Open Access

Identification of key genes associated with congenital heart defects in embryos of diabetic mice

  • Authors:
    • Nan Lin
    • Yan Cai
    • Linlin Zhang
    • Yahang Chen
  • View Affiliations

  • Published online on: December 20, 2017     https://doi.org/10.3892/mmr.2017.8330
  • Pages: 3697-3707
  • Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Maternal diabetes has been reported to be a critical factor for congenital heart defects (CHD) in offspring. The present study aimed to screen the key genes that may be involved in CHD in offspring of diabetic mothers. The present study obtained the gene expression profile of GSE32078, including three embryonic heart tissue samples at embryonic day 13.5 (E13.5), three embryonic heart tissue samples at embryonic day 15.5 (E15.5) from diabetic mice and their respective controls from normal mice. The cut‑off criterion of P<0.08 was set to screen differentially expressed genes (DEGs). Their enrichment functions were predicted by Gene Ontology. The enriched pathways were forecasted by Kyoto Encyclopedia of Genes and Genomes and Reactome analysis. Protein‑protein interaction (PPI) networks for DEGs were constructed using Cytoscape. The present study identified 869 and 802 DEGs in E13.5 group and E15.5 group, respectively and 182 DEGs were shared by the two developmental stages. The pathway enrichment analysis results revealed that DEGs including intercellular adhesion molecule 1 (Icam1) and H2‑M9 were enriched in cell adhesion molecules; DEGs including bone morphogenetic protein receptor type 1A, transforming growth factor β receptor 1 and SMAD specific E3 ubiquitin protein ligase 1 were enriched in the tumor growth factor‑β signaling pathway. In addition, DEGs including Icam1, C1s and Fc fragment of IgG receptor IIb were enriched in Staphylococcus aureus infection. Furthermore, the shared DEGs including Icam1, nuclear receptor corepressor 1 (Ncor1) and AKT serine/threonine kinase 3 (Akt3) had high connectivity degrees in the PPI network. The shared DEGs including Icam1, Ncor1 and Akt3 may be important in the cardiogenesis of embryos. These genes may be involved in the development of CHD in the offspring of diabetic mothers.

Introduction

Diabetes mellitus is a chronic disease characterized by hyperglycemia resulting from defects in insulin secretion, insulin action, or both (1). According to insulin-dependent and non-insulin-dependent, adult diabetes is classified: Type 1 and type 2 diabetes. A genetic defect on insulin was found in type 1 diabetes, whereas insulin resistance is the key metabolic abnormality in type 2 diabetes (2). In addition, maternal diabetes in pregnancy is defined as glucose intolerance or first recognition during pregnancy, who is easy to happen type 2 diabetes postpartum (3). Besides, maternal diabetes in pregnancy is related to an increased risk of congenital heart defects (CHD), macrosomia, miscarriage, and other birth defects in offspring (4,5). CHD was defined as deficits of the structure and function arising from cardiac embryogenesis stage (6). It has been reported that CHD is the major consequences in diabetic embryopathy (7).

An infant born to a diabetic mother has been shown to exhibit axial mesodermal dysplasia spectrum with atrioventricular septal defects (8). Many studies have reported that maternal diabetes altered expression of genes in developmental embryo (4,9,10). Bmp4, belongs to the TGF-β superfamily, is a myocardial signaling molecule which activated epithelial-mesenchymal transition (EMT) during cardiogenesis (11). The expression of Bmp4 has been reported to be downregulated by the Msx1 that expressed in atrioventricular canal endocardial cells during EMT (12,13). Pax3 is also essential for heart formation and outflow tract development in the mouse embryo (14). Study showed that the downregulation of Bmp4, Msx1 and Pax3 could contribute to the pathogenesis of maternal diabetes-induced CHD (15). In addition, it has been reported that hyperglycemia altered the expression of eNOC and VEGF that are involved in the regulation of vasculogenesis (16). Furthermore, a recent study on microarray analysis showed that several genes were altered in embryonic heart tissues from diabetic mother and were closely associated with CHD, such as Smyd1, Tsc1 and Gja1 (4). However, the exact pathogenesis of CHD in offspring of diabetic mother is still unknown.

In this study, we explored the gene expression profiles of embryonic heart tissue samples at embryonic day 13.5 (E13.5) and embryonic day 15.5 (E15.5) from diabetic mice, and their respective controls. The differentially expressed genes (DEGs) were screened in E13.5 and E15.5 groups. Gene Ontology (GO) functions, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathway enrichment analyses were then performed to identify the DEGs. We also constructed the protein-protein interaction (PPI) networks for the DEGs and analyzed several important shared genes that were associated with CHD. Our study aimed to identify the critical genes that might be involved in CHD in offspring of diabetic mother and to provide evidence to further clarify the relationship between CHD and diabetes.

Materials and methods

Affymetrix microarray data

The gene expression profile of GSE32078 was obtained from the study of Vijaya et al (4), which was deposited in Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/) of National Center of Biotechnology Information (NCBI). A total of 12 samples were available, including three embryonic heart tissue samples at E13.5 and three embryonic heart tissue samples at E15.5 from streptozotocin-induced diabetic Swiss albino mice (8–10 weeks), as well as their respective controls from normal mice. The day when a copulation plug was observed was counted as E0.5. Raw data were collected with the Affymetrix Mouse Genome 430 2.0 Array (Affymetrix, CA, USA). All raw data files were pretreated by RMA method in Affy package (17).

Screening of DEGs

The Linear Models for Microarray Data (LIMMA) package (18) was used to identify DEGs. All genes were tested with F-test and the DEGs with the threshold P-value <0.08 were screened.

GO and KEGG pathway enrichment analysis

ClusterProfiler, a new ontology-based tool, offers three methods (group GO, enrich GO, enrich KEGG) for genes classification and enrichment analyses (19,20). ClusterProfiler package was used to identify the main biological processes and metabolic pathways in DEGs. Default parameters (organism, mouse; ont, BP; P-value cut-off, 0.05; P-adjust method, none; readable, T) were used as the cut-off criteria for GO function enrichment analysis. And the default parameters (organism, mouse; P-value cut-off, 1; q-value cut-off, 1; readable, 1) were used as the cut-off criteria for KEGG pathway enrichment analysis.

Reactome pathway enrichment analysis

ReactomePA package was used to identify the main biological processes and metabolic pathways in DEGs (21,22). Default parameters (organism, mouse; P-value cut-off, 1; q-value cut-off, 1; minGSSize, 1; readable, 1) were used as the cut-off criteria for Reactome pathway enrichment analysis.

PPI network construction

The Search Tool for the Retrieval of Interacting Genes (STRING) database was used to obtain PPI data. The PPI networks of up- and downregulated DEGs were visualized by Cytoscape (http://cytoscape.org/) that is an open source software for visualizing complex networks and integrating these networks with any type of attribute data (23). The Required Confidence score >0.4 was chosen as the threshold.

Results

Screening of DEGs

The microarray dataset GSE32078 from GEO database was obtained to identify the DEGs in E13.5 and E15.5 hearts of embryos from diabetic mice compared with their respective controls. Totally, 869 DEGs were obtained in E13.5 group when compared with the control group, including 411 up- and 458 downregulated DEGs. Meanwhile, 802 genes were upregulated and 1,295 genes were downregulated in E15.5 group when compared with the control group. Finally, a total of 182 DEGs were shared between E13.5 and E15.5 groups, including 63 up- and 119 downregulated DEGs.

GO function enrichment analysis of DEGs

According to the GO enrichment analysis, we found that the upregulated DEGs in E13.5 group were mainly enriched in developmental process (P=2.32E-08) and the downregulated DEGs were mainly enriched in organic substance metabolic process (P=2.93E-06). In the E15.5 group, the upregulated DEGs were mainly enriched in cellular process (P=2.64E-19) and the downregulated DEGs were mainly enriched in cellular process (P=2.01E-19). Besides, we also performed the GO function enrichment analysis for the genes that were shared by E13.5 group and E15.5 group. The results showed that the upregulated DEGs were mainly related to cellular process (P=0.00650) and the downregulated DEGs were mainly related to organic substance metabolic process (P=0.00031) (Fig. 1).

KEGG and Reactome pathway analysis of DEGs

KEGG and Reactome enrichment pathway analysis were performed to explore the enriched pathways of the DEGs. The KEGG enrichment analysis results showed that the upregulated DEGs in E13.5 group were enriched in 64 pathways, such as Icam1, H2-M9, and 4930468A15Rik were enriched in cell adhesion molecules (CAMs) (P=1.56E-03). The downregulated DEGs in E13.5 group were enriched in 84 pathways, such as Bmpr1a, Tgfbr1 and Smurf1 were enriched in TGF-β signaling pathway (P=4.03E-03). The upregulated DEGs in E15.5 group were enriched in 129 pathways, such as Icam1, C1s and Fcgr2b were enriched in Staphylococcus aureus infection (P=1.80E-04). Meanwhile, the downregulated DEGs in E15.5 group were enriched in 156 pathways, such as Ero1lb, Dnajc3 and Dnajb11 were enriched in protein processing in endoplasmic reticulum (P=1.27E-03). Moreover, the shared DEGs that were upregulated such as Cmah and Gmppa were enriched in nucleotide sugar metabolism (P=2.65E-03). And the shared DEGs that were downregulated such as Akt3, Pak3 and Hgf were enriched in renal cell carcinoma (P=5.00E-04); Akt3, Pak3 and Pak1 were enriched in T cell receptor signaling pathway (P=2.60E-03) (Table I). Reactome enrichment analysis results showed that the upregulated DEGs in E13.5 group were enriched in 154 pathways and the downregulated DEGs in E13.5 group were enriched in 192 pathways. The upregulated DEGs in E15.5 group were enriched in 27 pathways and the downregulated DEGs in E15.5 group were enriched in 24 pathways. In addition, the shared DEGs that were upregulated such as Nr6a1 and Esrrg were enriched in nuclear receptor transcription pathway (P=5.09E-03) and the shared DEGs that were downregulated such as Pak3 and Pak1 were enriched in activation of Rac (P=1.85E-03) (Table II).

Table I.

Enrichment analysis of top five KEGG pathways for DEGs.

Table I.

Enrichment analysis of top five KEGG pathways for DEGs.

DEGsTermDescriptionP-valueGene IDCount
Upregulated DEGs in E13.5mmu04514Cell adhesion molecules (CAMs) 1.56×10−03Icam1, H2-M9, 4930468A15Rik, H2-D1, Itga4, H2-Ob, Cntnap2, Nrxn2  8
mmu04672Intestinal immune network for IgA production 3.77×10−03Gm13306, Itga4, Il15, H2-Ob  4
mmu05330Allograft rejection 8.24×10−03H2-M9, H2-D1, H2-Ob, Il12b  4
mmu04940Type I diabetes mellitus 1.24×10−02H2-M9, H2-D1, H2-Ob, Il12b  4
mmu04146Peroxisome 2.74×10−02Agps, Nudt19, Slc25a17, Acsbg2  4
Downregulated DEGs in E13.5mmu04350TGF-β signaling pathway 4.03×10−03Bmpr1a, Tgfbr1, Smurf1, Ltbp1 Lefty2, Thbs2,  6
mmu05211Renal cell carcinoma 9.14×10−03Akt3, Pak3, Hgf, Pak1, Pik3ca  5
mmu04080Neuroactive ligand-receptor interaction 1.05×10−02Prlr, Gabra1, Tshr, Gabra2, Gabrb2, Crhr211
mmu05223Non-small cell lung cancer 1.66×10−02Akt3, Pik3ca, Stk4, Rarb  4
mmu04210Apoptosis 1.87×10−02Irak3, Il3, Akt3, Endod1, Pik3ca  5
Upregulated DEGs in E15.5mmu05150Staphylococcus aureus infection 1.80×10−04Icam1, C1s, Fcgr2b, H2-Ab1, C2, Fpr2, Cfh, H2-Aa  8
mmu04514Cell adhesion molecules (CAMs) 1.18×10−03Icam1, Vcan, H2-K1, Cd80, Ncam113
mmu04145Phagosome 1.61×10−03Lamp2, H2-K1, Tap2, Tuba4a, Thbs2, Fcgr2b14
mmu05219Bladder cancer 2.20×10−03Rb1, Mmp9, Myc, E2f3, Egfr, E2f2  6
mmu05220Chronic myeloid leukemia 2.27×10−03Bcl2l1, Rb1, Myc, Cblb, E2f3, Sos2, E2f2, Abl1  8
Downregulated DEGs in E15.5mmu04141Protein processing in endoplasmic reticulum 1.27×10−03Ero1lb, Dnajc3, Dnajb11, Sec62, Derl1, Mapk818
mmu01100Metabolic pathways 2.25×10−03Phgdh, B3gnt5, Sucla2, Alad, B4galt6, Mmab77
mmu04977Vitamin digestion and absorption 4.84×10−03Tcn2, Rbp2, Plb1, Lrat, Slc5a6  5
mmu04010MAPK signaling pathway 9.22×10−03Cacna1a, Ptpn5, Mapk8, Cacna2d1, Akt3, Gna1222
mmu00512Mucin type O-Glycan biosynthesis 9.57×10−03Gcnt3, C1galt1, Galnt2, Galnt7, Galnt15  5
Upregulated genes in shared DEGsmmu00520Amino sugar and nucleotide sugar metabolism 2.65×10−03Cmah, Gmppa  2
mmu01100Metabolic pathways 6.08×10−01Lias, Gmppa  2
Downregulatedmmu05211Renal cell carcinoma 5.00×10−04Akt3, Pak3, Hgf, Pak1,  4
genes in shared DEGsmmu04660T cell receptor signaling pathway 2.60×10−03Akt3, Pak3, Pak1, Grap2  4
mmu04730Long-term depression 6.09×10−03Cacna1a, Gna12, Ppp1r17  3
mmu04210Apoptosis 9.95×10−03Akt3, Il3, Endod1  3
mmu04012ErbB signaling pathway 1.03×10−02Akt3, Pak3, Pak1  3
mmu04510Focal adhesion 1.96×10−02Akt3, Pak3, Hgf, Pak1,  4
mmu04010MAPK signaling pathway 5.02×10−02Akt3, Cacna1a, Gna12, Pak1  4
mmu04080Neuroactive ligand-receptor interaction 5.48×10−02Prlr, Tshr, Gabrb2, Crhr2  4
mmu04630Jak-STAT signaling pathway 4.49×10−02Akt3, Prlr, Il3  3
mmu00140Steroid hormone biosynthesis 3.31×10−02Hsd17b7, Hsd11b2  2

[i] E13.5, embryonic day 13.5; E15.5, embryonic day 15.5; DEGs, differentially expressed genes; KEGG, Kyoto Encyclopedia of Genes and Genomes; DEGs, differentially expressed genes.

Table II.

Enrichment analysis of top five Reactome pathways for DEGs.

Table II.

Enrichment analysis of top five Reactome pathways for DEGs.

DEGsTermDescriptionP-valueGene IDCount
Upregulated DEGs in E13.54809882Neuronal system 2.10×10−03Gjd2, Braf, Kcnb1, Chrna3, Gabrg2, Gls10
4810204Neurotransmitter receptor binding and downstream transmission in the post-synaptic cell 2.14×10−03Braf, Chrna3, Gabrg2, Arhgef9, Ap2a2, Epb4.1l1, Rps6ka2  7
4810917Interferon γ signaling 2.64×10−03Icam1, Oas1h, Irf7, Sp100, H2-D1  5
4809881Transmission across chemical synapses 3.10×10−03Braf, Chrna3, Gabrg2, Gls, Arhgef9, Ap2a2, Epb4.1l1, Rps6ka2  8
4810848Regulation of cytoskeletal remodeling and cell spreading by IPP complex components 5.26×10−03Parvb, Pxn  2
Downregulated DEGs in E13.54810408GABA receptor activation 3.56×10−04Gabra1, Gabra2, Gabrb2, Gngt2, Kcnj3, Kcnj9  6
4809881Transmission across chemical synapses 8.56×10−04Cacna1a, Gabra1, Gabra2, Gabrb2, Camk2b, Gngt210
4809882Neuronal system 9.71×10−04Cacna1a, Gabra1, Gabra2, Gabrb2, Camk2b, Kcnq512
4810957GABA A receptor activation 1.01×10−03Gabra1, Gabra2, Gabrb2  3
4810204Neurotransmitter receptor binding and downstream transmission in the post-synaptic cell 1.68×10−03Gabra1, Gabra2, Gabrb2, Camk2b, Gngt2, Kcnj3, Kcnj9, Chrnb3  8
Upregulated DEGs in E15.54810016Immune system 1.13×10−07Ifitm3, Ifitm6, Lgals3, H2-K1, Tap2, Bcl2l164
4810918Interferon signaling 5.69×10−05Icam1, Stat1, Irf1, Oas1h, Ncam1, Ifnar213
4810008EGFR interacts with phospholipase C-γ 5.88×10−05Adcy7, Plcg1, Adrbk1, Egfr, Creb1, Camk4, Adcy9  7
4810917Interferon γ signaling 6.00×10−05Icam1, Stat1, H2-K1, Irf1, Oas1h, Ncam1, Socs310
4810011PLCG1 events in ERBB2 signaling 7.26×10−05Adcy7, Plcg1, Adrbk1, Egfr, Creb1, Camk4, Adcy9  7
Downregulated DEGs in E15.54809896Metabolism of water-soluble vitamins and cofactors 2.69×10−03Amn, Mmaa, Mmab, Tcn2, Slc23a2, Mthfr, Rfk, Slc5a6, Mocs1, Ppcs10
4809897Metabolism of vitamins and cofactors 2.69×10−03Amn, Mmaa, Mmab, Tcn2, Slc23a2, Mthfr, Rfk, Slc5a6, Mocs1, Ppcs10
4809898Defective TCN2 causes hereditary megaloblastic anemia 2.69×10−03Amn, Mmaa, Mmab, Tcn2, Slc23a2, Mthfr, Rfk, Slc5a6, Mocs1, Ppcs10
4809899Defects in cobalamin (B12) metabolism 2.69×10−03Amn, Mmaa, Mmab, Tcn2, Slc23a2, Mthfr, Rfk, Slc5a6, Mocs1, Ppcs10
4809900Defects in vitamin and cofactor metabolism 2.69×10−03Amn, Mmaa, Mmab, Tcn2, Slc23a2, Mthfr, Rfk, Slc5a6, Mocs1, Ppcs10
Upregulated4810720Nuclear receptor genes in shared 5.09×10−03Nr6a1, Esrrg transcription pathway'  2
DEGs4810917Interferon γ signaling 9.26×10−03Icam1, Oas1h  2
4810918Interferon signaling 2.35×10−02Icam1, Oas1h  2
4810429Generic transcription pathway 3.07×10−02Nr6a1, Esrrg  2
4810016Immune system 6.30×10−02Icam1, Oas1h, C6, Fbxw11, Kif2a  5
Downregulated4810839Activation of Rac 1.85×10−03Pak3, Pak1  2
genes in shared4810573CD28 co-stimulation 5.02×10−03Pak1, Grap2  2
DEGs4810613Generation of second messenger molecules 5.02×10−03Pak1, Grap2  2
4810625Signaling by Robo receptor 9.02×10−03Pak3, Pak1  2
4810145FCERI mediated MAPK activation 1.55×10−02Pak1, Grap2  2

[i] E13.5, embryonic day 13.5; E15.5, embryonic day 15.5; DEGs, differentially expressed genes.

PPI network construction

STRING was used to construct the PPI networks for DEGs. The PPI networks for up- and downregulated DEGs in E13.5 group contained 114 and 112 nodes, respectively (Fig. 2); the PPI networks for up- and downregulated DEGs in E15.5 group contained 345 and 562 nodes, respectively (Fig. 3). The proteins such as Icam1, Akp3, Stat1 and Brca1 had high connectivity degrees in the PPI networks of E13.5 (Table III) and E15.5 (Table IV) groups. Additionally, the PPI network for shared DEGs contained 17 nodes and 12 PPI pairs (Fig. 4). The shared DEGs with top three connectivity degrees of the PPI network were Icam1, Ncor1 and Akt3 (Table V).

Table III.

DEGs with the top 10% connectivity degree in the PPI network in E13.5 group.

Table III.

DEGs with the top 10% connectivity degree in the PPI network in E13.5 group.

DEGsIDDegreeIDDegreeIDDegreeIDDegree
Upregulated DEGsIcam1  9H2-M95Ube2t4Calb13
H2-L  8Gabrg25Sgip14Itga43
Irf7  7Pxn5Ap2a23
Oas1h  5Il12b4Rnf23
Downregulated DEGsAkp313Ncor17Pik3ca6Akt35
Nr3c110Stat26Bmpr1a6Gna155
Ar  9Il36Gngt25Mut4

[i] DEGs, differentially expressed genes; PPI, protein-protein interaction; E13.5, embryonic day 13.5.

Table IV.

DEGs with the top 10% connectivity degree in the PPI network in E15.5 group.

Table IV.

DEGs with the top 10% connectivity degree in the PPI network in E15.5 group.

DEGsIDDegreeIDDegreeIDDegreeIDDegreeIDDegree
Upregulated DEGsStat138Mmp919Ifih116Ezr13Lgals3bp12
Egfr37Irf118Cxcl115Mbp13Oasl212
Icam127Ccr218Ifi3514Hnrnpa113Rbm2512
Myc27Fpr218Actb14Rmcs213Ccr912
Pten25Ncam117Ccl614Cpsf613H2-Aa12
Stat221Plcg117Cblb14Adrbk112Abl112
Yes120Creb116H2-K114Rsad212
Downregulated DEGsBrca133Dlgap520Tmem4817Rpn112Hao110
Chek130Creb120Hspa516Kcnj1112Ugt2b510
Bub129Aspm20Lsm414Med111Abcb1110
Rrm226Zwint20Akt314Calr11Gnao110
Sgol225Cenpm19Sec61a113Kalrn11Gabrg210
Cep5525Spc2419Lpar313Slc10a111Cd4010
Plk424Eme119Hjurp13Rgn10Chrm410
Tpx224Foxm119Nasp13Cfi10Ppp2r1a  9
Kif2323Xpo118Igf1r13Ddost10Nfatc2  9
Ska123Troap18Gucy1b313Prkx10Hpd  9
Prc123Oip517Ncor112Rae110Fanci  9

[i] DEGs, differentially expressed genes; PPI, protein-protein interaction; E15.5, embryonic day 15.5.

Table V.

Connectivity degrees in the PPI network for shared DEGs in E13.5 and E15.5 groups.

Table V.

Connectivity degrees in the PPI network for shared DEGs in E13.5 and E15.5 groups.

IDDegreeIDDegreeIDDegreeIDDegree
Icam13Oca21Hgf1Tpx21
Ncor13Slc45a21Esrrg1Il31
Akt32Appl11Pak31
Oas1h2Sgip11Pak11
Nr6a12Calb11Iqcd1

[i] DEGs, differentially expressed genes; PPI, protein-protein interaction; E13.5, embryonic day 13.5; E15.5, embryonic day 15.5.

Discussion

Maternal diabetes is a relatively common disease that results in an increased incidence of congenital malformations such as neural tube defects and heart defects (9). CHD are the most common type of birth defects and a main cause of birth defects-related mortality and morbidity (24,25). It has been reported that CHD are closely associated with maternal diabetes (26). Previous study had screened and analyzed several DEGs in embryonic heart tissues from diabetic mother (4). However, the molecular mechanism between CHD and diabetes remains largely unknown. In this study, we have screened 869 and 1,295 in E13.5 and E15.5 groups, respectively and 182 DEGs were shared by two groups. Moreover, the DEGs such as Icam1 and H2-M9 were significantly enriched in cell adhesion molecules (CAMs); DEGs such as Bmpr1a, Tgfbr1 and Smurf1 were enriched in TGF-β signaling pathway; DEGs such as Icam1, C1s and Fcgr2b were enriched in Staphylococcus aureus infection. Finally, several key shared DEGs that were the genes with the top three node degrees in the network were analyzed, including Icam1, Ncor1 and Akt3.

Our results showed that Icam1 had the highest connectivity degree not only in the PPI network of the upregulated DEGs in E13.5 group but also in the PPI network of the shared DEGs. ICAM1, also called CD54, is a cell surface glycoprotein which is typically expressed on endothelial cells and cells of the immune system (27) and the protein is considered to participate in atherogenesis by promoting monocyte accumulation in the arterial intima (28). An earlier study has found that the expression of ICAM1 on endothelial cells and circulating monocytes may be critical for the adhesion of the cells on the vascular endothelium (29). And the expression of ICAM1 has been also detected on cardiac myocytes both in adult humans with unexplained cardiac dysfunction (30) and animals with myocarditis (31,32). According to the pathway enrichment results in our study, we found that Icam1 were enriched in Staphylococcus aureus infection, CAMs and interferon γ signaling. Inflammation is showed to participate in the pathogenesis of type 2 diabetes (33). Upregulation of adhesion molecules such as ICAM1 is pivotal in the development of inflammatory responses (34). Besides, autoimmune-associated congenital heart block (CHB) may result from pathogenic cross-talk between inflammatory and profibrosing pathways (35). Therefore, Icam1 might be involved in the development of heart in mouse embryos.

Ncor1 also had a high node degree in the PPI network for the shared DEGs. It is well known that Ncor1 is a transcriptional coregulator that controls the activity of many transcription factors (such as MEF2, ERRs) and has wide-ranging effects on gene expression patterns (36). MEF2 family has been associated with regulation of myocardially-expressed genes within the heart, such as cardiac α-actin (37). MEF2 is critical for normal heart development and mitochondrial integrity (38). There is a strict relationship between oxygen consumption and cardiac work (39). Oxidative stress is thought to play a particularly critical role in the development of cardiovascular pathology (40). SIRT1 that is part of Ncor1/SMRT complex (36) could retard aging and confer oxidative stress resistance to the heart in vivo (41,42). SMRT (Ncor2), the homolog of Ncor1, is also considered to take part in heart formation (43). Thus, Ncor1, as a transcriptional coregulator, might regulate several genes that are related to the heart development through several different signaling pathways.

Akt3 was interacted with Ncor1 in the PPI network for the shared DEGs. Study has showed that Akt3 is a member of the Akt subfamily that comprises three closely related isoforms Akt1, Akt2 and Akt3. Akt regulates several cellular process including metabolism, cell growth, proliferation, survival and angiogenesis (44). Dysregulation of Akt leads to many diseases such as cancer, diabetes, cardiovascular and neurological diseases (44). Akt has an important role in the functional behavior in the cardiovasculature such as cardiomyocytes, thrombocytes and endothelial cells (44). Akt1 is demonstrated to be essential for heart development and function (45). In addition, Akt3 was found to be enriched in most of the KEGG pathways in shared DEGs, especially renal cell carcinoma. Renal insufficiency in patients with acquired heart failure and ischemic heart disease is related to higher mortality and morbidity (46). Therefore, Akt3 might to be related to the heart development during embryogenesis.

Additionally, there are some limitations in the present study. For example, much more samples should be used to clarify the finding; the expression level of Icam1, Ncor1, and Akt3 should be verified by RT-PCR in the maternal diabetes associated with CHD. However, the present study may provide a scientific guidance for future study to clarify the relationship between CHD and diabetes. It is helpful to explain the molecular mechanism of the CHD development in offspring of diabetic pregnancies. These DEGs may be the therapeutic target in the offspring of diabetic pregnancies with CHD.

In conclusion, we have identified many DEGs in embryonic heart tissue samples at E13.5 and E15.5 from diabetic mice using bioinformatics analysis. And we found that the shared DEGs such as Icam1, Ncor1, and Akt3 had high connectivity degrees in the network. Our study implied that maternal diabetes could affect Icam1, Ncor1, and Akt3 which are critical in the heart development during embryogenesis and might result in CHD. However, more research is needed to confirm these results and further explore the complex molecular mechanism.

References

1 

Association AD: Diagnosis and classification of diabetes mellitus. Diabetes Care. 33 Supplement 1:S62–S69. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Rother KI: Diabetes treatment-bridging the divide. N Engl J Med. 356:1499–1501. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Kim C, Newton KM and Knopp RH: Gestational diabetes and the incidence of type 2 diabetes: A systematic review. Diabetes Care. 25:1862–1868. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Vijaya M, Manikandan J, Parakalan R, Dheen ST, Kumar SD and Tay SS: Differential gene expression profiles during embryonic heart development in diabetic mice pregnancy. Gene. 516:218–227. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Herrera E and Ortega-Senovilla H: Disturbances in lipid metabolism in diabetic pregnancy-are these the cause of the problem? Best Pract Res Clin Endocrinol Metab. 24:515–525. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Fahed AC, Gelb BD, Seidman JG and Seidman CE: Genetics of congenital heart disease: The glass half empty. Circ Res. 112:707–720. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Molin DG, Roest PA, Nordstrand H, Wisse LJ, Poelmann RE, Eriksson UJ and Gittenberger-De Groot AC: Disturbed morphogenesis of cardiac outflow tract and increased rate of aortic arch anomalies in the offspring of diabetic rats. Birth Defects Res A Clin Mol Teratol. 70:927–938. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Dinleyici EC, Tekin N, Dinleyici M, Kilic Z, Adapinar B and Aksit MA: Severe fatal course of axial mesodermal dysplasia spectrum associated with complex cardiac defect in an infant of a mother with insulin dependent diabetes. Am J Med Genet A. 15:2156–2159. 2007. View Article : Google Scholar

9 

Pavlinkova G, Salbaum JM and Kappen C: Maternal diabetes alters transcriptional programs in the developing embryo. BMC Genomics. 10:2742009. View Article : Google Scholar : PubMed/NCBI

10 

Jiang B, Kumar SD, Loh WT, Manikandan J, Ling EA, Tay SS and Dheen ST: Global gene expression analysis of cranial neural tubes in embryos of diabetic mice. J Neurosci Res. 86:3481–3493. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Camenisch TD, Molin DG, Person A, Runyan RB, Gittenberger-de Groot AC, McDonald JA and Klewer SE: Temporal and distinct TGFbeta ligand requirements during mouse and avian endocardial cushion morphogenesis. Dev Biol. 248:170–181. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Han M, Yang X, Farrington JE and Muneoka K: Digit regeneration is regulated by Msx1 and BMP4 in fetal mice. Development. 130:5123–5132. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Wang J, Sridurongrit S, Dudas M, Thomas P, Nagy A, Schneider MD, Epstein JA and Kaartinen V: Atrioventricular cushion transformation is mediated by ALK2 in the developing mouse heart. Dev Biol. 286:299–310. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Conway SJ, Henderson DJ and Copp AJ: Pax3 is required for cardiac neural crest migration in the mouse: Evidence from the splotch (Sp2H) mutant. Development. 124:505–514. 1997.PubMed/NCBI

15 

Kumar SD, Dheen ST and Tay SS: Maternal diabetes induces congenital heart defects in mice by altering the expression of genes involved in cardiovascular development. Cardiovasc Diabetol. 6:342007. View Article : Google Scholar : PubMed/NCBI

16 

Kumar SD, Yong SK, Dheen ST, Bay BH and Tay SS: Cardiac malformations are associated with altered expression of vascular endothelial growth factor and endothelial nitric oxide synthase genes in embryos of diabetic mice. Exp Biol Med. 233:1421–1432. 2008. View Article : Google Scholar

17 

Gautier L, Cope L, Bolstad BM and Irizarry RA: Affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Wettenhall JM and Smyth GK: LimmaGUI: A graphical user interface for linear modeling of microarray data. Bioinformatics. 20:3705–3706. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Yu G, Wang LG, Han Y and He QY: Clusterprofiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Yu G: Using clusterProfiler to identify and compare functional profiles of gene lists. 2014.

21 

Haw R, Hermjakob H, D'Eustachio P and Stein L: Reactome pathway analysis to enrich biological discovery in proteomics data sets. Proteomics. 11:3598–3613. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Fabregat A, Sidiropoulos K, Garapati P, et al: The reactome pathway knowledgebase. Nucleic Acids Research. 44:D481–D487. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networksData Mining in Proteomics. Springer; pp. 291–303. 2011

24 

van der Linde D, Konings EE, Slager MA, Witsenburg M, Helbing WA, Takkenberg JJ and Roos-Hesselink JW: Birth prevalence of congenital heart disease worldwide: A systematic review and meta-analysis. J Am Coll Cardiol. 58:2241–2247. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Gilboa SM, Correa A, Botto LD, Rasmussen SA, Waller DK, Hobbs CA, Cleves MA and Riehle-Colarusso TJ; National Birth Defects Prevention Study, : Association between prepregnancy body mass index and congenital heart defects. Am J Obstet Gynecol. 202:51.e1–51.e10. 2010. View Article : Google Scholar

26 

Lisowski LA, Verheijen PM, Copel JA, Kleinman CS, Wassink S, Visser GH and Meijboom EJ: Congenital heart disease in pregnancies complicated by maternal diabetes mellitus. An international clinical collaboration, literature review, and meta-analysis. Herz. 35:19–26. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Buraczynska M, Zaluska W, Baranowicz-Gaszczyk I, Buraczynska K, Niemczyk E and Ksiazek A: The intercellular adhesion molecule-1 (ICAM-1) gene polymorphism K469E in end-stage renal disease patients with cardiovascular disease. Hum Immunol. 73:824–828. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Iiyama K, Hajra L, Iiyama M, Li H, DiChiara M, Medoff BD and Cybulsky MI: Patterns of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 expression in rabbit and mouse atherosclerotic lesions and at sites predisposed to lesion formation. Circ Res. 85:199–207. 1999. View Article : Google Scholar : PubMed/NCBI

29 

Meerschaert J and Furie MB: The adhesion molecules used by monocytes for migration across endothelium include CD11a/CD18, CD11b/CD18 and VLA-4 on monocytes and ICAM-1, VCAM-1, and other ligands on endothelium. J Immunol. 154:4099–4112. 1995.PubMed/NCBI

30 

Toyozaki T, Saito T, Takano H, Yorimitsu K, Kobayashi S, Ichikawa H, Takeda K and Inagaki Y: Expression of intercellular adhesion molecule-1 on cardiac myocytes for myocarditis before and during immunosuppressive therapy. Am J Cardiol. 72:441–444. 1993. View Article : Google Scholar : PubMed/NCBI

31 

Seko Y, Matsuda H, Kato K, Hashimoto Y, Yagita H, Okumura K and Yazaki Y: Expression of intercellular adhesion molecule-1 in murine hearts with acute myocarditis caused by coxsackievirus B3. J Clin Invest. 91:1327–1336. 1993. View Article : Google Scholar : PubMed/NCBI

32 

Seko Y, Yamazaki T, Shinkai Y, Yagita H, Okumura K, Naito S, Imataka K, Fujii J and Yazaki Y: Cellular and molecular bases for the immunopathology of the myocardial cell damage involved in acute viral myocarditis with special reference to dilated cardiomyopathy. Jpn Circ J. 56:1062–1072. 1992. View Article : Google Scholar : PubMed/NCBI

33 

Donath MY and Shoelson SE: Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 11:98–107. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Duan M, Yao H, Hu G, Chen X, Lund AK and Buch S: HIV Tat induces expression of ICAM-1 in HUVECs: Implications for miR-221/-222 in HIV-associated cardiomyopathy. PLoS One. 8:e601702013. View Article : Google Scholar : PubMed/NCBI

35 

Alvarez D, Briassouli P, Clancy RM, Zavadil J, Reed JH, Abellar RG, Halushka M, Fox-Talbot K, Barrat FJ and Buyon JP: A novel role of endothelin-1 in linking Toll-like receptor 7-mediated inflammation to fibrosis in congenital heart block. J Biol Chem. 286:30444–30454. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Yamamoto H, Williams EG, Mouchiroud L, Cantó C, Fan W, Downes M, Héligon C, Barish GD, Desvergne B, Evans RM, et al: NCoR1 is a conserved physiological modulator of muscle mass and oxidative function. Cell. 147:827–839. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Lockhart MM, Wirrig EE, Phelps AL, Ghatnekar AV, Barth JL, Norris RA and Wessels A: Mef2c regulates transcription of the extracellular matrix protein cartilage link protein 1 in the developing murine heart. PLoS One. 8:e570732013. View Article : Google Scholar : PubMed/NCBI

38 

Planavila A, Dominguez E, Navarro M, Vinciguerra M, Iglesias R, Giralt M, Lope-Piedrafita S, Ruberte J and Villarroya F: Dilated cardiomyopathy and mitochondrial dysfunction in Sirt1-deficient mice: A role for Sirt1-Mef2 in adult heart. J Mol Cell Cardiol. 53:521–531. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Ventura-Clapier R, Garnier A and Veksler V: Energy metabolism in heart failure. J Physiol. 555:1–13. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Pirinccioglu AG, Alyan O, Kizil G, Kangin M and Beyazit N: Evaluation of oxidative stress in children with congenital heart defects. Pediatr Int. 54:94–98. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Vinciguerra M, Santini MP, Martinez C, Pazienza V, Claycomb WC, Giuliani A and Rosenthal N: mIGF-1/JNK1/SirT1 signaling confers protection against oxidative stress in the heart. Aging Cell. 11:139–149. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Alcendor RR, Gao S, Zhai P, Zablocki D, Holle E, Yu X, Tian B, Wagner T, Vatner SF and Sadoshima J: Sirt1 regulates aging and resistance to oxidative stress in the heart. Circ Res. 100:1512–1521. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Mottis A, Mouchiroud L and Auwerx J: Emerging roles of the corepressors NCoR1 and SMRT in homeostasis. Genes Dev. 27:819–835. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Hers I, Vincent EE and Tavaré JM: Akt signalling in health and disease. Cell Signal. 23:1515–1527. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Chang Z, Zhang Q, Feng Q, Xu J, Teng T, Luan Q, Shan C, Hu Y, Hemmings BA, Gao X and Yang Z: Deletion of Akt1 causes heart defects and abnormal cardiomyocyte proliferation. Dev Biol. 347:384–391. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Dimopoulos K, Diller GP, Koltsida E, Pijuan-Domenech A, Papadopoulou SA, Babu-Narayan SV, Salukhe TV, Piepoli MF, Poole-Wilson PA, Best N, et al: Prevalence, predictors, and prognostic value of renal dysfunction in adults with congenital heart disease. Circulation. 117:2320–2328. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 17 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin N, Cai Y, Zhang L and Chen Y: Identification of key genes associated with congenital heart defects in embryos of diabetic mice. Mol Med Rep 17: 3697-3707, 2018
APA
Lin, N., Cai, Y., Zhang, L., & Chen, Y. (2018). Identification of key genes associated with congenital heart defects in embryos of diabetic mice. Molecular Medicine Reports, 17, 3697-3707. https://doi.org/10.3892/mmr.2017.8330
MLA
Lin, N., Cai, Y., Zhang, L., Chen, Y."Identification of key genes associated with congenital heart defects in embryos of diabetic mice". Molecular Medicine Reports 17.3 (2018): 3697-3707.
Chicago
Lin, N., Cai, Y., Zhang, L., Chen, Y."Identification of key genes associated with congenital heart defects in embryos of diabetic mice". Molecular Medicine Reports 17, no. 3 (2018): 3697-3707. https://doi.org/10.3892/mmr.2017.8330