SAP_fl+Cre 41,547,517 41,397,099 99.64 469 28,697,320 69.07 3,765,768 9.06 7,431,842 17.89
SAP_KO3 SAP_fl+Cre 46,143,349 45,812,985 99.28 443 30,710,635 66.55 5,476,238 11.87 8,174,538 17.72

Quality assessment

The quality of all samples was assessed with FastQC. Except for MDX_1, all samples were of high quality. Where applicable, a representative example (MDX_Ct1) is shown. Firstly, the insert size histogram (Fig. 3a) shows that the inferred insert size of each sample exceeded 150 base pairs, demonstating that the sequencing was not contaminated by adapter sequences. Secondly, the GC content plot (Fig. 3c) ideally shows a roughly normal distribution centred around the average GC content of the genome, which varies between species. The peaks observed in Fig. 3c are likely caused by sequences that are detected at high copy numbers, and should not pose problems for downstream analyses. Furthermore, Phred scores (Fig. 3d) are well within the green area of the graph indicating good base quality along the length of reads. As well, the gene coverage graph (Fig. 3e) of sample MDX_Ct1 shows that reads are distributed evenly along the length of the gene body. Because the gene coverage for all other samples is highly comparable to that of MDX_Ct1, only one example is shown. Lastly, the saturation report (Fig. 3f) represents the number of splice junctions detected using different subsets of the data from 5 to 100% of all reads. At sequencing depths sufficient to perform alternative splicing analysis, at least the red line, representing known junctions, should reach a plateau where adding more data does not much increase the number of detected junctions. Only MDX_1 does not reach this plateau.

Figure 3. Quality control.

Figure 3

(a) Histogram of inferred insert size for each sample, which represents distance between the two reads of one RNA fragment. (b) Principle component analyses (PCA) plots were performed to assess variability of samples within and between groups. Plot of the first two axes from a PCA based on the 500 genes with the most variable expression across all samples except MDX_1. CASK control (red, n=3) and KO (green, n=3); MDX control (orange, n=5) and KO (blue, n=4); SAP97 control (grey, n=3) and KO (black, n=3). (c) Distribution of GC content of the reads for each sample. (d) Base quality (Phred scores) along the length of the reads in each FastQC file of MDX_Ct1 as representative sample. The box plots are drawn as follows: red line, median; yellow box, range between upper and lower quartiles; whiskers, range between 10 and 90% quantiles. The blue line shows the mean quality. Y-axis represents quality scores across all bases. X-axis represents position in read (bp). (e) Gene body coverage. Distribution of reads along the length of the genes (5’-end on the left, 3’-end on the right). Shown image of sample MDX_Ct1 is representative for all samples. (f) Saturation report, depicting the number of splice junctions detected using different subsets of the data from 5 to 100% of all reads. Red, known junction based on the provided genome annotation; green, novel junctions; blue, all junctions. The red line reaches a plateau where adding more data does not increase the number of detected junctions, indicating that the sequencing depth suffices for performing alternative splicing analysis.

Gene expression variation of biological replicates

We performed Principle Component Analyses (PCA) to assess whether samples from the same experimental group have similar gene expression profiles (Fig. 3b). Of note, samples within each sample group still show considerable variation. The mixed genetic background of most sample groups may explain this variation; only the MDX control mice are on a pure Bl6/J background. The variation seen in MDX control mice is likely due to a batch effect, as two rounds of samples were sequenced. However, considering that PCA plots are based on the 500 genes with the highest variability in one sample, our genes of interest, including all ion channel genes, show similar expression levels throughout all samples.

Ion channel expression

Based on the list of ion channel genes from HUGO Gene Nomenclature Committee (https://www.genenames.org/cgi-bin/genefamilies/set/177), we distilled ion channel expression from WT mice expressed as TPM (Tables 1, Table 2 and Table 3 (available online only), Fig. 1).

Additional information

How to cite this article: Chevalier, M. et al, Transcriptomic analyses of murine ventricular cardiomyocytes. Sci. Data 5:180170 doi: 10.1038/sdata.2018.170 (2018).

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Material

sdata2018170-isa1.zip (3.7KB, zip)

Acknowledgments

The research was financially supported by Swiss National Science Foundation Grant 310030_165741 (HA). We thank the Next Generation Sequencing Platform of the University of Bern, in particular Muriel Fragnière, for performing the high-throughput sequencing experiments; prof. Rolf Jaggi for excellent technical advice on RNA isolation from cardiomyocytes; and Joseph Allan for language editing.

Footnotes

The authors declare no competing interests.

Data Citations

  1. 2017. Gene Expression Omnibus. GSE102772

References

  1. Cerrone M. et al. Plakophilin-2 is required for transcription of genes that control calcium cycling and cardiac rhythm. Nat Commun 8, 106 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Harrell M. D., Harbi S., Hoffman J. F., Zavadil J. & Coetzee W. A. Large-scale analysis of ion channel gene expression in the mouse heart during perinatal development. Physiol. Genomics 28, 273–283 (2007). [DOI] [PubMed] [Google Scholar]
  3. Petitprez S. et al. SAP97 and dystrophin macromolecular complexes determine two pools of cardiac sodium channels Nav1.5 in cardiomyocytes. Circ. Res. 108, 294–304 (2011). [DOI] [PubMed] [Google Scholar]
  4. Zimmer T., Haufe V. & Blechschmidt S. Voltage-gated sodium channels in the mammalian heart. Glob. Cardiol. Sci. Pract 4, 449–463 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Balse E. et al. Dynamic of ion channel expression at the plasma membrane of cardiomyocytes. Physiol. Rev. 92, 1317–1358 (2012). [DOI] [PubMed] [Google Scholar]
  6. Westenbroek R. E. et al. Localization of sodium channel subtypes in mouse ventricular myocytes using quantitative immunocytochemistry. J. Mol. Cell. Cardiol. 64, 69–78 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Stroud D. M. et al. Contrasting Nav1.8 Activity in Scn10a-/- Ventricular Myocytes and the Intact Heart. J. Am. Heart Assoc 5, 1–10 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Leonoudakis D., Conti L. R., Radeke C. M., McGuire L. M. & Vandenberg C. A. A multiprotein trafficking complex composed of SAP97, CASK, Veli, and Mint1 is associated with inward rectifier Kir2 potassium channels. J. Biol. Chem. 279, 19051–19063 (2004). [DOI] [PubMed] [Google Scholar]
  9. Eichel C. A. et al. Lateral membrane-specific MAGUK CASK down-regulates NaV1.5 channel in cardiac myocytes. Circ. Res. 119, 544–556 (2016). [DOI] [PubMed] [Google Scholar]
  10. Abriel H., Rougier J. S. & Jalife J. Ion channel macromolecular complexes in cardiomyocytes: roles in sudden cardiac death. Circ. Res. 116, 1971–1988 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Nafzger S. & Rougier J. S. Calcium/calmodulin-dependent serine protein kinase CASK modulates the L-type calcium current. Cell Calcium 61, 10–21 (2017). [DOI] [PubMed] [Google Scholar]
  12. Hsueh Y. P., Wang T. F., Yang F. C. & Sheng M. Nuclear translocation and transcription regulation by the membrane-associated guanylate kinase CASK/LIN-2. Nature 404, 298–302 (2000). [DOI] [PubMed] [Google Scholar]
  13. Sun R. et al. Human calcium/calmodulin-dependent serine protein kinase regulates the expression of p21 via the E2A transcription factor. Biochem. J. 419, 457–466 (2009). [DOI] [PubMed] [Google Scholar]
  14. Ashcroft F. M. From molecule to malady. Nature 440, 440–447 (2006). [DOI] [PubMed] [Google Scholar]
  15. Rickert-Sperling S., Kelly R. G. & Driscoll D. J. Congenital Heart Diseases: The Broken Heart 721–736 (Springer-Verlag, 2016). [Google Scholar]
  16. Lahrouchi N., Behr E. R. & Bezzina C. R. Next-generation sequencing in post-mortem genetic testing of young sudden cardiac death cases. Front. Cardiovasc. Med 3, 13 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Heinig M. A. et al. Natural genetic variation of the cardiac transcriptome in non-diseased donors and patients with dilated cardiomyopathy. Genome Biol. 18, 170 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. van den Berg C. W. et al. Transcriptome of human foetal heart compared with cardiomyocytes from pluripotent stem cells. Development 142, 3231–3238 (2015). [DOI] [PubMed] [Google Scholar]
  19. Agah R. et al. Gene recombination in postmitotic cells. Targeted expression of Cre recombinase provokes cardiac-restricted, site-specific rearrangement in adult ventricular muscle in vivo. J. Clin. Invest. 100, 169–179 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Gillet L. et al. Cardiac-specific ablation of synapse-associated protein SAP97 in mice decreases potassium currents but not sodium current. Heart Rhythm 12, 181–192 (2015). [DOI] [PubMed] [Google Scholar]
  21. Chapman V. M., Miller D. R., Armstrong D. & Caskey C. T. Recovery of induced mutations for X chromosome-linked muscular dystrophy in mice. Proc. Natl. Acad. Sci. USA 86, 1292–1296 (1989). [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Langmead B. & Salzberg S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Kim D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Anders S., Pyl P. T. & Huber W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Durinck S. et al. BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis. Bioinformatics 21, 3439–3440 (2005). [DOI] [PubMed] [Google Scholar]
  26. Love M. I., Huber W. & Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Citations

  1. 2017. Gene Expression Omnibus. GSE102772

Supplementary Materials

sdata2018170-isa1.zip (3.7KB, zip)

Articles from Scientific Data are provided here courtesy of Nature Publishing Group

RESOURCES