Skip to main content
Erschienen in: BMC Cardiovascular Disorders 1/2021

Open Access 01.12.2021 | Research

Identification of candidate biomarkers and therapeutic agents for heart failure by bioinformatics analysis

verfasst von: Vijayakrishna Kolur, Basavaraj Vastrad, Chanabasayya Vastrad, Shivakumar Kotturshetti, Anandkumar Tengli

Erschienen in: BMC Cardiovascular Disorders | Ausgabe 1/2021

Abstract

Introduction

Heart failure (HF) is a heterogeneous clinical syndrome and affects millions of people all over the world. HF occurs when the cardiac overload and injury, which is a worldwide complaint. The aim of this study was to screen and verify hub genes involved in developmental HF as well as to explore active drug molecules.

Methods

The expression profiling by high throughput sequencing of GSE141910 dataset was downloaded from the Gene Expression Omnibus (GEO) database, which contained 366 samples, including 200 heart failure samples and 166 non heart failure samples. The raw data was integrated to find differentially expressed genes (DEGs) and were further analyzed with bioinformatics analysis. Gene ontology (GO) and REACTOME enrichment analyses were performed via ToppGene; protein–protein interaction (PPI) networks of the DEGs was constructed based on data from the HiPPIE interactome database; modules analysis was performed; target gene—miRNA regulatory network and target gene—TF regulatory network were constructed and analyzed; hub genes were validated; molecular docking studies was performed.

Results

A total of 881 DEGs, including 442 up regulated genes and 439 down regulated genes were observed. Most of the DEGs were significantly enriched in biological adhesion, extracellular matrix, signaling receptor binding, secretion, intrinsic component of plasma membrane, signaling receptor activity, extracellular matrix organization and neutrophil degranulation. The top hub genes ESR1, PYHIN1, PPP2R2B, LCK, TP63, PCLAF, CFTR, TK1, ECT2 and FKBP5 were identified from the PPI network. Module analysis revealed that HF was associated with adaptive immune system and neutrophil degranulation. The target genes, miRNAs and TFs were identified from the target gene—miRNA regulatory network and target gene—TF regulatory network. Furthermore, receiver operating characteristic (ROC) curve analysis and RT-PCR analysis revealed that ESR1, PYHIN1, PPP2R2B, LCK, TP63, PCLAF, CFTR, TK1, ECT2 and FKBP5 might serve as prognostic, diagnostic biomarkers and therapeutic target for HF. The predicted targets of these active molecules were then confirmed.

Conclusion

The current investigation identified a series of key genes and pathways that might be involved in the progression of HF, providing a new understanding of the underlying molecular mechanisms of HF.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12872-021-02146-8.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Heart failure (HF) is a cardiovascular disease characterized by tachycardia, tachypnoea, pulmonary rales, pleural effusion, raised jugular venous pressure, peripheral oedema and hepatomegaly [1]. Morbidity and mortality linked with HF is a prevalent worldwide health problem holding a universal position as the leading cause of death [2]. The numbers of cases of HF are rising globally and it has become a key health issue. According to a survey, the prevalence HF is expected to exceed 50% of the global population [3]. Research suggests that modification in multiple genes and signaling pathways are associated in controlling the advancement of HF. However, a lack of investigation on the precise molecular mechanisms of HF development limits the treatment efficacy of the disease at present.
Previous study showed that HF was related to the expression of MECP2 [4] RBM20 [5], CaMKII [6], troponin I [7] and SERCA2a [8]. Toll-Like receptor signaling pathway [9], activin type II receptor signaling pathway [10], CaMKII signaling pathways [11], Drp1 signaling pathways [12] and JAK-STAT signaling pathway [13] were liable for progression of HF. More investigations are required to focus on treatments that enhance the outcome of patients with HF, to strictly make the diagnosis of the disease based on screening of biomarkers. These investigations can upgrade prognosis of patients by lowering the risk of advancement of HF and related complications. So it is essential to recognize the mechanism and find biomarkers with a good specificity and sensitivity.
The recent high-throughput RNA sequencing data has been widely employed to screen the differentially expressed genes (DEGs) between normal samples and HF samples in human beings, which makes it accessible for us to further explore the entire molecular alterations in HF at multiple levels involving DNA, RNA, proteins, epigenetic alterations, and metabolism [14]. However, there still exist obstacles to put these RNA seq data in application in clinic for the reason that the number of DEGs found by expression profiling by high throughput sequencing were massive and the statistical analyses were also too sophisticated [1519]
In this study, first, we had chosen dataset GSE141910 from Gene Expression Omnibus (GEO) (http:// www.​ncbi.​nlm.​nih.​gov/​geo/​) [20]. Second, we applied for limma tool in R software to obtain the differentially expressed genes (DEGs) in this dataset. Third, the ToppGene was used to analyze these DEGs including biological process (BP), cellular component (CC) and molecular function (MF) REACTOME pathways. Fourth, we established protein–protein interaction (PPI) network and then applied Cytotype PEWCC1 for module analysis of the DEGs which would identify some hub genes. Fifth, we established target gene—miRNA regulatory network and target gene—TF regulatory network. In addition, we further validated the hub genes by receiver operating characteristic (ROC) curve analysis and RT-PCR analysis. Finally, we performed molecular docking studies for over expressed hub genes. Results from the present investigation might provide new vision into potential prognostic and therapeutic targets for HF.

Materials and methods

Data resource

Expression profiling by high throughput sequencing with series number GSE141910 based on platform GPL16791 was downloaded from the GEO database. The dataset of GSE141910 contained 200 heart failure samples and 166 non heart failure samples. It was downloaded from the GEO database in NCBI based on the platform of GPL16791 Illumina HiSeq 2500 (Homo sapiens).

Identification of DEGs in HF

DEGs of dataset GSE141910 between HF groups and non heart failure groups were respectively analyzed using the limma package in R [21]. Fold changes (FCs) in the expression of individual genes were calculated and DEGs with P < 0.05, |log FC|> 1.158 for up regulated genes and |log FC|< − 0.83 for down regulated genes were considered to be significant. Hierarchical clustering and visualization were used by Heat-map package of R.

Functional enrichment analysis

Gene Ontology (GO) analysis and REACTOME pathway analysis were performed to determine the functions of DEGs using the ToppGene (ToppFun) (https://​toppgene.​cchmc.​org/​enrichment.​jsp) [22] GO terms (http://​geneontology.​org/​) [23] included biological processes (BP), cellular components (CC) and molecular functions (MF) of genomic products. REACTOME (https://​reactome.​org/​) [24] analyzes pathways of important gene products. ToppGene is a bioinformatics database for analyzing the functional interpretation of lists of proteins and genes. The cutoff value was set to P < 0.05.

Protein–protein interaction network construction and module screening

PPI networks are used to establish all protein coding genes into a massive biological network that serves an advance compassionate of the functional system of the proteome [25]. The HiPPIE interactome (https://​cbdm.​uni-mainz.​de/​hippie/​) [26] database furnish information regarding predicted and experimental interactions of proteins. In the current investigation, the DEGs were mapped into the HiPPIE interactome database to find significant protein pairs with a combined score of > 0.4. The PPI network was subsequently constructed using Cytoscape software, version 3.8.2 (www.​cytoscape.​org) [27]. The nodes with a higher node degree [28], higher betweenness centrality [29], higher stress centrality [30] and higher closeness centrality [31] were considered as hub genes. Additionally, cluster analysis for identifying significant function modules with a degree cutoff > 2 in the PPI network was performed using the PEWCC1 (http://​apps.​cytoscape.​org/​apps/​PEWCC1) [32] in Cytoscape.

Target gene—miRNA regulatory network construction

The miRNet database (https://​www.​mirnet.​ca/​) [33] contains information on miRNA and the regulated genes. Using information collected from the miRNet database, hub genes were matched with their associated miRNA. The target gene—miRNA regulatory network then was constructed using Cytoscape software. MiRNAs and target are selected based on highest node degree.

Target gene—TF regulatory network construction

The NetworkAnalyst database (https://​www.​networkanalyst.​ca/​) [34] contains information on TF and the regulated genes. Using information collected from the NetworkAnalyst database, hub genes were matched with their associated TF. The target gene—TF regulatory network then was constructed using Cytoscape software. TFs and target genes are selected based on highest node degree.

Receiver operating characteristic (ROC) curve analysis

Then ROC curve analysis was implementing to classify the sensitivity and specificity of the hub genes for HF diagnosis and we investigated how large the area under the curve (AUC) was by using the statistical package pROC in R software [35].

RT-PCR analysis

H9C2 cells (ATCC) were cultured in Dulbecco’s minimal essential medium (DMEM) (Sigma-Aldrich) supplemented with 10% fetal calf serum (Sigma-Aldrich) and 1% streptomycin (Sigma-Aldrich) at 37 °C in 5% CO2. HL-1 cells (ATCC) was culture in Claycomb medium (Sigma-Aldrich) supplemented with 10% fetal bovine serum (Sigma-Aldrich), 1% streptomycin (Sigma-Aldrich), 1% glutamax (Sigma-Aldrich) and 0.1 mM norepinephrine (Sigma-Aldrich) at 37 °C in 5% CO2. Total RNA was isolated from cell culture of H9C2 for HF and HL-1 for normal control using the TRI Reagent (Sigma, USA). cDNA was synthesized using 2.0 μg of total RNA with the Reverse transcription cDNA kit (Thermo Fisher Scientific, Waltham, MA, USA). The 7 Flex real-time PCR system (Thermo Fisher Scientific, Waltham, MA, USA) was employed to detect the relative mRNA expression. The relative expression levels were determined by the 2-ΔΔCt method and normalized to internal control beta-actin [36]. All RT-PCR reactions were performed in triplicate. The primers used to explore mRNA expression of ten hub genes were listed in Table 1.
Table 1
The sequences of primers for quantitative RT-PCR
Genes
Forward primers
Reverse primers
ESR1
CCTCTGGCTACCATTATGGG
AGTCATTGTGTCCTTGAATGC
PYHIN1
GCAAGATCAGTACGACAGAG
AGATAACTGAGCAACCTGTG
PPP2R2B
ACCAGAGACTATCTGACCG
GTAGTCATGAACCTGGTATGTC
LCK
CTAGTCCGGCTTTATGCAG
AAATCTACTAGGCTCCCGT
TP63
ATTCAATGAGGGACAGATTGC
GGGTCTTCTACATACTGGGC
PCLAF
GACCAATATAAACTGTGGCGGG
CCAGGGTAAACAAGGAGACGTT
CFTR
CTGTGGCCTTGGTTTACTG
CTCTGATCTCTGTACTTCACCA
TK1
AGATTCAGGTGATTCTCGGG
ACTTGTACTGGGCGATCTG
ECT2
GCTGTATTGTACGAGTATGCT
GTCACCAATTTGACAAGCTC
FKBP5
CCTAAGTTTGGCATTGACCC
CCAAGATTCTTTGGCCTTCTC

Identification of candidate small molecules

SYBYL-X 2.0 perpetual drug design software has been used for surflex-docking studies of the designed novel molecules and the standard on over expressed genes of PDB protein. Using ChemDraw Software, all designed molecules and standards were sketched, imported and saved using open babel free software in sdf. template. The protein of over expressed genes of ESR1, LCK, PPP2R2B, TP63 and their co-crystallised protein of PDB code 4PXM, 1KSW, 2HV7, 3VD8 and 6RU6 were extracted from Protein Data Bank [3740]. Optimizations of the designed molecules were performed by standard process by applying Gasteiger Huckel (GH) charges together with the TRIPOS force field. In addition, energy minimization was achieved using MMFF94s and MMFF94 algorithm methods. The preparation of the protein was done after protein incorporation. The co-crystallized ligand and all water molecules have been eliminated from the crystal structure; more hydrogen’s were added and the side chain was set, TRIPOS force field was used for the minimization of structure. The interaction efficiency of the compounds with the receptor was expressed in kcal/mol units by the Surflex-Dock score. The best location was integrated into the molecular region by the interaction between the protein and the ligand. Using Discovery Studio Visualizer, the visualisation of ligand interaction with receptor is performed.

Results

Identification of DEGs in HF

We identified 881 DEGs in the GSE141910 dataset using the limma package in R. Based on the limma analysis, using the adj P val < 0.05, |log FC|> 1.158 for up regulated genes and |log FC|< − 0.83 for down regulated genes, a total of 881 DEGs were identified, consisting of 442 genes were up regulated and 439 genes were down regulated. The DEGs are listed in Additional file 1: Table S1. The volcano plot for DEGs is illustrated in Fig. 1. Figure 2 is the hierarchical clustering heat-map.

Functional enrichment analysis

Results of GO analysis showed that the up regulated genes were significantly enriched in BP, CC, and MF, including biological adhesion, regulation of immune system process, extracellular matrix, cell surface, signaling receptor binding and molecular function regulator (Table 2); the down regulated genes were significantly enriched in BP, CC, and MF, including secretion, defense response, intrinsic component of plasma membrane, whole membrane, signaling receptor activity and molecular transducer activity (Table 2). Pathway analysis showed that the up regulated genes were significantly enriched in extracellular matrix organization and immunoregulatory interactions between a lymphoid and a non-lymphoid cell (Table 3); the down regulated genes were significantly enriched in neutrophil degranulation and SLC-mediated transmembrane transport (Table 3).
Table 2
The enriched GO terms of the up and down regulated differentially expressed genes
GO ID
CATEGORY
GO Name
P Value
FDR B&H
FDR B&Y
Bonferroni
Gene Count
Gene
Up regulated genes
GO:0022610
BP
biological adhesion
1.32E−13
3.37E−10
3.08E−09
6.75E−10
72
HLA-DQA1, DACT2, CD83, MDK, UBASH3A, ITGBL1, FAP, MFAP4, SERPINE2, NRXN2, COL14A1, CCR7, ALOX15, COL1A1, LAMB4, COL8A2, STAB2, COL16A1, COMP, TBX21, FERMT1, XG, CCDC80, APOA1, PODXL2, ZAP70, HAPLN1, TENM4, SKAP1, CNTNAP2, PDE5A, CARD11, CTNNA2, SLAMF7, ATP1B2, CX3CR1, LRRC15, IDO1, MYOC, SIGLEC8, ISLR, SMOC2, ITGAL, ITGB7, FREM1, PTN, KIRREL3, NTM, GLI2, FBLN7, DPT, NT5E, ECM2, LCK, OMG, OPCML, TGFB2, RASGRP1, CD2, CD3E, THBS4, CD5, CD6, THY1, TIGIT, CD27, CD40LG, ROBO2, GREM1, LY9, HBB, LEF1
GO:0002682
BP
regulation of immune system process
2.20E−10
2.25E−07
2.05E−06
1.12E−06
72
IL34, HLA-DQA1, ESR1, TLR7, CD83, IL17D, MDK, UBASH3A, TNFRSF4, PYHIN1, ZBP1, FCER1A, MS4A2, FCER2, FCN1, CCR7, SMPD3, CCL24, SCARA3, ALOX15, COL1A1, IL31RA, TBX21, XG, CXCL14, APOA1, ZAP70, SH2D1B, SKAP1, PDE5A, CARD11, SLAMF7, CTSG, CX3CR1, IDO1, CXCL10, ITGAL, ACE, SIT1, ITGB7, PTN, TBC1D10C, FCRL3, BPI, GLI2, KLRB1, NPPA, CAMK4, LCK, TGFB2, RASGRP1, CD1C, CD1E, CD2, CD3D, CD3E, THBS4, CD3G, CD247, CD5, CD6, THY1, TIGIT, MS4A1, CD27, GPR68, CD40LG, CD48, GREM1, SH2D1A, LEF1, LRRC17
GO:0031012
CC
extracellular matrix
1.09E−20
2.77E−18
1.89E−17
5.54E−18
52
MATN2, COL22A1, MDK, COLQ, MFAP4, SERPINE2, HMCN2, AEBP1, FCN1, CMA1, CTHRC1, COL14A1, SCARA3, COL1A1, LAMB4, COL8A2, COL9A1, COL9A2, COL10A1, MXRA5, FMOD, COL16A1, COMP, CCDC80, APOA1, HAPLN1, CTSG, ADAMTSL2, LRRC15, ASPN, MYOC, NDP, SMOC2, FREM1, PTN, SSC5D, SULF1, DPT, NPPA, ADAMTSL1, ECM2, OGN, ITIH5, TGFB2, LEFTY2, EYS, THBS4, P3H2, LTBP2, GREM1, LUM, LRRC17
GO:0009986
CC
cell surface
5.07E−17
8.61E−15
5.86E−14
2.58E−14
63
HYAL4, NRG1, HLA-DQA1, CD83, TNFRSF4, ITGBL1, FAP, SERPINE2, FCER1A, MS4A2, FCER2, FCN1, CXCL9, CCR7, IL31RA, SFRP4, STAB2, DUOX2, APOA1, ACKR4, FCRL6, SCUBE2, CNTNAP2, SLAMF7, CTSG, IL2RB, CX3CR1, LRRC15, CXCL10, NDP, ITGAL, ACE, ITGB7, GFRA3, PTN, PROM1, SSC5D, FCRL3, SULF1, MRC2, NTM, CLEC9A, NT5E, TGFB2, LHCGR, CD1C, HHIP, CD1E, CD2, CD3D, CD3E, CD3G, CD5, CD6, THY1, TIGIT, MS4A1, CD27, CD40LG, CD48, ROBO2, GREM1, LY9
GO:0005102
MF
signaling receptor binding
1.36E−09
5.99E−07
4.41E−06
1.20E−06
73
IL34, NRG1, HLA-DQA1, ESR1, GDF6, PENK, TAC4, KDM5D, IL17D, MDK, ITGBL1, FAP, SERPINE2, FCER2, NRXN2, FCN1, CLEC11A, UCHL1, AGTR2, CXCL9, NGEF, CTHRC1, C1QTNF2, CCL22, CCL24, CXCL11, COL16A1, COMP, WNT10B, WNT9A, CXCL14, APOA1, FCRL6, GNA14, OASL, RASL11B, LRRC15, CXCL10, ADAM18, MYOC, SYTL2, NDP, ACE, GDNF, ITGB7, GFRA3, PTN, LYPD1, SCG2, NPPA, NPPB, MCHR1, ECM2, CMTM2, ESM1, LCK, OGN, TGFB2, LEFTY2, CD2, CD3E, THBS4, CD3G, THY1, TIGIT, MS4A1, C1QTNF9, CD40LG, LTB, GREM1, SYTL1, LEF1, LGI1
GO:0098772
MF
molecular function regulator
3.79E−04
1.59E−02
1.17E−01
3.34E−01
58
IL34, NRG1, ESR1, GDF6, PENK, TAC4, IL17D, MDK, KCNIP1, SERPINE2, MYOZ1, NRXN2, CLEC11A, SCN2B, AGTR2, CXCL9, NGEF, CCL22, CCL24, CXCL11, HTR2B, PI16, SCG5, WNT10B, WNT9A, CXCL14, APOA1, LRRC55, PPP2R2B, ATP1B2, CXCL10, NDP, BIRC7, GDNF, PTN, TBC1D10C, LYPD1, SCG2, NPPA, NPPB, AZIN2, CMTM2, OGN, RGS4, ITIH5, TGFB2, LEFTY2, RASGRP1, THBS4, THY1, CD27, C1QTNF9, CD40LG, RGS17, LTB, GREM1, LEF1, INKA1
Down regulated genes
GO:0046903
BP
secretion
1.07E−11
5.64E−08
5.16E−07
5.64E−08
78
SERPINA3, HK3, SYN3, ACP3, TRPC4, CFTR, CD109, HMOX2, CHI3L1, F5, F8, F13A1, S100A8, S100A9, SAA1, FCER1G, MGST1, PIK3C2A, HP, AGTR1, PLA2G2A, CCR1, FGF10, C1QTNF1, PLA2G4F, FGR, MERTK, SERPINF2, ALOX5, SYT13, IL17RB, CNR1, ALOX15B, FLT3, ANPEP, P2RY12, ANXA3, FPR1, CR1, SLC1A1, SLC2A1, ARG1, ARNTL, SLC11A1, SLC22A16, LGI3, NSG1, ATP2A2, IL10, SIGLEC9, GPR84, NHLRC2, SSTR5, HPSE, KCNB1, IL1R2, PTX3, GLUL, SYN2, BANK1, WNK3, KNG1, CRISPLD2, CACNA1E, CD177, SIGLEC14, EDN1, EDN2, EDNRB, THBS1, RNASE2, CD38, TLR2, SERPINE1, ELANE, STEAP3, IL1RL1, MCEMP1
GO:0006952
BP
defense response
1.04E−06
1.63E−04
1.49E−03
5.50E−03
65
SERPINA3, EREG, VSIG4, TMIGD3, CLEC7A, RAET1E, CHI3L1, F8, CD163, S100A8, S100A9, SAA1, FCER1G, HP, HPR, AGTR1, PLA2G2A, CCR1, FGR, SERPINF2, ALOX5, ALOX5AP, IL17RB, CNR1, SELE, ADAMTS4, ANXA3, FPR1, APOB, SAMHD1, CR1, FCN3, AQP4, ARG1, SLC11A1, MARCO, IL10, BCL6, IL18R1, GGT5, IL1R2, PTX3, SIGLEC10, KNG1, CACNA1E, CD177, SOCS3, SIGLEC14, ADAMTS5, LBP, S1PR3, EDN1, EDNRB, FOSL1, THBS1, RNASE2, NAMPT, TLR2, SERPINE1, ELANE, IRAK3, ELF3, IL1RL1, CALCRL, OSMR
GO:0031226
CC
intrinsic component of plasma membrane
1.74E−10
4.55E−08
3.11E−07
9.10E−08
74
TPO, EREG, OPN4, TRPC4, CFTR, TMIGD3, KCNIP2, CD163, FCER1G, SCN3A, AGTR1, CCR1, C1QTNF1, MERTK, SYT13, IL17RB, CNR1, TRHDE, SELE, LRRC8E, FLT3, SLC4A7, P2RY12, SLC31A2, CR1, LGR5, AQP3, AQP4, SLC1A1, SLC2A1, SLC5A1, MSR1, SLC11A1, SIGLEC7, ART3, SLCO2A1, ATP2A2, MARCO, GABRR2, SIGLEC9, SLCO4A1, GPR84, SSTR2, SSTR5, IL18R1, LAPTM5, GGT5, SLC52A3, LYVE1, KCNA7, KCNB1, KCND3, NECTIN1, KCNK1, KCNK3, KCNS2, ADGRD1, CACNA1E, GPR4, GPR12, SLC38A4, GPR183, GPRC5A, RGR, S1PR3, RHAG, EDNRB, TGFBR3, TLR2, LGR6, CALCRL, OSMR, HAS2, CDH16
GO:0098805
CC
whole membrane
1.91E−03
4.99E−02
3.41E−01
9.97E−01
51
EREG, SYN3, ACP3, TRPC4, CFTR, CD109, HMOX2, CD163, FCER1G, MGST1, PLA2G4F, GPAT2, MOG, CNR1, SELE, ANPEP, P2RY12, ANXA3, FPR1, APOB, SCGN, CR1, AQP4, SLC1A1, SLC2A1, ARG1, MSR1, SLC11A1, NSG1, RAB39A, MARCO, SIGLEC9, GPR84, HPSE, LAPTM5, KCND3, SYN2, SLC9A7, WASF1, CD177, SIGLEC14, GRB14, STEAP4, EDNRB, GRIP1, CD38, TLR2, STEAP3, HAS2, SERPINA5, MCEMP1
GO:0038023
MF
signaling receptor activity
2.36E−04
1.97E−02
1.49E−01
2.53E−01
55
EREG, OPN4, CLEC7A, FCER1G, FCGR3A, AGTR1, CCR1, ADGRF5, ADGRF4, MERTK, IL17RB, CNR1, SELE, FLT3, MYOT, ANPEP, P2RY12, ANXA3, FPR1, CR1, LGR5, SIGLEC7, MARCO, PALLD, IL10, GABRR2, IL15RA, GPR82, DNER, PAQR5, GPR84, IL20RA, SSTR2, SSTR5, IL18R1, LYVE1, IL1R2, NECTIN1, ADGRD1, GPR4, GPR12, NPTX2, GPR183, GPRC5A, PKHD1L1, RGR, S1PR3, EDNRB, TGFBR3, TLR2, SERPINE1, IL1RL1, LGR6, CALCRL, OSMR
GO:0060089
MF
molecular transducer activity
4.71E−04
2.10E−02
1.59E−01
5.04E−01
58
EREG, OPN4, CLEC7A, FCER1G, FCGR3A, AGTR1, CCR1, ADGRF5, ADGRF4, MERTK, IL17RB, CNR1, SELE, FLT3, MYOT, ANPEP, P2RY12, ANXA3, FPR1, CR1, LGR5, SIGLEC7, MARCO, PALLD, IL10, GABRR2, IL15RA, GPR82, DNER, PAQR5, GPR84, IL20RA, STOX1, SSTR2, SSTR5, IL18R1, BLM, CDKL5, LYVE1, IL1R2, NECTIN1, ADGRD1, GPR4, GPR12, NPTX2, GPR183, GPRC5A, PKHD1L1, RGR, S1PR3, EDNRB, TGFBR3, TLR2, SERPINE1, IL1RL1, LGR6, CALCRL, OSMR
Biological Process(BP), Cellular Component(CC) and Molecular Functions (MF)
Table 3
The enriched pathway terms of the up and down regulated differentially expressed genes
Pathway ID
Pathway name
P-value
FDR B&H
FDR B&Y
Bonferroni
Gene count
Gene
Up regulated genes
1270244
Extracellular matrix organization
3.33E−08
1.80E−05
1.23E−04
1.80E−05
24
COL22A1, MFAP4, CMA1, COL14A1, COL1A1, COL8A2, COL9A1, COL9A2, COL10A1, FMOD, COL16A1, COMP, HAPLN1, ADAMTS14, CTSG, ASPN, ITGAL, ITGB7, CAPN6, TGFB2, P3H2, TLL2, LTBP2, LUM
1269201
Immunoregulatory interactions between a Lymphoid and a non-Lymphoid cell
8.13E−06
7.31E−04
5.03E−03
4.39E−03
13
SH2D1B, SLAMF7, SIGLEC8, ITGAL, ITGB7, KLRB1, CD1C, CD3D, CD3E, CD3G, CD247, CD40LG, SH2D1A
1269544
GPCR ligand binding
3.92E−04
1.51E−02
1.04E−01
2.12E−01
22
GNG8, PENK, F2RL2, AGTR2, APLNR, CXCL9, CCR7, CXCL11, OXER1, HTR2A, HTR2B, WNT10B, WNT9A, ACKR4, CRHBP, S1PR5, FZD2, CX3CR1, CXCL10, MCHR1, LHCGR, GPR68
1268749
Metabolism of Angiotensinogen to Angiotensins
5.26E−04
1.85E−02
1.27E−01
2.84E−01
4
CMA1, CTSG, ACE, GZMH
1269868
Muscle contraction
3.44E−02
4.03E−01
1.00E+00
1.00E+00
9
KCNIP1, RYR3, SCN2B, ATP1A4, ATP1B2, MYL1, KCNK17, NPPA, TNNI1
1269340
Hemostasis
6.57E−02
5.21E−01
1.00E+00
1.00E+00
20
GNG8, CEACAM3, F2RL2, SERPINE2, APOA1, GNA14, PDE5A, ATP1B2, IL2RB, CTSW, ISLR, ITGAL, LCK, TGFB2, LEFTY2, RASGRP1, CD2, P2RX6, CD48, HBB
1269171
Adaptive Immune System
1.32E−01
6.87E−01
1.00E+00
1.00E+00
23
NRG1, HLA-DQA1, ZAP70, SH2D1B, CARD11, SLAMF7, SIGLEC8, ITGAL, ITGB7, ASB18, IER3, MRC2, KLRB1, LCK, RASGRP1, CD1C, CD3D, CD3E, CD3G, CD247, FBXL16, CD40LG, SH2D1A
Down regulated genes
1457780
Neutrophil degranulation
4.82E−06
3.14E−03
2.22E−02
3.14E−03
28
SERPINA3, HK3, ACP3, HMOX2, CHI3L1, S100A8, S100A9, FCER1G, MGST1, HP, FGR, ALOX5, ANPEP, FPR1, CR1, ARG1, SLC11A1, SIGLEC9, GPR84, HPSE, PTX3, CRISPLD2, CD177, SIGLEC14, RNASE2, TLR2, ELANE, MCEMP1
1269907
SLC-mediated transmembrane transport
6.91E−04
4.74E−02
3.35E−01
4.51E−01
16
HK3, SLC7A11, SLC4A7, SLC1A1, SLC2A1, SLC5A1, SLC11A1, SLC22A16, SLCO2A1, SLCO4A1, GCKR, LCN15, SLC9A7, SLC25A18, SLC38A4, RHAG
1269545
Class A/1 (Rhodopsin-like receptors)
8.72E−04
4.74E−02
3.35E−01
5.69E−01
17
OPN4, SAA1, AGTR1, CCR1, CNR1, P2RY12, FPR1, SSTR2, SSTR5, KNG1, GPR4, GPR183, RGR, S1PR3, EDN1, EDN2, EDNRB
1269340
Hemostasis
2.18E−03
7.11E−02
5.02E−01
1.00E+00
26
SERPINA3, CD109, SLC7A11, F5, F8, F13A1, SERPINB8, FCER1G, FGR, MERTK, SERPINF2, SELE, P2RY12, APOB, KIF18B, ATP2A2, NHLRC2, KNG1, PDE11A, CD177, DOCK9, GRB7, GRB14, THBS1, SERPINE1, SERPINA5
1269903
Transmembranetransport of small molecules
4.89E−03
1.28E−01
9.01E−01
1.00E+00
26
HK3, TRPC4, CFTR, HMOX2, SLC7A11, ABCB1, SLC4A7, AQP3, AQP4, SLC1A1, SLC2A1, SLC5A1, SLC11A1, SLC22A16, SLCO2A1, ATP2A2, GABRR2, SLCO4A1, GCKR, LCN15, SLC9A7, WNK3, SLC25A18, SLC38A4, RHAG, STEAP3
1269203
Innate Immune System
9.62E−03
1.96E−01
1.00E+00
1.00E+00
42
SERPINA3, EREG, HK3, MARK3, ACP3, CLEC7A, HMOX2, CHI3L1, S100A8, S100A9, SAA1, FCER1G, MGST1, FCGR3A, HP, GRAP2, PLA2G2A, FGF5, FGF10, FGR, ALOX5, ANPEP, FPR1, APOB, CR1, FCN3, ARG1, SLC11A1, SIGLEC9, GPR84, HPSE, PTX3, WASF1, CRISPLD2, CD177, SIGLEC14, LBP, RNASE2, TLR2, ELANE, IRAK3, MCEMP1
1269310
Cytokine Signaling in Immune system
8.42E−02
4.76E−01
1.00E+00
1.00E+00
23
EREG, MARK3, F13A1, SAA1, FGF5, CCR1, FGF10, ALOX5, IL17RB, FLT3, FPR1, SAMHD1, IL10, IL15RA, IL20RA, BCL6, IL18R1, IL1R2, SOCS3, LBP, IRAK3, IL1RL1, OSMR

Protein–protein interaction (PPI) network and module analysis

Based on the HiPPIE interactome database, the PPI network for the DEGs (including 6541 nodes and 13,909 edges) was constructed (Fig. 3A). Up regulated gene with higher node degree, higher betweenness centrality, higher stress centrality and higher closeness centrality were as follows: ESR1, PYHIN1, PPP2R2B, LCK, TP63 and so on. Down regulated genes had higher node degree, higher betweenness centrality, higher stress centrality and higher closeness centrality were as follows PCLAF, CFTR, TK1, ECT2, FKBP5 and so on. The node degree, betweenness centrality, stress centrality and closeness centrality are listed in Table 4.
Table 4
Topology table for up and down regulated genes
Regulation
Node
Degree
Betweenness
Stress
Closeness
Up
ESR1
1094
0.250896
7.4E+08
0.392769
Up
PYHIN1
342
0.054258
1.1E+08
0.339882
Up
PPP2R2B
199
0.023115
35,268,762
0.346839
Up
LCK
162
0.033618
18,589,354
0.353915
Up
TP63
142
0.018577
39,426,608
0.319664
Up
CD247
129
0.013832
18,625,274
0.317784
Up
PTN
105
0.016638
36,892,166
0.300662
Up
APLNR
103
0.016611
40,715,208
0.288093
Up
APOA1
100
0.018834
13,935,332
0.315866
Up
CENPA
98
0.009846
31,131,318
0.301786
Up
SKAP1
97
0.01599
10,109,482
0.313338
Up
FSCN1
88
0.010226
9,387,016
0.335367
Up
SCN2B
86
0.01089
24,233,242
0.284756
Up
TMEM30B
79
0.016411
10,490,230
0.270807
Up
FOXS1
79
0.009408
26,398,078
0.293247
Up
COL1A1
76
0.010472
9,315,670
0.308098
Up
ZAP70
75
0.007096
5,315,196
0.317614
Up
UCHL1
74
0.009753
9,381,024
0.331525
Up
HBB
72
0.009984
14,209,592
0.304767
Up
NRG1
70
0.01151
13,449,066
0.29526
Up
LEF1
61
0.007998
18,260,094
0.290744
Up
NT5E
60
0.009599
11,470,634
0.301563
Up
MDK
59
0.006889
7,533,352
0.305822
Up
ISLR
58
0.010139
9,992,806
0.294012
Up
FATE1
57
0.011609
9,248,788
0.281581
Up
LRRC15
56
0.010069
4,772,792
0.299094
Up
MATN2
54
0.004491
9,580,600
0.286792
Up
LIPH
54
0.008197
6,873,658
0.282347
Up
MYOC
49
0.005138
12,313,276
0.291029
Up
SCARA3
49
0.006492
11,494,220
0.290151
Up
NPPA
46
0.009502
4,868,574
0.307143
Up
CD83
43
0.005149
5,484,906
0.272841
Up
COL14A1
41
0.006129
5,699,128
0.292893
Up
CTSG
40
0.003821
1,913,996
0.296155
Up
SFRP4
40
0.004006
7,876,966
0.282518
Up
TRAF3IP3
38
0.006447
4,263,002
0.290602
Up
CLEC11A
38
0.005085
3,520,012
0.283596
Up
ATP1B4
38
0.005722
2,476,482
0.245939
Up
CD3E
37
0.003892
1,537,326
0.297624
Up
SH2D1A
37
0.003969
2,117,220
0.308113
Up
DDX3Y
37
0.003754
1,632,916
0.32095
Up
PRPH
37
0.001871
1,994,266
0.301494
Up
BIRC7
35
0.004835
2,567,254
0.28549
Up
CARD11
35
0.002249
2,510,852
0.292173
Up
RXRG
35
0.002394
7,134,478
0.26225
Up
CCL22
34
0.005706
3,916,024
0.281423
Up
CD27
33
0.003915
2,076,446
0.294263
Up
GZMB
32
0.003277
6,141,522
0.285602
Up
THY1
32
0.003561
1,793,684
0.291509
Up
CHRNA3
32
0.004247
3,415,720
0.236631
Up
LSP1
32
0.003371
5,734,010
0.281836
Up
IL2RB
31
0.002351
1,938,172
0.305808
Up
HTR2B
30
0.004018
2,165,998
0.27434
Up
DLGAP1
29
0.003816
6,072,882
0.277754
Up
TRIM17
29
0.002943
5,543,810
0.275137
Up
CTNNA2
29
0.003554
3,511,510
0.30254
Up
SERPINE2
28
0.002577
3,031,536
0.27426
Up
CD1E
28
0.003282
3,419,392
0.255229
Up
MRC2
28
0.003395
2,950,548
0.296276
Up
C1QTNF2
28
0.003203
2,505,388
0.270147
Up
SH2D1B
27
0.001864
1,043,570
0.29028
Up
BRINP1
27
0.001516
4,402,948
0.27726
Up
PDIA2
27
0.001967
2,895,182
0.286453
Up
CHD5
27
0.001918
5,223,636
0.286503
Up
FAP
27
0.003531
5,820,086
0.268583
Up
IL31RA
26
0.002073
1,606,954
0.263603
Up
GAP43
25
0.002745
2,793,268
0.279858
Up
CD5
25
0.001695
655,904
0.295861
Up
UBASH3A
25
0.001652
1,347,264
0.290951
Up
ROBO2
25
0.002959
3,726,616
0.267048
Up
ITGB7
24
0.002686
3,351,660
0.276124
Up
HTR2A
24
0.002571
2,472,010
0.275833
Up
MOXD1
24
0.002391
2,492,756
0.259926
Up
ASB18
24
9.77E−04
3,202,162
0.273001
Up
CD2
23
0.002221
926,408
0.287954
Up
BCL11B
23
8.18E−04
1,888,298
0.28836
Up
STAT4
23
0.001786
2,435,506
0.277166
Up
NGEF
23
0.001809
1,548,206
0.277636
Up
SMPD3
23
0.002518
2,175,774
0.281
Up
FZD2
22
0.003673
3,239,390
0.250335
Up
DUSP15
22
0.001253
2,474,532
0.284472
Up
CD3D
21
0.001725
1,111,132
0.288589
Up
SYT17
21
0.002549
2,482,574
0.285802
Up
FCGR3B
21
0.002748
1,492,022
0.282286
Up
EGR2
21
0.002934
3,438,856
0.266406
Up
ZBP1
21
0.001876
2,664,938
0.26006
Up
CAMK4
21
0.001773
3,472,134
0.272716
Up
DMC1
20
0.002511
4,659,098
0.254277
Up
GDNF
20
0.002515
3,274,958
0.244751
Up
FCN1
20
0.002571
1,243,380
0.236742
Up
LUM
20
0.002276
1,515,870
0.283903
Up
GZMA
20
0.001051
3,258,230
0.276498
Up
TGFB2
20
0.002259
1,632,566
0.277119
Up
SLAMF7
20
0.00211
1,035,482
0.271708
Up
MS4A1
20
0.002857
1,169,078
0.288398
Up
ETV4
20
0.001747
1,674,080
0.301883
Up
GLI2
20
0.001398
2,977,194
0.285902
Up
PHLDA1
19
4.37E−04
818,256
0.298194
Up
COL8A2
19
0.00147
1,089,762
0.273835
Up
GABRD
19
0.002826
2,629,544
0.25748
Up
LMF1
19
0.004342
2,024,228
0.265132
Up
F2RL2
19
0.001554
790,338
0.282933
Up
LYPD1
19
0.003123
3,995,458
0.266276
Up
CAPN6
19
0.001415
3,046,736
0.267802
Up
SOX8
19
0.003361
2,763,024
0.251306
Up
IER3
18
0.001921
3,613,164
0.282982
Up
BEX1
18
0.001034
1,286,206
0.273606
Up
COLQ
18
0.001173
1,414,826
0.261234
Up
NTM
18
0.00284
2,486,684
0.275102
Up
RPS4Y1
18
0.001013
1,200,768
0.287713
Up
FERMT1
18
0.001713
4,279,868
0.270315
Up
RGS17
18
0.002928
3,868,106
0.249895
Up
TNNI1
17
0.001349
1,550,004
0.266765
Up
MYOZ1
17
0.00128
2,111,156
0.283203
Up
KLHDC8A
17
0.001147
7,007,508
0.251036
Up
MYL1
17
7.90E−04
1,213,666
0.289945
Up
DIO2
16
0.001161
1,959,228
0.279416
Up
ITGAL
16
0.001182
1,521,116
0.271527
Up
CRABP2
16
4.13E−04
675,182
0.272171
Up
HSH2D
16
0.001425
889,856
0.26034
Up
CD48
3
0
0
0.265422
Up
CD3G
2
0
0
0.23833
Up
LY9
2
0
0
0.240141
Up
SIT1
2
0
0
0.264221
Up
ATP1A4
2
1.16E−04
79,140
0.235049
Up
FMOD
2
3.96E−05
20,526
0.240707
Up
CCDC80
2
3.58E−05
499,704
0.288908
Up
CCR7
2
0
0
0.244312
Up
KCNIP1
1
0
0
0.219995
Up
CD6
1
0
0
0.22832
Up
FCRL3
1
0
0
0.241062
Up
SERTAD4
1
0
0
0.257531
Up
PRF1
1
0
0
0.222162
Up
C1QTNF9
1
0
0
0.226548
Up
OPCML
1
0
0
0.215756
Up
ESM1
1
0
0
0.213551
Up
CD40LG
1
0
0
0.240053
Up
S1PR5
1
0
0
0.24224
Up
AGTR2
1
0
0
0.259256
Up
NPPB
1
0
0
0.211726
Up
SCG5
1
0
0
0.238721
Up
PDE5A
1
0
0
0.243548
Up
RYR3
1
0
0
0.274755
Up
RASEF
1
0
0
0.274755
Up
PODXL2
1
0
0
0.213106
Up
OGN
1
0
0
0.226548
Up
PLCH2
1
0
0
0.238721
Up
SCG2
1
0
0
0.267704
Up
P3H2
1
0
0
0.207132
Up
C12orf75
1
0
0
0.217608
Up
ACE
1
0
0
0.241159
Up
GNA14
1
0
0
0.217608
Up
HDC
1
0
0
0.216614
Up
CMA1
1
0
0
0.226713
Up
CEACAM3
1
0
0
0.265519
Down
PCLAF
817
0.135529
4.95E+08
0.365547
Down
CFTR
800
0.168404
4.5E+08
0.378823
Down
TK1
188
0.034997
43,663,230
0.331089
Down
ECT2
164
0.020509
39,431,940
0.325989
Down
FKBP5
157
0.028064
15,963,868
0.346288
Down
ANLN
153
0.021564
38,168,832
0.325066
Down
ATP2A2
148
0.027131
19,656,040
0.363859
Down
BCL6
142
0.022279
29,419,916
0.314181
Down
TOP2A
132
0.018571
16,838,266
0.361426
Down
ZBTB16
132
0.025165
14,500,206
0.349976
Down
S100A9
124
0.01355
11,186,464
0.352219
Down
CEP55
123
0.019583
21,505,878
0.316891
Down
BLM
108
0.014259
18,458,556
0.321945
Down
AGTR1
100
0.019518
14,083,216
0.313564
Down
SAMHD1
94
0.011463
12,340,270
0.337357
Down
S100A8
88
0.011637
8,662,548
0.361486
Down
GRAP2
86
0.011721
16,819,438
0.305936
Down
CBS
83
0.011248
20,466,334
0.301591
Down
SOCS3
83
0.011071
9,067,820
0.324888
Down
GFI1B
80
0.011791
21,469,034
0.299012
Down
APOB
78
0.014102
9,290,092
0.319133
Down
PCK1
77
0.004102
12,732,476
0.305408
Down
MARK3
76
0.008497
19,265,788
0.304512
Down
HMOX2
75
0.011098
15,258,770
0.312053
Down
PCNT
74
0.011297
9,190,430
0.312261
Down
PIK3C2A
69
0.005568
8,768,122
0.313053
Down
KIF14
69
0.01035
12,506,564
0.304668
Down
WASF1
67
0.009478
18,219,554
0.29633
Down
ARNTL
65
0.00974
19,494,854
0.295526
Down
ALOX5
65
0.010921
7,343,824
0.306424
Down
MCM10
64
0.006773
8,807,396
0.306438
Down
THBS1
64
0.008915
6,203,038
0.312694
Down
VSIG4
64
0.010353
10,534,518
0.302037
Down
WWC1
64
0.007241
14,604,594
0.301647
Down
MELK
63
0.008554
18,629,232
0.283953
Down
P2RY12
63
0.008962
9,063,088
0.286641
Down
PPL
62
0.007851
16,137,388
0.297313
Down
MYBL2
59
0.006189
16,766,208
0.291964
Down
FAM107A
59
0.006172
12,510,302
0.289688
Down
GRIP1
58
0.008069
3,384,760
0.320055
Down
ELF3
56
0.004945
7,205,142
0.309118
Down
PALLD
55
0.00509
15,467,012
0.293274
Down
CTH
54
0.007754
5,179,060
0.296908
Down
EIF4EBP1
53
0.005367
9,748,852
0.303946
Down
KNG1
53
0.007017
4,204,766
0.304243
Down
GLUL
51
0.00728
10,616,752
0.30116
Down
SLC2A1
51
0.004557
8,307,646
0.303974
Down
HP
51
0.006741
3,398,298
0.314741
Down
RPGR
50
0.004441
10,097,488
0.29384
Down
TLR2
50
0.00754
8,322,330
0.294595
Down
GRB7
49
0.004147
4,832,846
0.308113
Down
PPEF1
49
0.001983
3,327,232
0.298276
Down
TXNRD1
49
0.00627
2,860,122
0.328561
Down
NAMPT
48
0.005035
10,420,710
0.290538
Down
BMP7
47
0.007622
4,442,306
0.286981
Down
CA14
47
0.005218
4,884,858
0.279189
Down
CCR1
46
0.008305
11,217,842
0.27812
Down
CDC45
45
0.004479
3,231,162
0.30618
Down
ARG1
45
0.004931
2,347,856
0.32381
Down
SPC24
43
0.005356
6,589,710
0.294834
Down
FGR
43
0.003434
3,020,950
0.303452
Down
KIF5C
42
0.004876
2,365,086
0.319586
Down
IL1R2
42
0.006825
9,489,620
0.289265
Down
SERPINA3
42
0.005518
4,047,110
0.293168
Down
DEPDC1B
42
0.002978
9,632,346
0.260838
Down
SLC4A7
41
0.006314
2,752,106
0.316232
Down
SERPINA5
41
0.003604
13,750,404
0.273206
Down
MPP3
40
0.008262
9,328,402
0.297003
Down
NCEH1
40
0.009405
3,554,728
0.304214
Down
SLC1A1
38
0.008678
2,278,154
0.320573
Down
CLSPN
38
0.003668
3,801,300
0.294343
Down
BCAT1
38
0.0052
9,066,238
0.269657
Down
MYH6
38
0.005049
1,731,786
0.308578
Down
IL20RA
37
0.005252
8,769,348
0.267901
Down
HOOK1
37
0.005558
7,195,380
0.279177
Down
FLT3
37
0.002948
1,938,440
0.292408
Down
ADAMTS4
37
0.005524
2,338,476
0.307519
Down
CAMSAP3
36
0.003339
4,795,892
0.29578
Down
PLA2G2A
35
0.003637
1,686,594
0.300565
Down
FOSL1
34
0.004151
10,955,318
0.269402
Down
NQO1
34
0.001945
5,351,260
0.289201
Down
ELANE
34
0.005024
2,289,646
0.302834
Down
KCND3
34
0.002555
9,153,178
0.28203
Down
EPN3
34
0.005073
7,423,282
0.280302
Down
GPR183
34
0.003642
4,243,792
0.256783
Down
CD109
34
0.006381
3,655,940
0.303565
Down
TUBA3E
34
0.003459
6,711,886
0.289048
Down
TGFBR3
33
0.005143
1,983,082
0.267386
Down
NID1
33
0.004536
1,702,236
0.311503
Down
STEAP3
33
0.004665
2,788,716
0.285365
Down
AMD1
32
0.005714
3,154,782
0.29099
Down
EDNRB
31
0.003092
7,273,156
0.265368
Down
IL17RB
31
0.004227
6,381,040
0.261527
Down
SLC19A2
30
0.004653
2,505,974
0.281302
Down
SLC22A16
30
0.004545
3,831,872
0.240618
Down
PHACTR3
29
0.002193
6,417,862
0.280976
Down
LAPTM5
29
0.003298
2,735,158
0.274317
Down
ANGPTL4
29
0.003467
1,447,446
0.325163
Down
PPM1E
29
0.002894
5,733,032
0.270427
Down
E2F2
28
0.002816
5,508,320
0.28041
Down
SERPINE1
28
0.001474
2,497,574
0.271302
Down
ACPP
28
0.003084
2,749,550
0.291223
Down
KRT7
28
0.002861
1,288,592
0.315774
Down
SERPINB8
28
0.002944
3,167,812
0.28186
Down
FREM2
28
0.003954
3,395,758
0.276661
Down
RNF157
28
0.002172
6,196,626
0.265551
Down
PPIP5K2
28
0.003886
8,572,108
0.270014
Down
F8
27
0.002839
4,879,016
0.274836
Down
TUBAL3
27
0.002055
1,052,840
0.318915
Down
ELL2
26
0.003971
6,281,508
0.255859
Down
GRB14
25
0.002326
3,092,024
0.28378
Down
IRAK3
25
0.00257
6,900,170
0.265897
Down
MANEA
25
0.004508
5,075,608
0.263869
Down
CLEC7A
25
0.004246
4,293,212
0.277095
Down
KLF10
24
0.001607
3,013,994
0.281339
Down
GNMT
24
0.00165
3,015,768
0.269136
Down
ART3
24
0.002904
2,401,360
0.255748
Down
LRRC8E
24
0.003739
4,188,308
0.288665
Down
SLA
23
0.001714
1,003,510
0.289329
Down
CLEC4G
23
0.002667
2,376,260
0.277495
Down
TUBB4A
5
1.28E−04
181,440
0.250652
Down
CD38
4
0
0
0.268176
Down
FCGR3A
4
1.01E−04
73,756
0.268385
Down
F5
3
8.10E−07
708
0.248641
Down
EHF
2
7.11E−06
4180
0.254842
Down
KIAA1549
2
4.16E−04
193,604
0.261391
Down
S100A3
2
1.84E−05
45,822
0.254376
Down
ADH1B
2
3.40E−05
28,184
0.233546
Down
PAPSS2
2
1.05E−05
8726
0.251868
Down
PTX3
1
0
0
0.19143
Down
IL15RA
1
0
0
0.234199
Down
EDN1
1
0
0
0.209723
Down
SERPINF2
1
0
0
0.232451
Down
ZNF366
1
0
0
0.282018
Down
ACR
1
0
0
0.214588
Down
MATN3
1
0
0
0.222881
Down
CNR1
1
0
0
0.216205
Down
LBP
1
0
0
0.240053
Down
ALOX5AP
1
0
0
0.23456
Down
SCGN
1
0
0
0.23353
Down
MAMDC2
1
0
0
0.248745
Down
CDKL5
1
0
0
0.219891
Down
CENPM
1
0
0
0.231833
Down
KCNIP2
1
0
0
0.219995
Down
CPM
1
0
0
0.24533
Down
GPSM2
1
0
0
0.245855
Down
LSAMP
1
0
0
0.215756
Down
KCNK3
1
0
0
0.219353
Down
ALOX15B
1
0
0
0.234981
Down
ST6GALNAC3
1
0
0
0.233263
Down
GPRC5A
1
0
0
0.274755
Down
SLC31A2
1
0
0
0.215287
Down
MARVELD2
1
0
0
0.218671
Down
SNTG2
1
0
0
0.229
Down
TRHDE
1
0
0
0.208786
Down
SIGLEC7
1
0
0
0.245229
Down
SMTNL2
1
0
0
0.265519
Down
ANXA3
1
0
0
0.274755
Down
F13A1
1
0
0
0.248745
Down
ANKRD7
1
0
0
0.233438
Down
KCNS2
1
0
0
0.219721
Down
SIGLEC9
1
0
0
0.227565
Down
SIGLEC10
1
0
0
0.282018
Down
C20orf197
1
0
0
0.282018
Down
SCGB1D2
1
0
0
0.226548
Down
IL1RL1
1
0
0
0.21698
Down
PLIN2
1
0
0
0.241935
Down
CD163
1
0
0
0.239403
Down
HPR
1
0
0
0.240053
Additionally, two significant modules, including module 1 (10 nodes and 24 edges) and module 2 (5 nodes and 10 edges) (Fig. 3B) and module 3 (55 nodes and 115 edges), were acquired by PEWCC1 plug-in (Fig. 3C). Furthermore, GO terms and REACTOME pathways were significantly enriched by module 1, including adaptive immune system, immunoregulatory interactions between a lymphoid and a non-lymphoid cell, hemostasis, biological adhesion and regulation of immune system process. Meanwhile, the nodes in module 2 were significantly enriched in GO terms and REACTOME pathways, including neutrophil degranulation and secretion.

Target gene—miRNA regulatory network construction

Associations between 2063 miRNAs and their 319 target genes were collected from the target gene—miRNA regulatory network (Fig. 4). MiRNAs of hsa-mir-4533, hsa-mir-548ac, hsa-mir-548i, hsa-mir-5585-3p, hsa-mir-6750-3p, hsa-mir-200c-3p, hsa-mir-1273 g-3p, hsa-mir-1244, hsa-mir-4789-3p and hsa-mir-766-3p, which exhibited a high degree of interaction, were selected from this network. Furthermore, the results also showed that FSCN1 was the target of hsa-mir-4533, ESR1 was the target of hsa-mir-548ac, TMEM30B was the target of hsa-mir-548i, SCN2B was the target of hsa-mir-5585-3p, CENPA was the target of hsa-mir-6750-3p, FKBP5 was the target of hsa-mir-200c-3p, PCLAF was the target of hsa-mir-1273g-3p, CEP55 was the target of hsa-mir-1244, ATP2A2 was the target of hsa-mir-4789-3p and TK1 was the target of hsa-mir-766-3p, and are listed in Table 5.
Table 5
miRNA—target gene and TF—target gene interaction
Regulation
Target Genes
Degree
MicroRNA
Regulation
Target Genes
Degree
TF
Up
FSCN1
99
hsa-mir-4533
Up
FSCN1
62
ESRRA
Up
ESR1
72
hsa-mir-548ac
Up
APOA1
48
RERE
Up
TMEM30B
64
hsa-mir-548i
Up
COL1A1
21
HMG20B
Up
SCN2B
46
hsa-mir-5585-3p
Up
HBB
16
THRAP3
Up
CENPA
35
hsa-mir-6750-3p
Up
LCK
15
ATF1
Up
APOA1
22
hsa-mir-6722-5p
Up
FOXS1
14
YBX1
Up
PPP2R2B
14
hsa-mir-149-3p
Up
CENPA
10
SAP30
Up
TP63
12
hsa-mir-1178-3p
Up
SCN2B
5
RCOR2
Up
PYHIN1
5
hsa-mir-205-3p
Up
TMEM30B
5
ZNF24
Up
APLNR
2
hsa-mir-10b-5p
Up
APLNR
4
FOXJ2
Up
PTN
1
hsa-mir-155-5p
Up
NRG1
2
SUZ12
Up
LCK
1
hsa-mir-335-5p
Up
PTN
2
L3MBTL2
Up
CD247
1
hsa-mir-346
Up
UCHL1
2
MAZ
Down
FKBP5
88
hsa-mir-200c-3p
Up
ESR1
1
EZH2
Down
PCLAF
62
hsa-mir-1273g-3p
Up
ZAP70
1
ZFX
Down
CEP55
57
hsa-mir-1244
Down
SOCS3
48
MXD3
Down
ATP2A2
55
hsa-mir-4789-3p
Down
BCL6
44
ARID4B
Down
TK1
45
hsa-mir-766-3p
Down
FKBP5
43
CBFB
Down
ZBTB16
43
hsa-mir-1976
Down
ANLN
38
TAF7
Down
SAMHD1
26
hsa-mir-3124-3p
Down
ATP2A2
35
CREM
Down
TOP2A
17
hsa-mir-186-5p
Down
CBS
31
IKZF1
Down
BCL6
13
hsa-mir-339-5p
Down
BLM
19
ZNF501
Down
ECT2
13
hsa-mir-132-3p
Down
ECT2
15
KLF16
Down
CFTR
9
hsa-mir-145-5p
Down
CEP55
10
FOSL2
Down
S100A9
7
hsa-mir-4679
Down
GRAP2
10
CEBPD
Down
AGTR1
5
hsa-mir-410-3p
Down
ZBTB16
4
TRIM28
Down
ANLN
5
hsa-mir-503-5p
Down
S100A8
3
STAT3
Down
BLM
3
hsa-mir-193b-3p
Down
S100A9
2
CEBPG
    
Down
AGTR1
1
EZH2

Target gene—TF regulatory network construction

Associations between 330 TFs and their 247 target genes were collected from the target gene—TF regulatory network (Fig. 5). TFs of ESRRA, RERE, HMG20B, THRAP3, ATF1, MXD3, ARID4B, CBFB, TAF7 and CREM, which exhibited a high degree of interaction, were selected from this network. Furthermore, the results also showed that FSCN1 was the target of ESRRA, APOA1 was the target of RERE, COL1A1 was the target of HMG20B, HBB was the target of THRAP3, LCK was the target of ATF1, SOCS3 was the target of MXD3, BCL6 was the target of ARID4B, FKBP5 was the target of CBFB, ANLN was the target of TAF7 and ATP2A2 was the target of CREM, and are listed in Table 5.

Receiver operating characteristic (ROC) curve analysis

First of all, we performed the ROC curve analysis among 10 hub genes based on the GSE141910. The results showed that ESR1, PYHIN1, PPP2R2B, LCK, TP63, PCLAF, CFTR, TK1, ECT2 and FKBP5 achieved an AUC value of > 0.7, demonstrating that these ten genes have high sensitivity and specificity for HF, suggesting they can be served as biomarkers for the diagnosis of HF (Fig. 6).

RT-PCR analysis

RT-PCR was used to validate the hub genes between normal and HF cell lines. The results suggested that the mRNA expression level of ESR1, PYHIN1, PPP2R2B, LCK and TP63 were significantly increased in HF compared with that in normal, while PCLAF, CFTR, TK1, ECT2 and FKBP5 were significantly decreased in HF compared with that in normal and are shown in Fig. 7.

Identification of candidate small molecules

In the present study docking simulations are performed to spot the active site and foremost interactions accountable for complex stability with the receptor binding sites. In heart failure recognized over expressed genes and their proteins of x-ray crystallographic structure are chosen from PDB for docking studies. Most generally, medications containing benzothiadiazine ring hydrochlorothiazide are used in heart failure either alone or in conjunction with other drugs, based on this the molecules containing heterocyclic ring of benzothiadiazine are designed and hydrochlorothiazide is uses as a reference standard. Docking experiments using Sybyl-X 2.1.1. drug design perpetual software were used on the designed molecules. Docking studies were performed in order to understand the biding interaction of standard hydrochlorothiazide and designed molecules on over expressed protein. The X- RAY crystallographic structure of one proteins from each over expressed genes of ESR1, LCK, PPP2R2B, PYHIN1, TP63 and their co-crystallised protein of PDB code 4PXM, 1KSW, 2HV7, 3VD8 and 6RU6 respectively were selected for the docking studies to identify and predict the potential molecule based on the binding score with the protein and successful in heart failure. For the docking tests, a total of 34 molecules were built and the molecule with binding score greater than 5 is believed to be good. The designed molecules obtained docking score of 5 to 7 were HIM10, HTZ5, HIM6, HTZ31, HIM3, HIM14, HIM1, HIM7 and HIM11, HIM16, HTZ9, HIM17, HIM12, HTZ12, HIM6, HTZ7, HIM10, HTZ3 and HIM8, HTZ9, HIM6, HIM4, HIM13, HTZ16, HIM9, HIM7, HTZ5, HIM16, HTZ7, HIM10, HIM5, HIM12, HIM15, HTZ12, HIM3, HIM14 and HIM14, HIM6, HIM17, HTZ7, HIM10, HIM1, HTZ9, HIM3, HIM16, HIM15, HIM8, HIM9, HIM7, HTZ10, HTZ3, HTZ5, HTZ1, HIM13, HTZ4, HIM11, HTZ12, HTZ14, HIM2 and HIM7, HTZ13, HTZ5, HIM15, HIM12, HIM6, HTZ11, HIM14, HTZ9, HIM11, HIM13, HIM9, HIM8, HIM10, HIM1, HIM5, HIM4, HTZ12, HIM2, HIM17, HIM3, HTZ1, HTZ8, HIM3, HTZ14, HTZ3 with proteins 4PXM and, 1KSW and 2HV7 and 3VD8 and 6RUR respectively (Fig. 8). The molecules obtained binding score of less than 5 were HTZ13, HTZ12, HTZ10, HIM3, HIM15, HIM16, HIM13, HIM8, HTZ16, HIM2, HIM4, HIM17, HTZ17, HIM11, HTZ5, HTZ3, HIM9, HTZ15, HTZ5, HTZ9, HTZ11, HIM5, HTZ8 and HTZ14, HIM14, HTZ13, HIM13, HTZ16, HIM2, HIM3, HTZ10, HIM7, HIM1, HTZ1, HTZ4, HIM8, HIM5, HTZ2, HIM9, HTZ5, HTZ15, HTZ3, HIM4, HIM15, HTZ17, HTZ8, HTZ11 and HTZ14, HIM2, HIM1, HTZ11, HIM17, HTZ13, HTZ4, HTZ2, HIM3, HTZ15, HTZ8, HTZ17, HTZ1, HTZ3 and HTZ8, HIM4, HTZ16, HTZ15, HIM5, HTZ11, HTZ13, HIM3, HTZ17, HTZ2 and HTZ7, HTZ4, HTZ2, HTZ17, HTZ15 with proteins 4PXM and, 1KSW and 2HV7 and 3VD8 and 6RUR respectively. The molecules obtained very less binding score are HTZ1, HIM12, HTZ2, HTZ4 with protein 4PXM and the standard hydrochlorothiazide (HTZ) obtained less binding score with all proteins, the values are depicted in Table 6.
Table 6
Docking results of Designed Molecules on Over Expressed Proteins
Sl. No/
Code
Over expressed gene: ESR1
Over expressed gene: LCK
Over expressed gene: PPP2R2B
Over expressed gene: PYHIN 1
Over expressed gene: TP63
PDB: 4PXM
PDB:1KSW
PDB: 2HV7
PDB: 3VD8
PDB: 6RU6
Total Score
Crash
(-Ve)
Polar
Total Score
Crash
(-Ve)
Polar
Total Score
Crash
(−Ve)
Polar
Total Score
Crash
(−Ve)
Polar
Total Score
Crash
(−Ve)
Polar
HIM1
5.097
− 4.375
0.114
4.258
− 1.555
1.784
4.904
− 0.870
0.004
5.794
− 0.514
3.828
5.770
− 1.440
2.231
HIM2
4.057
− 6.172
0.039
4.624
− 0.997
1.959
4.906
− 0.468
1.517
5.052
− 0.780
3.415
5.414
− 1.529
2.444
HIM3
5.353
− 5.309
0.161
4.578
− 1.492
1.790
5.042
− 1.845
1.073
5.680
− 0.804
3.956
5.350
− 1.170
2.316
HIM4
3.976
− 5.132
0.167
3.839
− 1.635
1.196
6.328
− 1.172
1.128
4.966
− 0.666
1.818
5.627
− 1.416
2.320
HIM5
2.707
− 7.759
0.179
4.067
− 0.997
1.987
5.254
− 0.674
1.618
4.563
− 1.068
2.935
5.698
− 1.240
2.485
HIM6
5.948
− 3.902
1.796
5.229
− 0.707
3.656
6.766
− 1.424
1.858
6.670
− 0.941
5.519
6.218
− 1.468
2.578
HIM7
5.019
− 7.055
0.203
4.382
− 2.443
3.329
6.028
− 0.629
1.660
5.374
− 1.876
0.670
6.627
− 1.484
2.579
HIM8
4.429
− 3.983
0.344
4.150
− 4.382
4.210
6.794
− 1.279
1.129
5.468
− 0.769
3.444
5.842
− 2.088
2.272
HIM9
3.722
− 5.956
0.197
4.051
− 2.006
0.002
6.116
− 0.597
1.717
5.407
− 0.565
1.370
5.877
− 2.054
0.792
HIM10
6.771
− 3.977
1.836
5.176
− 3.512
4.023
5.332
− 1.378
3.349
6.071
− 0.923
3.854
5.825
− 0.966
3.672
HIM11
3.775
− 6.079
0.898
6.998
− 2.086
3.842
8.678
− 1.065
2.876
5.087
− 0.881
1.854
5.948
− 1.015
1.202
HIM12
0.190
− 8.149
0.022
5.302
− 2.305
3.475
5.227
− 1.636
0.710
7.322
− 1.128
4.099
6.237
− 2.562
2.171
HIM13
4.523
− 4.537
0.014
4.840
− 0.664
3.305
6.181
− 2.966
3.523
5.281
− 0.503
3.981
5.905
− 1.136
2.218
HIM14
5.247
− 3.183
0.000
4.888
− 1.296
2.563
5.037
− 0.377
1.647
7.057
− 0.799
4.256
6.116
− 1.366
2.438
HIM15
4.633
− 4.173
0.180
3.756
− 0.710
2.072
5.188
− 1.559
1.143
5.570
− 1.125
3.718
6.238
− 1.708
2.443
HIM16
4.588
− 2.883
0.000
6.027
− 1.099
3.903
5.606
− 0.987
4.197
5.661
− 0.926
2.751
7.263
− 1.533
4.212
HIM17
3.944
− 4.806
0.236
5.329
− 0.590
2.798
4.830
− 1.682
1.617
6.234
− 0.830
3.912
5.366
− 1.257
3.022
HTZ1
0.593
− 7.518
0
4.221
− 0.692
1.975
3.993
− 0.539
0.003
5.284
− 0.566
1.432
5.227
− 1.045
0.903
HTZ2
− 1.770
− 8.477
0.000
4.055
− 1.438
2.877
4.388
− 1.665
1.170
4.100
− 0.546
3.017
4.563
− 0.976
1.266
HTZ3
4.649
− 5.870
0.148
5.104
− 0.861
3.922
4.243
− 1.539
1.933
5.304
− 1.370
1.398
5.138
− 1.217
1.930
HTZ4
− 3.169
− 12.002
0.482
4.173
− 1.898
1.864
4.654
− 1.627
1.128
5.163
− 0.745
1.304
5.084
− 1.143
1.018
HTZ5
4.021
− 12.325
0.246
3.215
− 1.481
4.232
3.256
− 6.374
2.317
2.382
− 5.263
1.238
4.623
− 0.951
1.280
HTZ6
6.605
− 3.866
1.6487
4.004
− 1.104
2.851
5.834
− 1.310
3.111
5.286
− 1.530
1.436
6.336
− 2.326
3.781
HTZ7
4.977
− 5.434
0.655
5.197
− 2.040
3.250
5.352
− 1.371
1.172
6.138
− 1.734
1.627
4.908
− 1.057
1.335
HTZ8
1.025
− 8.223
0.000
3.549
− 1.310
2.403
4.024
− 3.825
2.440
4.980
− 0.593
1.474
5.164
− 1.291
0.999
HTZ9
3.386
− 7.041
0.194
5.567
− 1.622
3.057
6.792
− 2.581
1.088
5.794
− 0.683
1.774
6.053
− 1.408
1.037
HTZ10
4.744
− 5.463
0.837
4.520
− 2.469
3
7.758
− 1.518
3.765
5.345
− 1.133
3.661
7.507
− 2.080
4.086
HTZ11
2.991
− 6.177
0
3.453
− 0.721
1.266
4.841
− 1.738
0.045
4.368
− 0.805
1.074
6.176
− 1.380
1.511
HTZ12
4.810
− 6.157
0.275
5.296
− 2.814
3.605
5.138
− 1.840
2.189
5.083
− 0.870
1.536
5.592
− 1.321
1.525
HTZ13
4.868
− 3.837
0
4.863
− 0.535
2.405
4.656
− 0.681
3.152
4.246
− 2.335
0.529
6.404
− 0.975
2.954
HTZ14
5.646
− 3.473
0
4.948
− 0.801
2.324
4.953
− 1.672
1.066
5.058
− 1.174
1.121
5.114
− 1.299
1.296
HTZ15
3.428
− 4.957
0.348
3.949
− 0.614
1.873
4.049
− 0.787
1.224
4.796
− 1.066
1.489
3.510
− 0.607
0.461
HTZ16
4.227
− 4.787
0.298
4.654
− 1.534
2.096
6.143
− 1.204
2.879
4.854
− 0.994
1.564
7.102
− 0.917
3.001
HTZ17
3.784
− 5.018
0.380
3.661
− 0.897
1.676
4.016
− 1.179
1.384
4.039
− 0.569
2.706
4.256
− 1.040
1.236
HTZ
STD
4.722
− 1.084
1.063
3.319
− 0.890
3.033
3.564
− 0.272
2.367
3.394
− 0.882
1.169
4.237
− 0.801
1.855

Discussion

HF is the most prevalent form of cardiovascular disease among the elderly. A complete studies of HF, comprising pathogenic factors, pathological processes, clinical manifestations, early clinical diagnosis, clinical prevention, and drug therapy targets urgency to be consistently analyzed. In the present investigation, bioinformatics analysis was engaged to explore HF biomarkers and the pathological processes in myocardial tissues, acquired from HF groups and non heart failure groups. We analyzed GSE141910 expression profiling by high throughput sequencing obtained 881 different genes between HF groups and non heart failure groups, 442 up regulated and 439 down regulated genes. HBA2 and HBA1 have a key role in hypertension [41], but these genes might be linked with development HF. SFRP4 was linked with progression of myocardial ischemia [42]. Emmens et al. [43] and Broch et al. [44] found that PENK (proenkephalin) and IL1RL1 were up regulated in HF. ALOX15B has lipid accumulation and inflammation activity and is highly expressed in atherosclerosis [45]. Studies have shown that expression of MYH6 was associated with hypertrophic cardiomyopathy [46].
In functional enrichment analysis, some genes involved with regulation of cardiovascular system processes were enriched in HF. Liu et al. [47], Kosugi et al. [48], McMacken et al. [49], Pan and Zhang [50], Li et al. [51] and Jiang et al. [52] presented that expression of HLA-DQA1, KDM5D, UCHL1, SAA1, ARG1 and LYVE1 were associated with progression of cardiomyopathy. Hou et al. [53] and Olesen et al. [54] demonstrated that DACT2 and KCND3 were found to be substantially related to atrial fibrillation. Ge and Concannon [55], Ferjeni et al. [56], Anquetil et al. [57], Glawe et al. [58], Kawabata et al. [59], Li et al. [60], Buraczynska et al. [61], Amini et al. [62], Yang et al. [63], Du Toit et al. [64], Hirose et al. [65], Zhang et al. [66], Griffin et al. [67], Zouidi et al. [68], Trombetta et al. [69], Alharbi et al. [70], Ikarashi et al. [71], Dharmadhikari et al. [72], Sutton et al. [73] and Deng et al. [74] reported that UBASH3A, ZAP70, IDO1, ITGAL (integrin subunit alpha L). ITGB7, RASGRP1, CNR1, SLC2A1, SLC11A1, GPR84, SSTR5, KCNB1, GLUL (glutamate-ammonia ligase), BANK1, CACNA1E, LGR5, AQP3, SIGLEC7, SSTR2 and DNER (delta/notch like EGF repeat containing) could be an index for diabetes, but these genes might be responsible for progression of HF. Experiments show that expression of FAP (fibroblast activation protein alpha) [75], THBS4 [76], CD27 [77], LEF1 [78], CTHRC1 [79], ESR1 [80], CXCL9 [81], SERPINA3 [82], TRPC4 [83], F13A1 [84], PIK3C2A [85], KCNIP2 [86] and GPR4 [87] contributed to myocardial infarction. MFAP4 [88], ALOX15 [89], COL1A1 [90], APOA1 [91], PDE5A [92], CX3CR1 [93], THY1 [94], GREM1 [95], FMOD (fibromodulin) [96], NPPA (natriuretic peptide A) [97], LTBP2 [98], LUM (lumican) [99], IL34 [100], NRG1 [101], CXCL14 [102], CXCL10 [103], ACE (angiotensin I converting enzyme) [104], CFTR (ystic fibrosis transmembrane conductance regulator) [105], S100A8 [106], S100A9 [106], HP (haptoglobin) [107], AGTR1 [108], ATP2A2 [109], IL10 [110], EDN1 [111], TLR2 [112], MCEMP1 [113], TPO (thyroid peroxidase) [114], CD163 [115], IL18R1 [116], KCNA7 [117] and CALCRL (calcitonin receptor like receptor) [118] have an important role in HF. Li et al. [119], Deckx et al. [120], Ichihara et al. [121] and Paik et al. [122] showed that the SERPINE2, OGN (osteoglycin), AGTR2 and WNT10B promoted cardiac interstitial fibrosis. Cai et al. [123], Mo et al. [124], Sun et al. [125], Martinelli et al. [126], Zhao et al. [127], Assimes et al. [128] and Piechota et al. [129] showed that CCR7, FCN1, ESM1, F8 (coagulation factor VIII), C1QTNF1, ALOX5 and MSR1 were an important target gene for coronary artery disease. STAB2 have been suggested to be associated with venous thromboembolic disease [130]. Genes such as COMP (cartilage oligomeric matrix protein) [131], CHI3L1 [132], PLA2G2A [133], P2RY12 [134], CR1 [135], HPSE (heparanase) [136], PTX3 [137] and SERPINE1 [138] were related to atherosclerosis. CCDC80 [139], CMA1 [140], MDK (midkine) [141], GNA14 [142], SCG2 [143], NPPB (natriuretic peptide B) [144], FGF10 [145], ARNTL (aryl hydrocarbon receptor nuclear translocator like) [146], WNK3 [147], EDNRB (endothelin receptor type B) [148], THBS1 [149], SELE (selectin E) [150], SLC4A7 [151], AQP4 [152] and KCNK3 [153] are thought to be responsible for progression of hypertension, but these genes might to be associated with progression of HF. CNTNAP2 [154], GLI2 [155], DPT (dermatopontin) [156], AEBP1 [157], ITIH5 [158], CXCL11 [159], GDNF (glial cell derived neurotrophic factor) [160], MCHR1 [161], FLT3 [162], ELANE (elastase, neutrophil expressed) [163], OSMR (oncostatin M receptor) [164] and IL15RA [165] are involved in development of obesity, but these genes might be key for progression of HF. CTSG (cathepsin G) is a protein coding gene plays important roles in aortic aneurysms [166]. Evidence from Safa et al. [167], Chen et al. [168], Zhou et al. [169], Hu et al. [170], Lou et al. [171], Zhang et al. [172] and Chen et al. [173] study indicated that the expression of CCL22, CCR1, FPR1, KNG1, CRISPLD2, CD38 and GPRC5A were linked with progression of ischemic heart disease. Li et al. [174] showed that STEAP3 expression can be associated with cardiac hypertrophy progression.
The HiPPIE interactome database was used to construct the PPI network, and modules analysis was performed. We finally screened out up regulated hub genes and down regulated hub genes, including ESR1, PYHIN1, PPP2R2B, LCK, TP63, PCLAF, CD247, CD2, CD5, CD48, CFTR, TK1, ECT2, FKBP5, S100A9 and S100A8 from the PPI network and its modules. TP63 might serve as a potential prognostic factor in cardiomyopathy [175]. The expression of FKBP5 is related to the progression of coronary artery disease [176]. CD247 plays a central role in hypertension [177], but this gene might be involved in the HF. PYHIN1, PPP2R2B, LCK (LCK proto-oncogene, Src family tyrosine kinase), PCLAF (PCNA clamp associated factor), TK1, ECT2, CD2, CD5 and CD48 might be the novel biomarker for HF.
The miRNet database and NetworkAnalyst database were used to construct the target gene—miRNA regulatory network and target gene—TF regulatory network. We finally screened out target genes, miRNA, TFs, including FSCN1, ESR1, TMEM30B, SCN2B, CENPA, FKBP5, PCLAF, CEP55, ATP2A2, TK1, APOA1, COL1A1, HBB, LCK, SOCS3, BCL6, ANLN, hsa-mir-4533, hsa-mir-548ac, hsa-mir-548i, hsa-mir-5585-3p, hsa-mir-6750-3p, hsa-mir-200c-3p, hsa-mir-1273 g-3p, hsa-mir-1244, hsa-mir-4789-3p, hsa-mir-766-3p, ESRRA, RERE, HMG20B, THRAP3, ATF1, MXD3, ARID4B, CBFB, TAF7 and CREM from the target gene—miRNA regulatory network and target gene—TF regulatory network. SCN2B [178] and SOCS3 [179] are considered as a markers for HF and might be a new therapeutic target. BCL6 levels are correlated with disease severity in patients with atherosclerosis [180]. A previous study showed that hsa-mir-1273 g-3p [181], hsa-mir-4789-3p [182] and ATF1 [183] could involved in hypertension, but these markers might be responsible for progression of HF. hsa-miR-518f, was demonstrated to be associated with cardiomyopathy [184]. An evidence demonstrating a role for ESRRA (estrogen related receptor alpha) [185] and THRAP3 [186] in diabetes, but these genes might be liable for development of HF. FSCN1, TMEM30B, CENPA (centromere protein A), CEP55, HBB (hemoglobin subunit beta), ANLN (anillin actin binding protein), hsa-mir-4533, hsa-mir-548ac, hsa-mir-548i, hsa-mir-5585-3p, hsa-mir-6750-3p, hsa-mir-200c-3p, hsa-mir-1244, RERE(arginine-glutamic acid dipeptide repeats), HMG20B, MXD3, ARID4B, CBFB (core-binding factor subunit beta), TAF7 and CREM (cAMP response element modulator) might be the novel biomarker for HF.
The molecules HIM6, HIM10 obtained good binding score of more 5 to 6.999 with all proteins and the molecules HIM11, HIM12, HIM14, HTZ9, HTZ10 and HTZ12 obtained binding score above 5 and less than 9 with PDB protein code of 2HV7, 3VD8 and 6RUR respectively. The molecule HIM11 obtained highest binding score of 8.678 with 2HV7 and its interaction with amino acids are molecule HIM11 (Fig. 9) has obtained with a high binding score with PDB protein 2HV7, the interactions of molecule is the C6 side chin acyl carbonyl C=O formed hydrogen bond interaction with amino acid GLN-207 with bond length 1.92 A° and 3’ N–H group of imidazole ring formed hydrogen bond interaction with VAL-305 with bond length 2.36 A° respectively. It also formed other interactions of carbon hydrogen bond of –CH3 group of carboxylate at C6 with PRO-304 and amide-pi stacked and pi–pi stacked interaction of electrons of aromatic ring A with ALA-204 and ring C with HIS-155 and HIS-308. Molecule formed pi-alkyl interaction of ring B with PRO-304 and all interactions with amino acids and bond length are depicted by 3D and 2D figures (Fig. 10 and Fig. 11).

Conclusions

The present investigation aimed at characterizing the expression profiling by high throughput sequencing of the HF patients. Our bioinformatics analyses revealed key gene signatures as candidate biomarkers in HF. Hub genes (ESR1, PYHIN1, PPP2R2B, LCK, TP63, PCLAF, CFTR, TK1, ECT2 and FKBP5) were diagnosed as an essential genetic factors in HF. In general, DEGs linked with HF genes, including already known markers of HF and other HF related diseases, and novel biomarkers, were diagnosed. Potential implicated miRNAs and TFs were also diagnosed. The diagnosed hub genes might represent candidate diagnostic and prognostic biomarkers, and therapeutic targets. The current investigation reported novel genes and signaling pathways in HF, and further investigation is required.

Acknowledgements

I thank Michael Patrick Morley, Perelman School of Medicine at the University of Pennsylvania, Penn Cardiovascular Institute, Philadelphia, USA, very much, the author who deposited their profiling by high throughput sequencing dataset GSE141910, into the public GEO database.

Declarations

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.
No informed consent because this study does not contain human or animals participants.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
39.
Zurück zum Zitat Okawa T, Aramaki Y, Yamamoto M, Kobayashi T, Fukumoto S, Toyoda Y, Henta T, Hata A, Ikeda S, Kaneko M, et al. Design, synthesis, and evaluation of the highly selective and potent g-protein-coupled receptor Kinase 2 (GRK2) inhibitor for the potential treatment of heart failure. J Med Chem. 2017;60(16):6942–90. https://doi.org/10.1021/acs.jmedchem.7b00443.CrossRefPubMed Okawa T, Aramaki Y, Yamamoto M, Kobayashi T, Fukumoto S, Toyoda Y, Henta T, Hata A, Ikeda S, Kaneko M, et al. Design, synthesis, and evaluation of the highly selective and potent g-protein-coupled receptor Kinase 2 (GRK2) inhibitor for the potential treatment of heart failure. J Med Chem. 2017;60(16):6942–90. https://​doi.​org/​10.​1021/​acs.​jmedchem.​7b00443.CrossRefPubMed
47.
Zurück zum Zitat Liu W, Li WM, Sun NL. Relationship between HLA-DQA1 polymorphism and genetic susceptibility to idiopathic dilated cardiomyopathy. Chin Med J (Engl). 2004;117(10):1449–52. Liu W, Li WM, Sun NL. Relationship between HLA-DQA1 polymorphism and genetic susceptibility to idiopathic dilated cardiomyopathy. Chin Med J (Engl). 2004;117(10):1449–52.
52.
Zurück zum Zitat Jiang X, Cui J, Yang C, Song Y, Yuan J, Liu S, Hu F, Yang W, Qiao S. Elevated lymphatic vessel density measured by Lyve-1 expression in areas of replacement fibrosis in the ventricular septum of patients with hypertrophic obstructive cardiomyopathy (HOCM). Heart Vessels. 2020;35(1):78–85. https://doi.org/10.1007/s00380-019-01463-5.CrossRefPubMed Jiang X, Cui J, Yang C, Song Y, Yuan J, Liu S, Hu F, Yang W, Qiao S. Elevated lymphatic vessel density measured by Lyve-1 expression in areas of replacement fibrosis in the ventricular septum of patients with hypertrophic obstructive cardiomyopathy (HOCM). Heart Vessels. 2020;35(1):78–85. https://​doi.​org/​10.​1007/​s00380-019-01463-5.CrossRefPubMed
54.
57.
Zurück zum Zitat Anquetil F, Mondanelli G, Gonzalez N, Rodriguez Calvo T, Zapardiel Gonzalo J, Krogvold L, Dahl-Jørgensen K, Van den Eynde B, Orabona C, Grohmann U, et al. Loss of IDO1 expression from human pancreatic β-cells precedes their destruction during the development of type 1 diabetes. Diabetes. 2018;67(9):1858–66. https://doi.org/10.2337/db17-1281.CrossRefPubMedPubMedCentral Anquetil F, Mondanelli G, Gonzalez N, Rodriguez Calvo T, Zapardiel Gonzalo J, Krogvold L, Dahl-Jørgensen K, Van den Eynde B, Orabona C, Grohmann U, et al. Loss of IDO1 expression from human pancreatic β-cells precedes their destruction during the development of type 1 diabetes. Diabetes. 2018;67(9):1858–66. https://​doi.​org/​10.​2337/​db17-1281.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Kawabata Y, Nishida N, Awata T, Kawasaki E, Imagawa A, Shimada A, Osawa H, Tanaka S, Takahashi K, Nagata M, et al. Genome-wide association study confirming a strong effect of HLA and identifying variants in CSAD/lnc-ITGB7-1 on chromosome 12q13.13 associated with susceptibility to fulminant type 1 diabetes. Diabetes. 2019;68(3):665–75. https://doi.org/10.2337/db18-0314.CrossRefPubMed Kawabata Y, Nishida N, Awata T, Kawasaki E, Imagawa A, Shimada A, Osawa H, Tanaka S, Takahashi K, Nagata M, et al. Genome-wide association study confirming a strong effect of HLA and identifying variants in CSAD/lnc-ITGB7-1 on chromosome 12q13.13 associated with susceptibility to fulminant type 1 diabetes. Diabetes. 2019;68(3):665–75. https://​doi.​org/​10.​2337/​db18-0314.CrossRefPubMed
62.
Zurück zum Zitat Amini S, Javanmardi M, Mokarizadeh A, Maroofi F, Jalali C, Azadi NA, Mohammadi H, Abdi M. Association of HaeIII single nucleotide polymorphisms in the SLC2A1 gene with risk of diabetic nephropathy; evidence from Kurdish patients with type 2 diabetes mellitus. QJM. 2016;109(6):399–404. https://doi.org/10.1093/qjmed/hcv149.CrossRefPubMed Amini S, Javanmardi M, Mokarizadeh A, Maroofi F, Jalali C, Azadi NA, Mohammadi H, Abdi M. Association of HaeIII single nucleotide polymorphisms in the SLC2A1 gene with risk of diabetic nephropathy; evidence from Kurdish patients with type 2 diabetes mellitus. QJM. 2016;109(6):399–404. https://​doi.​org/​10.​1093/​qjmed/​hcv149.CrossRefPubMed
65.
Zurück zum Zitat Hirose H, Yamasaki T, Ogino M, Mizojiri R, Tamura-Okano Y, Yashiro H, Muraki Y, Nakano Y, Sugama J, Hata A, et al. Discovery of novel 5-oxa-2,6-diazaspiro[3.4]oct-6-ene derivatives as potent, selective, and orally available somatostatin receptor subtype 5 (SSTR5) antagonists for treatment of type 2 diabetes mellitus. Bioorg Med Chem. 2017;25(15):4175–93. https://doi.org/10.1016/j.bmc.2017.06.007.CrossRefPubMed Hirose H, Yamasaki T, Ogino M, Mizojiri R, Tamura-Okano Y, Yashiro H, Muraki Y, Nakano Y, Sugama J, Hata A, et al. Discovery of novel 5-oxa-2,6-diazaspiro[3.4]oct-6-ene derivatives as potent, selective, and orally available somatostatin receptor subtype 5 (SSTR5) antagonists for treatment of type 2 diabetes mellitus. Bioorg Med Chem. 2017;25(15):4175–93. https://​doi.​org/​10.​1016/​j.​bmc.​2017.​06.​007.CrossRefPubMed
74.
Zurück zum Zitat Deng Z, Shen J, Ye J, Shu Q, Zhao J, Fang M, Zhang T. Association between single nucleotide polymorphisms of delta/notch-like epidermal growth factor (EGF)-related receptor (DNER) and Delta-like 1 Ligand (DLL 1) with the risk of type 2 diabetes mellitus in a Chinese Han population. Cell Biochem Biophys. 2015;71(1):331–5. https://doi.org/10.1007/s12013-014-0202-3.CrossRefPubMed Deng Z, Shen J, Ye J, Shu Q, Zhao J, Fang M, Zhang T. Association between single nucleotide polymorphisms of delta/notch-like epidermal growth factor (EGF)-related receptor (DNER) and Delta-like 1 Ligand (DLL 1) with the risk of type 2 diabetes mellitus in a Chinese Han population. Cell Biochem Biophys. 2015;71(1):331–5. https://​doi.​org/​10.​1007/​s12013-014-0202-3.CrossRefPubMed
84.
Zurück zum Zitat Ansani L, Marchesini J, Pestelli G, Luisi GA, Scillitani G, Longo G, Milani D, Serino ML, Tisato V, Gemmati D. F13A1 gene variant (V34L) and residual circulating FXIIIA levels predict short- and long-term mortality in acute myocardial infarction after coronary angioplasty. Int J Mol Sci. 2018;19(9):2766. https://doi.org/10.3390/ijms19092766.CrossRefPubMedCentral Ansani L, Marchesini J, Pestelli G, Luisi GA, Scillitani G, Longo G, Milani D, Serino ML, Tisato V, Gemmati D. F13A1 gene variant (V34L) and residual circulating FXIIIA levels predict short- and long-term mortality in acute myocardial infarction after coronary angioplasty. Int J Mol Sci. 2018;19(9):2766. https://​doi.​org/​10.​3390/​ijms19092766.CrossRefPubMedCentral
92.
Zurück zum Zitat Westermann D, Becher PM, Lindner D, Savvatis K, Xia Y, Fröhlich M, Hoffmann S, Schultheiss HP, Tschöpe C. Selective PDE5A inhibition with sildenafil rescues left ventricular dysfunction, inflammatory immune response and cardiac remodeling in angiotensin II-induced heart failure in vivo. Basic Res Cardiol. 2012;107(6):308. https://doi.org/10.1007/s00395-012-0308-y.CrossRefPubMed Westermann D, Becher PM, Lindner D, Savvatis K, Xia Y, Fröhlich M, Hoffmann S, Schultheiss HP, Tschöpe C. Selective PDE5A inhibition with sildenafil rescues left ventricular dysfunction, inflammatory immune response and cardiac remodeling in angiotensin II-induced heart failure in vivo. Basic Res Cardiol. 2012;107(6):308. https://​doi.​org/​10.​1007/​s00395-012-0308-y.CrossRefPubMed
99.
Zurück zum Zitat Engebretsen KV, Lunde IG, Strand ME, Waehre A, Sjaastad I, Marstein HS, Skrbic B, Dahl CP, Askevold ET, Christensen G, et al. Lumican is increased in experimental and clinical heart failure, and its production by cardiac fibroblasts is induced by mechanical and proinflammatory stimuli. FEBS J. 2013;280(10):2382–98. https://doi.org/10.1111/febs.12235.CrossRefPubMed Engebretsen KV, Lunde IG, Strand ME, Waehre A, Sjaastad I, Marstein HS, Skrbic B, Dahl CP, Askevold ET, Christensen G, et al. Lumican is increased in experimental and clinical heart failure, and its production by cardiac fibroblasts is induced by mechanical and proinflammatory stimuli. FEBS J. 2013;280(10):2382–98. https://​doi.​org/​10.​1111/​febs.​12235.CrossRefPubMed
105.
Zurück zum Zitat Meissner A, Yang J, Kroetsch JT, Sauvé M, Dax H, Momen A, Noyan-Ashraf MH, Heximer S, Husain M, et al. Tumor necrosis factor-α-mediated downregulation of the cystic fibrosis transmembrane conductance regulator drives pathological sphingosine-1-phosphate signaling in a mouse model of heart failure. Circulation. 2012;125(22):2739–50. https://doi.org/10.1161/CIRCULATIONAHA.111.047316.CrossRefPubMed Meissner A, Yang J, Kroetsch JT, Sauvé M, Dax H, Momen A, Noyan-Ashraf MH, Heximer S, Husain M, et al. Tumor necrosis factor-α-mediated downregulation of the cystic fibrosis transmembrane conductance regulator drives pathological sphingosine-1-phosphate signaling in a mouse model of heart failure. Circulation. 2012;125(22):2739–50. https://​doi.​org/​10.​1161/​CIRCULATIONAHA.​111.​047316.CrossRefPubMed
108.
Zurück zum Zitat de Denus S, Dubé MP, Fouodjio R, Huynh T, LeBlanc MH, Lepage S, Sheppard R, Giannetti N, Lavoie J, Mansour A, et al. A prospective study of the impact of AGTR1 A1166C on the effects of candesartan in patients with heart failure. Pharmacogenomics. 2018;19(7):599–612. https://doi.org/10.2217/pgs-2018-0004.CrossRefPubMed de Denus S, Dubé MP, Fouodjio R, Huynh T, LeBlanc MH, Lepage S, Sheppard R, Giannetti N, Lavoie J, Mansour A, et al. A prospective study of the impact of AGTR1 A1166C on the effects of candesartan in patients with heart failure. Pharmacogenomics. 2018;19(7):599–612. https://​doi.​org/​10.​2217/​pgs-2018-0004.CrossRefPubMed
110.
Zurück zum Zitat Amir O, Rogowski O, David M, Lahat N, Wolff R, Lewis BS. Circulating interleukin-10: association with higher mortality in systolic heart failure patients with elevated tumor necrosis factor-alpha. Isr Med Assoc J. 2010;12(3):158–62.PubMed Amir O, Rogowski O, David M, Lahat N, Wolff R, Lewis BS. Circulating interleukin-10: association with higher mortality in systolic heart failure patients with elevated tumor necrosis factor-alpha. Isr Med Assoc J. 2010;12(3):158–62.PubMed
117.
Zurück zum Zitat Bardien-Kruger S, Wulff H, Arieff Z, Brink P, Chandy KG, Corfield V. Characterisation of the human voltage-gated potassium channel gene, KCNA7, a candidate gene for inherited cardiac disorders, and its exclusion as cause of progressive familial heart block I (PFHBI). Eur J Hum Genet. 2002;10(1):36–43. https://doi.org/10.1038/sj.ejhg.5200739. CrossRefPubMed Bardien-Kruger S, Wulff H, Arieff Z, Brink P, Chandy KG, Corfield V. Characterisation of the human voltage-gated potassium channel gene, KCNA7, a candidate gene for inherited cardiac disorders, and its exclusion as cause of progressive familial heart block I (PFHBI). Eur J Hum Genet. 2002;10(1):36–43. https://​doi.​org/​10.​1038/​sj.​ejhg.​5200739. CrossRefPubMed
118.
Zurück zum Zitat Totsune K, Takahashi K, Mackenzie HS, Murakami O, Arihara Z, Sone M, Mouri T, Brenner BM, Ito S. Increased gene expression of adrenomedullin and adrenomedullin-receptor complexes, receptor-activity modifying protein (RAMP)2 and calcitonin-receptor-like receptor (CRLR) in the hearts of rats with congestive heart failure. Clin Sci (Lond). 2000;99(6):541–6.CrossRef Totsune K, Takahashi K, Mackenzie HS, Murakami O, Arihara Z, Sone M, Mouri T, Brenner BM, Ito S. Increased gene expression of adrenomedullin and adrenomedullin-receptor complexes, receptor-activity modifying protein (RAMP)2 and calcitonin-receptor-like receptor (CRLR) in the hearts of rats with congestive heart failure. Clin Sci (Lond). 2000;99(6):541–6.CrossRef
119.
Zurück zum Zitat Li X, Wang G, QiLi M, Liang H, Li T, E X, Feng Y, Zhang Y, Liu X, Qian M, et al. Aspirin Reduces Cardiac Interstitial Fibrosis by Inhibiting Erk1/2-Serpine2 and P-Akt Signalling Pathways. Cell Physiol Biochem. 2018;45(5):1955–1965. https://doi.org/10.1159/000487972 Li X, Wang G, QiLi M, Liang H, Li T, E X, Feng Y, Zhang Y, Liu X, Qian M, et al. Aspirin Reduces Cardiac Interstitial Fibrosis by Inhibiting Erk1/2-Serpine2 and P-Akt Signalling Pathways. Cell Physiol Biochem. 2018;45(5):1955–1965. https://​doi.​org/​10.​1159/​000487972
120.
126.
Zurück zum Zitat Martinelli N, Girelli D, Lunghi B, Pinotti M, Marchetti G, Malerba G, Pignatti PF, Corrocher R, Olivieri O, Bernardi F. Polymorphisms at LDLR locus may be associated with coronary artery disease through modulation of coagulation factor VIII activity and independently from lipid profile. Blood. 2010;116(25):5688–97. https://doi.org/10.1182/blood-2010-03-277079.CrossRefPubMed Martinelli N, Girelli D, Lunghi B, Pinotti M, Marchetti G, Malerba G, Pignatti PF, Corrocher R, Olivieri O, Bernardi F. Polymorphisms at LDLR locus may be associated with coronary artery disease through modulation of coagulation factor VIII activity and independently from lipid profile. Blood. 2010;116(25):5688–97. https://​doi.​org/​10.​1182/​blood-2010-03-277079.CrossRefPubMed
133.
Zurück zum Zitat Monroy-Muñoz IE, Angeles-Martinez J, Posadas-Sánchez R, Villarreal-Molina T, Alvarez-León E, Flores-Dominguez C, Cardoso-Saldaña G, Medina-Urrutia A, Juárez-Rojas JG, Posadas-Romero C, et al. PLA2G2A polymorphisms are associated with metabolic syndrome and type 2 diabetes mellitus. Results from the genetics of atherosclerotic disease Mexican study. Immunobiology. 2017;222(10):967–72. https://doi.org/10.1016/j.imbio.2016.08.014.CrossRefPubMed Monroy-Muñoz IE, Angeles-Martinez J, Posadas-Sánchez R, Villarreal-Molina T, Alvarez-León E, Flores-Dominguez C, Cardoso-Saldaña G, Medina-Urrutia A, Juárez-Rojas JG, Posadas-Romero C, et al. PLA2G2A polymorphisms are associated with metabolic syndrome and type 2 diabetes mellitus. Results from the genetics of atherosclerotic disease Mexican study. Immunobiology. 2017;222(10):967–72. https://​doi.​org/​10.​1016/​j.​imbio.​2016.​08.​014.CrossRefPubMed
142.
Zurück zum Zitat Kohara K, Tabara Y, Nakura J, Imai Y, Ohkubo T, Hata A, Soma M, Nakayama T, Umemura S, Hirawa N, et al. Identification of hypertension-susceptibility genes and pathways by a systemic multiple candidate gene approach: the millennium genome project for hypertension. Hypertens Res. 2008;31(2):203–12. https://doi.org/10.1291/hypres.31.203.CrossRefPubMed Kohara K, Tabara Y, Nakura J, Imai Y, Ohkubo T, Hata A, Soma M, Nakayama T, Umemura S, Hirawa N, et al. Identification of hypertension-susceptibility genes and pathways by a systemic multiple candidate gene approach: the millennium genome project for hypertension. Hypertens Res. 2008;31(2):203–12. https://​doi.​org/​10.​1291/​hypres.​31.​203.CrossRefPubMed
143.
Zurück zum Zitat Wen G, Wessel J, Zhou W, Ehret GB, Rao F, Stridsberg M, Mahata SK, Gent PM, Das M, Cooper RS, et al. An ancestral variant of Secretogranin II confers regulation by PHOX2 transcription factors and association with hypertension. Hum Mol Genet. 2007;16(14):1752–64. https://doi.org/10.1093/hmg/ddm123.CrossRefPubMed Wen G, Wessel J, Zhou W, Ehret GB, Rao F, Stridsberg M, Mahata SK, Gent PM, Das M, Cooper RS, et al. An ancestral variant of Secretogranin II confers regulation by PHOX2 transcription factors and association with hypertension. Hum Mol Genet. 2007;16(14):1752–64. https://​doi.​org/​10.​1093/​hmg/​ddm123.CrossRefPubMed
162.
164.
Zurück zum Zitat Piquer-Garcia I, Campderros L, Taxerås SD, Gavaldà-Navarro A, Pardo R, Vila M, Pellitero S, Martínez E, Tarascó J, Moreno P, et al. A role for oncostatin M in the impairment of glucose homeostasis in obesity. J Clin Endocrinol Metab. 2020;105(3):e337–48. https://doi.org/10.1210/clinem/dgz090.CrossRef Piquer-Garcia I, Campderros L, Taxerås SD, Gavaldà-Navarro A, Pardo R, Vila M, Pellitero S, Martínez E, Tarascó J, Moreno P, et al. A role for oncostatin M in the impairment of glucose homeostasis in obesity. J Clin Endocrinol Metab. 2020;105(3):e337–48. https://​doi.​org/​10.​1210/​clinem/​dgz090.CrossRef
171.
Zurück zum Zitat Lou Y, Wang S, Qu J, Zheng J, Jiang W, Lin Z, Zhang S. miR-424 promotes cardiac ischemia/reperfusion injury by direct targeting of CRISPLD2 and regulating cardiomyocyte pyroptosis. Int J Clin Exp Pathol. 2018;11(7):3222–35.PubMedPubMedCentral Lou Y, Wang S, Qu J, Zheng J, Jiang W, Lin Z, Zhang S. miR-424 promotes cardiac ischemia/reperfusion injury by direct targeting of CRISPLD2 and regulating cardiomyocyte pyroptosis. Int J Clin Exp Pathol. 2018;11(7):3222–35.PubMedPubMedCentral
176.
Zurück zum Zitat Brandt J, Warnke K, Jörgens S, Arolt V, Beer K, Domschke K, Haverkamp W, Kuhlmann SL, Müller-Nordhorn J, Rieckmann N, et al. Association of FKBP5 genotype with depressive symptoms in patients with coronary heart disease: a prospective study. J Neural Transm (Vienna). 2020;127(12):1651–62. https://doi.org/10.1007/s00702-020-02243-6.CrossRef Brandt J, Warnke K, Jörgens S, Arolt V, Beer K, Domschke K, Haverkamp W, Kuhlmann SL, Müller-Nordhorn J, Rieckmann N, et al. Association of FKBP5 genotype with depressive symptoms in patients with coronary heart disease: a prospective study. J Neural Transm (Vienna). 2020;127(12):1651–62. https://​doi.​org/​10.​1007/​s00702-020-02243-6.CrossRef
185.
Zurück zum Zitat Larsen LH, Rose CS, Sparsø T, Overgaard J, Torekov SS, Grarup N, Jensen DP, Albrechtsen A, Andersen G, Ek J, et al. Genetic analysis of the estrogen-related receptor alpha and studies of association with obesity and type 2 diabetes. Int J Obes (Lond). 2007;31(2):365–70. https://doi.org/10.1038/sj.ijo.0803408.CrossRef Larsen LH, Rose CS, Sparsø T, Overgaard J, Torekov SS, Grarup N, Jensen DP, Albrechtsen A, Andersen G, Ek J, et al. Genetic analysis of the estrogen-related receptor alpha and studies of association with obesity and type 2 diabetes. Int J Obes (Lond). 2007;31(2):365–70. https://​doi.​org/​10.​1038/​sj.​ijo.​0803408.CrossRef
Metadaten
Titel
Identification of candidate biomarkers and therapeutic agents for heart failure by bioinformatics analysis
verfasst von
Vijayakrishna Kolur
Basavaraj Vastrad
Chanabasayya Vastrad
Shivakumar Kotturshetti
Anandkumar Tengli
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
BMC Cardiovascular Disorders / Ausgabe 1/2021
Elektronische ISSN: 1471-2261
DOI
https://doi.org/10.1186/s12872-021-02146-8

Weitere Artikel der Ausgabe 1/2021

BMC Cardiovascular Disorders 1/2021 Zur Ausgabe

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.