Skip to main content

ORIGINAL RESEARCH article

Front. Cardiovasc. Med., 04 August 2022
Sec. General Cardiovascular Medicine
Volume 9 - 2022 | https://doi.org/10.3389/fcvm.2022.946229

Identification of key genes for hypertrophic cardiomyopathy using integrated network analysis of differential lncRNA and gene expression

Jing Cao1 Lei Yuan2*
  • 1Department of Cardiovascular Medicine, Third Xiangya Hospital, Central South University, Changsha, China
  • 2Department of Medical Affairs, Xiangya Hospital, Central South University, Changsha, China

Objective: Hypertrophic cardiomyopathy (HCM) is a complex heterogeneous heart disease. Recent reports found that long non-coding RNAs (lncRNAs) play an important role in the progression of cardiovascular diseases. The present study aimed to identify the novel lncRNAs and messenger RNAs (mRNAs) and determine the key pathways involved in HCM.

Methods: The lncRNA and mRNA sequencing datasets of GSE68316 and GSE130036 were downloaded from the Gene Expression Omnibus (GEO) database. An integrated co-expression network analysis was conducted to identify differentially expressed lncRNAs and differentially expressed mRNAs in patients with HCM. Then, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were explored to identify the biological functions and signaling pathways of the co-expression network. Protein–protein interaction (PPI) and hub gene networks were constructed by using Cytoscape software. Plasma samples of patients with HCM and the GSE89714 dataset were used to validate the bioinformatics results.

Results: A total of 1,426 differentially expressed long non-coding RNAs (lncRNAs) and 1,715 differentially expressed mRNAs were obtained from GSE68316, of which 965 lncRNAs and 896 mRNAs were upregulated and 461 lncRNAs and 819 mRNAs were downregulated. A total of 469 differentially expressed lncRNAs and 2,407 differentially expressed mRNAs were screened from GSE130036, of which 183 lncRNAs and 1,283 mRNAs were upregulated and 286 lncRNAs and 1,124 mRNAs were downregulated. A co-expression network was constructed and contained 30 differentially expressed lncRNAs and 63 differentially expressed mRNAs, which were primarily involved in ‘G-protein beta/gamma-subunit complex binding,' ‘polyubiquitin modification-dependent protein binding,' ‘Apelin signaling pathway,' and ‘Wnt signaling pathway.' The 10 hub genes in the upregulated network [G Protein Subunit Alpha I2 (GNAI2), G Protein Subunit Alpha I1 (GNAI1), G Protein Subunit Alpha I3 (GNAI3), G Protein Subunit Gamma 2 (GNG2), G Protein Subunit Beta 1 (GNB1), G Protein Subunit Gamma 13 (GNG13), G Protein Subunit Gamma Transducin 1 (GNGT1), G Protein Subunit Gamma 12 (GNG12), AKT Serine/Threonine Kinase 1 (AKT1) and GNAS Complex Locus (GNAS)] and the 10 hub genes in the downregulated network [Nucleotide-Binding Oligomerization Domain Containing Protein 2 (NOD2), Receptor-Interacting Serine/Threonine Kinase 2 (RIPK2), Nucleotide-Binding Oligomerization Domain Containing Protein 1 (NOD1), Mitochondrial Antiviral Signaling Protein (MAVS), Autophagy Related 16-Like 1 (ATG16L1), Interferon Induced With Helicase C Domain 1 (IFIH1), Autophagy Related 5 (ATG5), TANK-Binding Kinase 1 (TBK1), Caspase Recruitment Domain Family Member 9 (CARD9), and von Willebrand factor (VWF)] were screened using cytoHubba. The expression of LA16c-312E8.2 and RP5-1160K1.3 in the plasma of patients with HCM was elevated, and the expression of the MIR22 host gene (MIR22HG) was decreased, which was consistent with our analysis, while the expression of LINC00324 and Small Nucleolar RNA Host Gene 12 (SNHG12) was not significantly different between the two groups. Verification analyses performed on GSE89714 showed the upregulated mRNAs of Chloride Voltage-Gated Channel 7 (CLCN7), N-Acetylglucosamine-1-Phosphate Transferase Subunit Gamma (GNPTG), Unk Like Zinc Finger (UNKL), Adenosine Monophosphate Deaminase 2 (AMPD2), GNAI3, WD Repeat Domain 81 (WDR81), and Serpin Family F Member 1 (SERPINF1) and downregulated mRNAs of TATA-Box Binding Protein Associated Factor 12 (TAF12) co-expressed with five crucial lncRNAs. Moreover, GNAI2, GNAI3, GNG12, and vWF were upregulated and GNAS was downregulated in the top 10 hub genes of upregulated and downregulated PPI networks.

Conclusion: These findings from integrative biological analysis of lncRNA-mRNA co-expression networks explored the key genes and pathways and provide new insights into the understanding of the mechanism and discovering new therapeutic targets for HCM. Three differentially expressed pivotal lncRNAs (LA16c-312E8.2, RP5-1160K1.3, and MIR22HG) in the co-expression network may serve as biomarkers and intervention targets for the diagnosis and treatment of HCM.

Introduction

Hypertrophic cardiomyopathy (HCM) is a complex heterogeneous heart disease that has been recognized to be a significant cause of atrial fibrillation, heart failure, and arrhythmic sudden death and is one of the main causes of sudden cardiac death in young adults (1). Epidemiological investigations have shown that the prevalence of phenotypic expression of HCM in the adult general population was about 0.2% (1:500) (2, 3). HCM is characterized by myocardial hypertrophy, asymmetrical ventricular septal hypertrophy, ventricular narrowing, and abnormal myocardial cell hypertrophy. In the absence of other cardiac or systemic diseases, such as hypertension or aortic stenosis, echocardiography of a hypertrophic but undilated left ventricle is the easiest and most reliable method for the clinical diagnosis of HCM (4). The pathogenesis of HCM is strongly associated with the mutation of the genes encoding proteins of thick and thin myofilament contractile components of the cardiac sarcomere or Z-disk (5, 6). Although myosin heavy chain 7 (MYH7) and myosin binding protein C3 (MYBPC3), which encode β-myosin heavy chain and myosin binding protein C, are the two most common mutations (7, 8), the great heterogeneity and diversity in the molecular pathways make the exact mechanism of HCM remain indistinct.

About 99% of genomic sequences in the human genome do not encode proteins, but they are highly transcriptional and produce a broad spectrum of non-coding RNAs (ncRNAs) that show regulatory and structural functions. The encyclopedia of DNA elements (ENCODE) project and other studies showed that limiting the pathogenesis analysis of diseases to the protein-coding regions of the human genome is insufficient since many non-coding variations are associated with important human diseases (9–11). The most studied ncRNAs in the heart are microRNAs, and little is known about the other ncRNAs (12–14). Long non-coding RNAs (lncRNAs) are defined as RNAs with transcripts >200 nucleotides that do not encode protein. Recent reports demonstrated that lncRNAs play an important role in the progression of several cardiovascular diseases, such as acute myocardial infarction, heart failure, and atrial fibrillation (15–17). In HCM, lncRNAs are verified to be involved in vital biological processes of myocardial fibrosis, myocardial hypertrophy, and atherosclerosis (18, 19). For example, the overexpression of cardiomyocyte-specific non-coding repressor of factor of Nuclear factor of activated T-cells (NFAT) (NRON) exacerbated transverse aortic constriction (TAC)-induced hypertrophy in mice heart (19), and ROR sponges miR-133 to cause the re-expression of atrial natriuretic peptide and B-type natriuretic peptide, leading to the exacerbation of cardiac hypertrophy eventually (20). However, the role of lncRNAs in the progression of HCM remains to be further explored.

Currently, big data mining and precision medicine have gained considerable attention. In this study, we aimed to perform in-depth data mining based on former microarray studies. We identified differentially expressed lncRNAs and mRNAs in HCM progression by a comprehensive analysis of the public datasets GSE68316 and GSE13036, including 35 patients with HCM and 14 controls. Subsequently, we constructed the co-expression network of lncRNA-mRNA and performed gene ontology (GO) and Kyoto Encyclopedia of Genes/Genomes (KEGG) pathway enrichment analyses on the differentially expressed genes in the network. Finally, we constructed the protein–protein interaction (PPI) network of the differentially expressed genes to reveal the potential role of HCM-related mRNAs and lncRNAs. This study will provide useful information to explore the potential candidate biomarkers for HCM diagnosis, prognosis, and intervention targets.

Materials and methods

Data of gene expression profiles

The expression profile data of LncRNA and mRNA with the sequence numbers GSE68316 and GSE130036 were obtained from the Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo/) database. The left ventricular tissues of 35 patients with HCM and 14 healthy donors along with their clinical data were obtained, including 7 HCM and 5 healthy donors from GSE68316 and 28 HCM and 9 healthy donors from GSE130036. Gene expression profile dataset GSE89714 was collected from the GEO database to verify the key genes involved in HCM, which included the left ventricular tissues of 5 patients with HCM and 4 healthy donors. The characteristics of these expression profiles are presented in Supplementary Table 1.

Screening of differentially expressed lncRNAs and mRNAs

R language was used to analyze the initial data and identify differential lncRNAs and mRNAs. The Affy package was applied to perform data normalization, including converting the data to raw data and correcting the background. The limma software package was performed to screen the data for differentially expressed lncRNAs and mRNAs. The thresholds were |log2 (fold change) |>1 with the adjusted p-value < 0.05, and the genes that met the criteria were considered as differentially expressed lncRNAs and mRNAs. The gglot2 and heatmap packages were used to create volcano maps and heatmaps of differentially expressed lncRNAs and mRNAs to make them visible.

Co-expression network analysis of differentially expressed lncRNAs and mRNAs in patients with HCM

The Pearson correlation analysis of the differentially expressed lncRNAs and mRNAs was performed by the cor () function in the R language. The absolute value of the Pearson correlation coefficient ≥0.75 of lncRNA-mRNA pairs was considered to be significantly correlated and selected in the co-expression network. Subsequently, the co-expression network was visualized using Cytoscape software.

Gene ontology (GO) enrichment and Kyoto Encyclopedia of genes and genomes (KEGG) pathway analysis of genes in the network

The functions of genes enriched in the co-expression network, including cellular components, biological processes, and molecular functions, were determined by using gene ontology (GO) enrichment analysis. The enriched pathways of genes were analyzed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. ClusterProfiler package in R was applied to GO enrichment and KEGG pathway analyses with a threshold of adjusted p-value < 0.05.

Protein–protein interaction (PPI) network analysis of genes in the network

To predict the physical and functional interactions of proteins, protein–protein interaction (PPI) networks were constructed by the online Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), and the combined score >0.4 was used as the cutoff criterion. Following the construction of the PPI network, the Cytohubba plugin in Cytoscape software was adopted to calculate the degree of each protein node. In this study, the top 10 nodes were regarded to be hub genes.

Plasma collection and real-time quantitative PCR (qPCR)

The research design, protocol, and the use of human plasma samples were approved by the Medical Ethics Committee of the Xiangya Hospital of Central South University. The diagnosis of HCM was carried out according to the European Society of Cardiology Guidelines (21). 4 patients diagnosed with HCM and 4 healthy controls were included in this study. The clinical characteristics of the patients are listed in Supplementary Table 4. The peripheral whole blood of patients was collected in a tube containing an EDTA anticoagulant after overnight fasting, and plasma was separated by centrifugation. Total RNA was extracted from plasma samples using a miRNeasy Serum/Plasma Kit (Qiagen) according to the manufacturer's instructions. The quality and concentration of RNA were evaluated using NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific). Complementary DNAs (cDNAs) were synthesized from the total RNA using the PrimeScript RT reagent Kit (Takara) in accordance with the manufacturer's instructions. The lncRNAs were quantified by performing real-time qPCR on a QuantStudio 5 Real-Time PCR System with SYBR Premix Ex Taq assays (Takara). The fold difference in the expression level between the two groups was calculated using the 2−ΔΔCt method. The primer sequences are summarized in Supplementary Table 5.

Statistical analysis

The results are expressed as mean±SD. The T-tests or nonparametric tests were used to compare the differences between the HCM group and the control group. Graphs were constructed with R software, GraphPad Prism, and online visualization tools. A two-tailed P-value of < 0.05 was considered to be statistically significant. Statistical analyses were done using GraphPad Prism 8.0 and SPSS 20.0.

Results

Identification of differentially expressed lncRNAs and mRNAs in patients with HCM compared with the controls

The gene expression data of HCM were obtained from GSE68316 and GSE130036 datasets, which included 35 patients with HCM and 14 controls, and the basic information of the datasets is presented in Supplementary Table 1. After normalization, a total of 1,426 differential expressed lncRNAs and 1,715 differential expressed mRNAs were obtained from GSE68316 (adjusted P < 0.05, fold change >2). Among them, 965 lncRNAs were upregulated and 461 lncRNAs were downregulated; 896 mRNAs were upregulated and 819 mRNAs were downregulated. The volcano plots of differentially expressed genes are shown in Figures 1A,C, and the cluster heatmaps are shown in Figures 2A,C. A total of 469 differentially expressed lncRNAs and 2,407 differentially expressed mRNAs were obtained from GSE130036, of which 183 lncRNAs and 1,283 mRNAs were upregulated and 286 lncRNAs and 1,124 mRNAs were downregulated (adjusted P < 0.05, fold change >2). The volcano plots of differentially expressed genes in GSE130036 are shown in Figures 1B,D, and the cluster heatmaps are shown in Figures 2B,D. The top 10 differentially expressed lncRNAs involved in the HCM of the two datasets are listed in Table 1, and the top 10 differentially expressed mRNAs are listed in Table 2.

FIGURE 1
www.frontiersin.org

Figure 1. A Volcano plot of all the differentially expressed lncRNAs and mRNAs between patients with HCM and the controls. (A) A volcano plot of differentially expressed lncRNAs in the GSE68316 dataset. (B) A volcano plot of differentially expressed lncRNAs in the GSE130036 dataset. (C) A volcano plot of differentially expressed mRNAs in the GSE68316 dataset. (D) A volcano plot of differentially expressed mRNAs in GSE130036 dataset. Green dots represent differentially expressed genes with log-fold change <-1, red dots represent differentially expressed genes with log-fold change >1, and black spots represent genes with no significant difference in expression, adjusted p-value <0.05.

FIGURE 2
www.frontiersin.org

Figure 2. A cluster heat map of differentially expressed lncRNAs and mRNAs between HCM patients and the controls. (A) A heat map of differentially expressed lncRNAs in the GSE68316 dataset. (B) A heat map of differentially expressed lncRNAs in the GSE130036 dataset. (C) A heat map of differentially expressed mRNAs in the GSE68316 dataset. (D) A heat map of differentially expressed mRNAs in the GSE130036 dataset. The gradient color change from blue to red represents the changing process from downregulation to upregulation.

TABLE 1
www.frontiersin.org

Table 1. Top 10 upregulated and downregulated long noncoding RNAs in the myocardial tissues of patients with HCM in GSE68316 and GSE130036.

TABLE 2
www.frontiersin.org

Table 2. Top 10 upregulated and downregulated mRNAs in the myocardial tissues of patients with HCM in GSE68316 and GSE130036.

Subsequently, the Venn diagrams of upregulated and downregulated genes are shown in Figure 3. A total of 34 common differentially expressed lncRNAs with 15 upregulated and 19 downregulated lncRNAs (Figures 3A,C) and a total of 54 common differentially expressed mRNAs with 25 upregulated and 29 downregulated mRNAs (Figures 3B,D) were thus identified.

FIGURE 3
www.frontiersin.org

Figure 3. A Venn diagram of common differentially expressed lncRNAs and mRNAs. (A) A Venn diagram of upregulated common differentially expressed lncRNAs. (B) A Venn diagram of upregulated common differentially expressed mRNAs. (C) A Venn diagram of downregulated common differentially expressed lncRNAs. (D) A Venn diagram of downregulated common differentially expressed mRNAs.

Differential lncRNA-mRNA co-expression network construction and analysis

To determine the function of differentially expressed lncRNAs and their role in HCM, a gene co-expression network between lncRNAs and mRNAs was constructed. Then, Pearson's correlation coefficients in all samples were calculated for correlation analysis of mRNAs and lncRNAs, and lncRNA-mRNA pairs with a coefficient of ≥0.75 were selected. A total of 63 lncRNA-mRNA pairs were observed to be involved in 30 differentially expressed lncRNAs (13 upregulated and 17 downregulated) and 63 differentially expressed mRNAs (27 in upregulated network and 36 in downregulated network). In the upregulated co-expression network, 27 lncRNA-mRNA pairs were observed with 13 differentially expressed lncRNAs and 27 mRNAs (Figure 4A). About 36 lncRNA-mRNA pairs were observed in the downregulated co-expression network, including 17 differentially expressed lncRNAs and 36 mRNAs (Figure 4B).

FIGURE 4
www.frontiersin.org

Figure 4. Co-expression network analysis of common differentially expressed lncRNAs and mRNAs in HCM. (A) Co-expression network of upregulated common differentially expressed lncRNAs and mRNAs. (B) Co-expression network of downregulated common differentially expressed lncRNAs and mRNAs. Red spots represent lncRNAs, blue spots represent mRNAs, and lines represent the co-expression relationships between lncRNAs and mRNAs.

GO function enrichment analysis of genes in the co-expression network

Furthermore, to ascertain the potential functions of the identified differentially expressed lncRNAs and mRNAs in the co-expression network, GO function enrichment analysis was performed (Figure 5). GO analysis mainly described three categories of results: ‘molecular functions' (MF), ‘cellular components' (CC), and ‘biological processes' (BP). The analyses exhibited the top 10 significant enrichment of MF, CC, and BP in terms of upregulated and downregulated genes in the co-expression network. As shown in Figure 5A, the enrichment analysis of upregulated co-expression network related to molecular functions was primarily enriched in G-protein beta/gamma-subunit complex binding (P = 0.0002), GDP binding (P = 0.0031), and GTPase activity (P = 0.0055), and the analysis related to cellular components were mainly enriched in heterotrimeric G-protein complex (p = 0.0006), GTPase complex (P = 0.0006), and the extrinsic component of the cytoplasmic side of the plasma membrane (P = 0.0005). Additionally, the analysis of biological processes showed that genes were mainly involved in the positive regulation of cholesterol metabolic process (P = 0.0112), endothelial cell chemotaxis to fibroblast growth factor (P = 0.0112), and miRNA catabolic process (P = 0.0112). On the other hand, the enrichment analysis related to the molecular functions of downregulated co-expression network focused on K63-linked polyubiquitin modification-dependent protein binding (P = 0.0007), polyubiquitin modification-dependent protein binding (P = 0.0041), and serine-type endopeptidase inhibitor activity (P = 0.0132). Also, the cellular components were primarily enriched in mitotic spindle midzone (P = 0.0002), spindle midzone (P = 0.0015), and myelin sheath (P = 0.0030). In regard to biological process enrichment analysis of downregulated co-expression network, genes were mainly involved in the hydrogen peroxide biosynthetic process (P = 0.0004), the antibiotic biosynthetic process (P = 0.0007), and glutamate secretion (P = 0.0029) (Figure 5B).

FIGURE 5
www.frontiersin.org

Figure 5. Gene Ontology (GO) analysis of genes in the co-expression network. GO analysis shows: (A) The top 10 enriched terms associated with the molecular function (MF), cellular component (CC), and biological process (BP) of genes in the upregulated co-expression network. (B) The top 10 enriched terms associated with MF, CC, and BP of genes in the downregulated co-expression network.

KEGG pathway enrichment analysis of genes in the co-expression network

To further identify the biological processes associated with genes in the co-expression networ, KEGG pathway enrichment analyses were performed. As shown in Figure 6A, a total of nine key pathways were found through the KEGG pathway enrichment analysis of upregulated genes, which were primarily enriched in the Apelin signaling pathway (P = 0.0095), the Rap1 signaling pathway (P = 0.0215), and the regulation of the actin cytoskeleton (P = 0.0223). However, the downregulated genes in the co-expression network were significantly enriched in the Wnt signaling pathway (P = 0.0347), the folate biosynthesis pathway (P = 0.0473), and the transcriptional misregulation pathway (P = 0.0483) (Figure 6B).

FIGURE 6
www.frontiersin.org

Figure 6. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of genes in the co-expression network. (A) KEGG pathways enriched by genes in the upregulated co-expression network. (B) KEGG pathways enriched by genes in the downregulated co-expression network.

PPI network construction analysis

The PPI network analysis aims to study disease-related molecular mechanisms in co-expression networks and identify novel therapeutic targets from a systematic perspective. Consequently, a PPI network analysis of differentially expressed genes in the co-expression network was conducted with the STRING database. A total of 65 nodes and 362 edges were screened in the upregulated PPI network (Figure 7A). Nodes with high topological scores were considered to likely play an important role in disease, and G Protein Subunit Alpha I2 (GNAI2), G Protein Subunit Alpha I1 (GNAI1), G Protein Subunit Alpha I3 (GNAI3), G Protein Subunit Gamma 2 (GNG2), G Protein Subunit Beta 1 (GNB1), G Protein Subunit Gamma 13 (GNG13), G Protein Subunit Gamma Transducin 1 (GNGT1), G Protein Subunit Gamma 12 (GNG12), AKT Serine/Threonine Kinase 1 (AKT1), and GNAS Complex Locus (GNAS) were the top 10 hub genes in the PPI network of upregulated genes (Figure 7B, Supplementary Table 2). A total of 69 nodes and 314 edges were screened in the downregulated PPI network (Figure 7C). Nucleotide-Binding Oligomerization Domain Containing Protein 2 (NOD2), Receptor-Interacting Serine/Threonine Kinase 2 (RIPK2), Nucleotide-Binding Oligomerization Domain Containing 1 (NOD1), Mitochondrial Antiviral Signaling Protein (MAVS), Autophagy Related 16-Like 1 (ATG16L1), Interferon Induced With Helicase C Domain 1 (IFIH1), Autophagy Related 5 (ATG5), TANK Binding Kinase 1 (TBK1), Caspase Recruitment Domain Family Member 9 (CARD9), and von Willebrand factor (VWF) were the top 10 hub genes in the PPI network of downregulated genes (Figure 7D, Supplementary Table 2).

FIGURE 7
www.frontiersin.org

Figure 7. Protein–Protein Interaction (PPI) network analysis of genes in the co-expression network. (A,B) PPI network and hub genes in the upregulated co-expression network. (C,D) PPI network and hub genes in the downregulated co-expression network. The edges represent the interaction between two nodes, and the number of edges in one node is called the degree of the PPI network.

Verification of crucial lncRNAs and mRNAs in patients with HCM

In addition to expressing in specific tissues, lncRNA also exists stably in the circulating peripheral blood and hence can be used as biomarkers for the diagnosis and treatment of diseases (22). In order to verify the key lncRNAs and their diagnostic value for HCM, we collected plasma samples of patients with HCM and evaluated the level of five crucial lncRNAs (two upregulated and three downregulated) in the co-expression network by real-time qPCR. As shown in Figure 8A, lncRNA LA16c-312E8.2 (control: 0.99 ± 0.42 vs. HCM: 2.44 ± 0.93, P = 0.0303) and RP5-1160K1.3 (control: 1.15 ± 0.42 vs HCM: 2.97 ± 1.10, P = 0.0219) were upregulated and lncRNA MIR22 host gene (MIR22HG) (control: 1.19 ± 0.46 vs. HCM: 0.49 ± 0.23, P = 0.0360) was downregulated in patients with HCM, which was consistent with our previous analysis results. However, the levels of LINC00324 (control: 1.11 ± 0.46 vs. HCM: 0.93 ± 0.48, P = 0.6037) and Small Nucleolar RNA Host Gene 12 (SNHG12) (control: 1.14 ± 0.40 vs. HCM: 0.88 ± 0.53, P = 0.4657) were not significantly different between the two groups.

FIGURE 8
www.frontiersin.org

Figure 8. Verification of crucial lncRNAs and mRNAs in HCM patients. (A) The expression of five crucial lncRNAs in plasma of HCM patients and the controls were measured by real-time qPCR. (B) The expression level of mRNAs (co-expressed with five crucial lncRNAs) in the verification dataset GSE89714. (C) The expression level of the top 10 hub genes of upregulated and downregulated PPI network in the verification dataset GSE89714. ***p < 0.001.

In addition to verify the expression of lncRNAs in peripheral blood, we selected GSE89714 as the validation dataset to verify the expression levels of mRNAs (co-expressed with five crucial lncRNAs) in the co-expression network and the top 10 hub genes in the PPI network between HCM and healthy control cardiac tissues. As seen in Figure 8B, Chloride Voltage-Gated Channel 7 (CLCN7), N-Acetylglucosamine-1-Phosphate Transferase Subunit Gamma (GNPTG), and Unk Like Zinc Finger (UNKL), which co-expressed with LA16c-312E8.2, were upregulated in the cardiac tissue of patients with HCM, whereas Coiled-Coil Domain Containing 154 (CCDC154), Proline and Glutamate Rich with Coiled Coil 1 (PERCC1), and C16orf91 showed no significant differences between the two groups; LncRNA RP5-1160K1.3 co-expressed mRNAs Adenosine Monophosphate Deaminase 2 (AMPD2) and GNAI3 were upregulated, and G Protein Subunit Alpha Transducin 2 (GNAT2) and G Protein-Coupled Receptor 61 (GPR61) showed no significant changes. In the co-expressed mRNAs of MIR22HG, WD Repeat Domain 81 (WDR81) and Serpin Family F Member 1 (SERPINF1) were upregulated in the HCM group, and TLC Domain Containing 2 (TLCD2), Pre-mRNA Processing Factor 8 (PRPF8), and Serpin Family F Member 2 (SERPINF2) showed no significant differences. In the co-expressed mRNAs of SNHG12, TATA-Box Binding Protein Associated Factor 12 (TAF12) was downregulated in HCM cardiac tissues, and other genes like Regulator of Chromosome Condensation 1 (RCC1), TRNA Selenocysteine 1 Associated Protein 1 (TRNAU1AP), and RAB42 showed no significant differences. Unfortunately, there were no significant differences in the expression of LINC00324 co-expressed mRNAs, which include CST telomere replication complex component 1 (CTC1), Phosphoribosylformylglycinamidine synthase (PFAS), Transmembrane Protein 107 (TMEM107), Aurora Kinase B (AURKB), and C17orf59, compared with the control group. In regard to the top 10 hub genes, the analysis of the dataset GSE89714 found that GNAI2, GNAI3, and AKT1 were upregulated, whereas GNAS was downregulated in the upregulated PPI network. Other genes, including GNAI1, GNG2, GNB1, GNG13, GNGT1, and GNG12, showed no significant differences between HCM and control cardiac tissue. In the downregulated network, only one gene (vWF) was statistically different (Figure 8C).

Discussion

Hypertrophic cardiomyopathy, a type of hereditary cardiomyopathy, is the most common risk factor for sudden death in young people. With the emergence of next-generation sequencing (NGS) technologies, the list of variants and genes implicated in HCM is also expanding. These discoveries allow the precise identification of at-risk individuals prior to clinical diagnosis and provide novel therapeutic approaches for the modulation and prevention of HCM (23–26). A growing number of studies showed that lncRNAs play a momentous regulatory function in the pathophysiology of cardiovascular diseases, such as acute myocardial infarction, heart failure, and atrial fibrillation (15–17). Sequencing studies gradually found that lncRNAs also play an important role in the pathogenesis of HCM (27, 28), but the comprehensive evaluation of multiple datasets has not been thoroughly investigated. In the present study, we identified a total of 1,861 lncRNAs (1,133 upregulated and 728 downregulated) and 4,068 mRNAs (2,154 upregulated and 1,914 downregulated) were differentially expressed between the HCM and controls in the GSE68316 and GSE130036 datasets. Among these differentially expressed genes, 34 lncRNAs (15 upregulated and 19 downregulated) and 54 mRNAs (25 upregulated and 29 downregulated) were the commonly found differentially expressed genes. Co-expression network construction and subsequent GO enrichment and KEGG pathway analysis showed that the upregulated co-expression network was mainly enriched in G-protein beta/gamma-subunit complex binding, heterotrimeric G-protein complex, the Apelin signaling pathway, and the Rap1 signaling pathway. In addition, the downregulated network was mainly enriched in K63-linked polyubiquitin modification-dependent protein binding, mitotic spindle midzon, the Wnt signaling pathway, and the folate biosynthesis pathway. Of note, plasma sample validation of patients with HCM prompts that three key lncRNAs (LA16c-312E8.2, RP5-1160K1.3, and MIR22HG) may serve as biomarkers and intervention targets in HCM. These analyses provide novel insights to explore the potential mechanisms underlying HCM progression.

LncRNA has been found to regulate cardiomyocyte hypertrophy, apoptosis, angiogenesis, atherosclerosis, and other pathophysiological processes and play an important role in the development of cardiovascular disease (29–32). In recent years, it has also been reported that lncRNA plays an important role in HCM. Janika Viereck et al. reported that LncRNA H19 is highly conserved and downregulated in the failing hearts of mice, pigs, and humans. The H19 gene therapy prevents and reverses experimental pressure-overload-induced heart failure according to interaction with the polycomb repressive complex 2, suppressing H3K27 trimethylation, which, in turn, leads to reduced NFAT expression and activity (33). On the other hand, Xiao et al. (34) found that lncRNA X Inactive Specific Transcript (XIST) expression was significantly upregulated in hypertrophic mouse hearts and phenylephrine-treated cardiomyocytes. XIST promoted the progression of cardiac hypertrophy through competitively binding with miR-101 to enhance the expression of TLR2. After knocking down XIST, PE-induced cardiomyocyte hypertrophy was attenuated. In our research, a total of 1861 differentially expressed lncRNAs were identified by integrating datasets GSE68316 and GSE130036, among which 34 lncRNAs were expressed in both datasets. These common differentially expressed lncRNAs were analyzed to be co-expressed with several genes.

LncRNA LA16c-312E8.2 is also named LOC101929440. In addition to upregulated expression in HCM, LA16c-312E8.2 has also been reported to be differentially expressed in HER-2 enriched subtypes of breast cancer and pancreatic cancer (35, 36). However, the exact function and regulatory role of LA16c-312E8.2 has not yet been studied. The mRNAs co-expressed with LA16C-312E8.2 included CCDC154, CLCN7, GNPTG, PERCC1, UNKL, and C16orf91. CCDC154 is mainly involved in osteopetrosis and hypoplastic left heart syndrome (37, 38), and CLCN7 is mainly involved in osteopetrosis and angiogenesis (39). Diseases associated with GNPTG include mucolipidosis III gamma and mucolipidosis (40, 41), and those associated with PERCC1 include diarrhea 11, malabsorptive, congenital, and hepatocellular carcinoma (42–44). UNKL is associated with mucolipidosis (45), while the function and role of C16orf91 have not been reported. Among these co-expressed mRNAs, GNPTG and UNKL were involved in mucolipidosis that is associated with dilated cardiomyopathy in mucolipidosis type 2 (46), and CLCN7 was related to angiogenesis (39). These three genes are associated with cardiovascular diseases but have not been reported in HCM, which may be related to the lack of adequate research on HCM. In this study, our verification results showed that LncRNA LA16c-312E8.2 was upregulated in the plasma samples of patients with HCM compared to the controls. Besides, verification analysis of the public dataset showed that CCDC154, GNPTG, and UNKL were upregulated in the heart tissues of patients with HCM in the GSE89714 dataset, but there were no significant differences in other genes between the patients with HCM and controls.

MIR22HG, also known as C17orf91, is a downregulated lncRNA in HCM. The actions and functions of the MIR22HG gene are complex and have not been fully elucidated, but it has been reported to be involved in the regulation of cell proliferation and death according to several signaling pathways, including Wnt/β-catenin, epithelial-mesenchymal transition (EMT), notch, and STAT3 (47, 48). These pathways may promote the process of myocardial fibrosis in HCM. Previous studies reported that MIR22HG aggravates hypoxia-induced injury in cardiomyocytes and endothelial cells (49, 50); however, the exact mechanism of this gene in HCM has not been elucidated. MIR22HG was co-expressed with SERPINF1, SERPINF2, TLCD2, WDR81, and PRPF8. SERPINF1 and SERPINF2 are members of the serpin family. SERPINF1 strongly inhibits angiogenesis, and SERPINF2 is involved in alpha-2-plasmin inhibitor deficiency, vasculitis, and left ventricular diastolic dysfunction (51, 52). TLCD2 participates in Chromosome 17P13.3, Centromeric, Duplication Syndrome and is reported to be associated with increased left ventricular mass and cardiac hypertrophy (53). WDR81 is necessary for Purkinje and photoreceptor cell survival and has been reported to be associated with cerebellar ataxia and mental retardation. PRPF8 is essential for oxidative stress injury-induced mitophagy, which in turn leads to intracellular energy metabolism disorders (54). Although these genes have not been reported in HCM, they are involved in pathophysiological processes, including left ventricular diastolic dysfunction, myocardial hypertrophy, and disturbances in cellular energy metabolism, which are closely related to HCM. Therefore, it deserves our attention and further study.

We further analyzed the co-expression network of lncRNA-mRNA and found two hub gene networks. In the upregulated network, 10 hub genes were enumerated, which were involved in regulating the G protein-coupled receptor (GPCR) signaling pathway and GTPase activity. GPCR mediates many pathophysiological processes, such as cardiac contractility, hypertrophy, proliferation, survival, and fibrosis, which are closely correlated with HCM (55–57). Xin Liu et al. reported that ERK1/2 communicates with the GPCR signaling pathways to promote HCM upon Ang-II stimulation (58). In addition, Feng Xie et al. found that the GPCR family member APJ interacts with its ligand and promotes cardiac hypertrophy through the PI3K-autophagy and endoplasmic reticulum stress-autophagy pathways (59). Although the position of GCRP in cardiac hypertrophy is now well-recognized, the roles and mechanisms of GCRP family members in HCM remain poorly understood. Therefore, we need to focus on and study these genes and their related GPCR pathways in the next step. With regard to the downregulated network, 10 hub genes were filtered out, which were enriched in positive regulation of tumor necrosis factor production and nucleotide-binding oligomerization domain (NOD)-like receptor signaling pathway. A clinical study found that, after non-surgical septal reduction therapy for patients with hypertrophic obstructive cardiomyopathy, the expression of tumor necrosis factor-α was decreased and cardiac hypertrophy was regressed (60). Knockout of tumor necrosis factor-related protein reduced myocardial hypertrophy in mice (61). NOD leucine-rich repeat (LRR) protein family plays an important role in regulating the intracellular recognition of bacterial components by immune cells and is crucial for the maintenance of immune homeostasis (62). Jing Zong et al. found that NOD2 expression was upregulated in cardiomyocytes of aortic banding hypertrophic mice. NOD2 deficiency promoted cardiac hypertrophy and fibrosis by activating TLR4 and the MAPKs, NF-kB, and TGF-β/Smad pathways (63). Furthermore, the data showed that NOD-like receptor NLRP3 expression was downregulated in the aortic banding hypertrophic mice. NLRP3 deficiency accelerated cardiac hypertrophy, fibrosis, and inflammation responses with deteriorating cardiac dysfunction (64). Although the role of NOD in HCM is still controversial at present, its status and importance for cardiac hypertrophy have been widely recognized. Therefore, it is crucial to explore and elucidate the mechanism of these hub genes in HCM.

This study has certain limitations. Although both datasets contain several hundred differentially expressed lncRNAs and mRNAs, only a few of them overlapped. It is likely that the dominant mutant genotypes of the patients with HCM in the GSE130036 dataset were MYH7 and MYBPC3 mutations, while the dominant mutant genotypes of the patients with HCM in the GSE68316 dataset were not clear. It is well-known that numerous mutant genotypes lead to HCM, but pathophysiological processes caused by each genotype are not the same, resulting in different gene expressions in patients with different genotypes (65, 66). Notably, inconsistent sample size, sequencing methods, and baseline data of patients between the two datasets may also account for a few of the co-differentially expressed genes. In addition, due to the difficulties in obtaining cardiac tissues from patients with HCM, we did not validate the results in cardiac tissues. Finally, we could obtain limited plasma samples from patients with HCM for validation, and this may be the reason for the inconsistent results of lncRNA LINC00324 and SNHG12 between GEO analysis and our real-time qPCR data. In the future, more samples are needed to verify the abovementioned results.

Conclusion

The present study aimed to analyze and elucidate the differentially expressed lncRNAs and mRNAs by integrative biological analysis of lncRNA-mRNA co-expression networks in patients with HCM. Three key lncRNAs (LA16c-312E8.2, RP5-1160K1.3, and MIR22HG) identified in patients with HCM may serve as biomarkers and intervention targets for HCM. These findings will provide a new thought in understanding the cause and mechanism of HCM and discovering new biomarkers or therapeutic targets for clinic treatment.

Data availability statement

The datasets presented in this study can be found in online repositories. The names of the repository/repositories and accession number(s) can be found in the article/Supplementary material.

Ethics statement

This study was reviewed and approved by the Committee at Xiangya Hospital of Central South University (IRB No. 202113548).

Author contributions

JC conducted the statistical analysis, carried out the experiments, and drafted the article. LY contributed to reviewing, editing, and revising the article. All authors contributed to manuscript revision, read, and approved the submitted version.

Funding

The work was supported by grants from Xiangya Hospital, Wei Ming Clinical, and Rehabilitation Research Fund of Peking University (No. xywmII09).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fcvm.2022.946229/full#supplementary-material

References

1. Veselka J, Anavekar NS, Charron P. Hypertrophic obstructive cardiomyopathy. Lancet. (2017) 389:1253–67. doi: 10.1016/S0140-6736(16)31321-6

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Maron BJ, Gardin JM, Flack JM, Gidding SS, Kurosaki TT, Bild DE. Prevalence of hypertrophic cardiomyopathy in a general population of young adults. Echocardiographic analysis of 4111 subjects in the CARDIA Study. Coronary artery risk development in (Young) adults. Circulation. (1995) 92:785–9. doi: 10.1161/01.CIR.92.4.785

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Zou Y, Song L, Wang Z, Ma A, Liu T, Gu H, et al. Prevalence of idiopathic hypertrophic cardiomyopathy in China: a population-based echocardiographic analysis of 8,080 adults. Am J Med. (2004) 116:14–8. doi: 10.1016/j.amjmed.2003.05.009

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Geske JB, Ommen SR, Gersh BJ. Hypertrophic cardiomyopathy: Clinical Update. JACC Heart Fail. (2018) 6:364–75. doi: 10.1016/j.jchf.2018.02.010

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Cirino AL, Harris S, Lakdawala NK, Michels M, Olivotto I, Day SM, et al. Role of genetic testing in inherited cardiovascular disease: a review. Jama Cardiol. (2017) 2:1153–60. doi: 10.1001/jamacardio.2017.2352

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Burke MA, Cook SA, Seidman JG, Seidman CE. Clinical and mechanistic insights into the genetics of cardiomyopathy. J Am Coll Cardiol. (2016) 68:2871–86. doi: 10.1016/j.jacc.2016.08.079

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Fourey D, Care M, Siminovitch KA, Weissler-Snir A, Hindieh W, Chan RH, et al. Prevalence and clinical implication of double mutations in hypertrophic cardiomyopathy: revisiting the gene-dose effect. Circ Cardiovasc Genet. (2017) 10:1685. doi: 10.1161/CIRCGENETICS.116.001685

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Biagini E, Olivotto I, Iascone M, Parodi MI, Girolami F, Frisso G, et al. Significance of sarcomere gene mutations analysis in the end-stage phase of hypertrophic cardiomyopathy. Am J Cardiol. (2014) 114:769–76. doi: 10.1016/j.amjcard.2014.05.065

PubMed Abstract | CrossRef Full Text | Google Scholar

9. An integrated encyclopedia of DNA elements in the human genome. Nature. (2012) 489:57–74. doi: 10.1038/nature11247

PubMed Abstract | CrossRef Full Text

10. Hoffman MM, Ernst J, Wilder SP, Kundaje A, Harris RS, Libbrecht M, et al. Integrative annotation of chromatin elements from ENCODE data. Nucleic Acids Res. (2013) 41:827–41. doi: 10.1093/nar/gks1284

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Hardison RC. Genome-wide epigenetic data facilitate understanding of disease susceptibility association studies. J Biol Chem. (2012) 287:30932–40. doi: 10.1074/jbc.R112.352427

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Barwari T, Joshi A, Mayr M. MicroRNAs in cardiovascular disease. J Am Coll Cardiol. (2016) 68:2577–84. doi: 10.1016/j.jacc.2016.09.945

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Lu D, Thum T. RNA-based diagnostic and therapeutic strategies for cardiovascular disease. Nat Rev Cardiol. (2019) 16:661–74. doi: 10.1038/s41569-019-0218-x

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Philippen LE, Dirkx E, Wit JB, Burggraaf K, de Windt LJ Da CMP. Antisense MicroRNA therapeutics in cardiovascular disease: quo vadis? Mol Ther. (2015) 23:1810–8. doi: 10.1038/mt.2015.133

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Zhang Y, Jiao L, Sun L, Li Y, Gao Y, Xu C, et al. LncRNA ZFAS1 as a SERCA2a inhibitor to cause intracellular Ca(2+) overload and contractile dysfunction in a mouse model of myocardial infarction. Circ Res. (2018) 122:1354–68. doi: 10.1161/CIRCRESAHA.117.312117

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Liang H, Su X, Wu Q, Shan H, Lv L, Yu T, et al. LncRNA 2810403D21Rik/Mirf promotes ischemic myocardial injury by regulating autophagy through targeting Mir26a. Autophagy. (2020) 16:1077–91. doi: 10.1080/15548627.2019.1659610

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Wang LY, Shen H, Yang Q, Min J, Wang Q, Xi W, et al. LncRNA-LINC00472 contributes to the pathogenesis of atrial fibrillation (Af) by reducing expression of JP2 and RyR2 via miR-24. Biomed Pharmacother. (2019) 120:109364. doi: 10.1016/j.biopha.2019.109364

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Wang L, Wang J, Li G, Xiao J. Non-coding RNAs in physiological cardiac hypertrophy. Adv Exp Med Biol. (2020) 1229:149–61. doi: 10.1007/978-981-15-1671-9_8

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Hoepfner J, Leonardy J, Lu D, Schmidt K, Hunkler HJ, Biss S, et al. The long non-coding RNA NRON promotes the development of cardiac hypertrophy in the murine heart. Mol Ther. (2022) 30:1265–74. doi: 10.1016/j.ymthe.2021.11.018

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Jiang F, Zhou X, Huang J. Long non-coding RNA-ROR mediates the reprogramming in cardiac hypertrophy. PLoS ONE. (2016) 11:e152767. doi: 10.1371/journal.pone.0152767

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Elliott PM, Anastasakis A, Borger MA, Borggrefe M, Cecchi F, Charron P, et al. 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy: the task force for the diagnosis and management of hypertrophic cardiomyopathy of the european society of cardiology (ESC). Eur Heart J. (2014) 35:2733–79. doi: 10.1093/eurheartj/ehu284

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Viereck J, Thum T. Circulating noncoding rnas as biomarkers of cardiovascular disease and injury. Circ Res. (2017) 120:381–99. doi: 10.1161/CIRCRESAHA.116.308434

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Cecconi M, Parodi MI, Formisano F, Spirito P, Autore C, Musumeci MB, et al. Targeted next-generation sequencing helps to decipher the genetic and phenotypic heterogeneity of hypertrophic cardiomyopathy. Int J Mol Med. (2016) 38:1111–24. doi: 10.3892/ijmm.2016.2732

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Ashley EA, Reuter CM, Wheeler MT. Genome sequencing in hypertrophic cardiomyopathy. J Am Coll Cardiol. (2018) 72:430–3. doi: 10.1016/j.jacc.2018.05.029

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Chen R, Ge T, Jiang W, Huo J, Chang Q, Geng J, et al. Identification of biomarkers correlated with hypertrophic cardiomyopathy with co-expression analysis. J Cell Physiol. (2019) 234:21999–2008. doi: 10.1002/jcp.28762

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Walsh R, Offerhaus JA, Tadros R, Bezzina CR. Minor hypertrophic cardiomyopathy genes, major insights into the genetics of cardiomyopathies. Nat Rev Cardiol. (2022) 19:151–67. doi: 10.1038/s41569-021-00608-2

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Yuan Y, Wang J, Chen Q, Wu Q, Deng W, Zhou H, et al. Long non-coding RNA cytoskeleton regulator RNA (CYTOR) modulates pathological cardiac hypertrophy through miR-155-mediated IKKi signaling. Biochim Biophys Acta Mol Basis Dis. (2019) 1865:1421–7. doi: 10.1016/j.bbadis.2019.02.014

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Gao J, Collyer J, Wang M, Sun F, Xu F. Genetic dissection of hypertrophic cardiomyopathy with myocardial RNA-Seq. Int J Mol Sci. (2020) 21:3040. doi: 10.3390/ijms21093040

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Lee JH, Gao C, Peng G, Greer C, Ren S, Wang Y, et al. Analysis of transcriptome complexity through RNA sequencing in normal and failing murine hearts. Circ Res. (2011) 109:1332–41. doi: 10.1161/CIRCRESAHA.111.249433

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Wang K, Long B, Zhou LY, Liu F, Zhou QY, Liu CY, et al. CARL lncRNA inhibits anoxia-induced mitochondrial fission and apoptosis in cardiomyocytes by impairing miR-539-dependent PHB2 downregulation. Nat Commun. (2014) 5:3596. doi: 10.1038/ncomms4596

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Simion V, Zhou H, Haemmig S, Pierce JB, Mendes S, Tesmenitsky Y, et al. A macrophage-specific lncRNA regulates apoptosis and atherosclerosis by tethering HuR in the nucleus. Nat Commun. (2020) 11:6135. doi: 10.1038/s41467-020-19664-2

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Wu R, Hu W, Chen H, Wang Y, Li Q, Xiao C, et al. A Novel human long non-coding RNA SCDAL promotes angiogenesis through SNF5-mediated GDF6 expression. Adv Sci. (2021) 8:e2004629. doi: 10.1002/advs.202004629

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Viereck J, Buhrke A, Foinquinos A, Chatterjee S, Kleeberger JA, Xiao K, et al. Targeting muscle-enriched long non-coding RNA H19 reverses pathological cardiac hypertrophy. Eur Heart J. (2020) 41:3462–74. doi: 10.1093/eurheartj/ehaa519

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Xiao L, Gu Y, Sun Y, Chen J, Wang X, Zhang Y, et al. The long noncoding RNA XIST regulates cardiac hypertrophy by targeting miR-101. J Cell Physiol. (2019) 234:13680–92. doi: 10.1002/jcp.28047

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Rossignoli F, Spano C, Grisendi G, Foppiani EM, Golinelli G, Mastrolia I, et al. MSC-delivered soluble TRAIL and paclitaxel as novel combinatory treatment for pancreatic adenocarcinoma. Theranostics. (2019) 9:436–48. doi: 10.7150/thno.27576

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Yang F, Lyu S, Dong S, Liu Y, Zhang X, Wang O. Expression profile analysis of long noncoding RNA in HER-2-enriched subtype breast cancer by next-generation sequencing and bioinformatics. Onco Targets Ther. (2016) 9:761–72. doi: 10.2147/OTT.S97664

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Liao W, Zhao R, Lu L, Zhang R, Zou J, Xu T, et al. Overexpression of a novel osteopetrosis-related gene CCDC154 suppresses cell proliferation by inducing G2/M arrest. Cell Cycle. (2012) 11:3270–9. doi: 10.4161/cc.21642

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Liu X, Yagi H, Saeed S, Bais AS, Gabriel GC, Chen Z, et al. The complex genetics of hypoplastic left heart syndrome. Nat Genet. (2017) 49:1152–9. doi: 10.1038/ng.3870

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Peng H, He HB, Wen T. A Novel Variant in CLCN7 Regulates the coupling of angiogenesis and osteogenesis. Front Cell Dev Biol. (2020) 8:599826. doi: 10.3389/fcell.2020.599826

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Di Lorenzo G, Westermann LM, Yorgan TA, Sturznickel J, Ludwig NF, Ammer LS, et al. Pathogenic variants in GNPTAB and GNPTG encoding distinct subunits of GlcNAc-1-phosphotransferase differentially impact bone resorption in patients with mucolipidosis type II and III. GENET MED. (2021) 23:2369–77. doi: 10.1038/s41436-021-01285-9

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Westermann LM, Fleischhauer L, Vogel J, Jenei-Lanzl Z, Ludwig NF, Schau L, et al. Imbalanced cellular metabolism compromises cartilage homeostasis and joint function in a mouse model of mucolipidosis type III gamma. Dis Model Mech. (2020) 13:6425. doi: 10.1242/dmm.046425

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Oz-Levi D, Olender T, Bar-Joseph I, Zhu Y, Marek-Yagel D, Barozzi I, et al. Noncoding deletions reveal a gene that is critical for intestinal function. Nature. (2019) 571:107–11. doi: 10.1038/s41586-019-1312-2

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Yang LY Li L, Jiang H, Shen Y, Plunkett W. Expression of ERCC1 antisense RNA abrogates gemicitabine-mediated cytotoxic synergism with cisplatin in human colon tumor cells defective in mismatch repair but proficient in nucleotide excision repair. Clin Cancer Res. (2000) 6:773–81.

PubMed Abstract | Google Scholar

44. Goulet O, Pigneur B, Charbit-Henrion F. Congenital enteropathies involving defects in enterocyte structure or differentiation. Best Pract Res Clin Gastroenterol. (2022) 56–57:101784. doi: 10.1016/j.bpg.2021.101784

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Lores P, Visvikis O, Luna R, Lemichez E, Gacon G. The SWI/SNF protein BAF60b is ubiquitinated through a signalling process involving Rac GTPase and the RING finger protein Unkempt. FEBS J. (2010) 277:1453–64. doi: 10.1111/j.1742-4658.2010.07575.x

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Carboni E, Sestito S, Lucente M, Morrone A, Zampini L, Chimenz R, et al. Dilated cardiomyopathy in mucolipidosis type 2. J Biol Regul Homeost Agents. (2020) 34(4 Suppl. 2):71–7. Available online at: https://www.biolifesas.org/EN/Y2020/V34/I4(S2)/71

PubMed Abstract | Google Scholar

47. Zhang L, Li C, Su X. Emerging impact of the long noncoding RNA MIR22HG on proliferation and apoptosis in multiple human cancers. J Exp Clin Cancer Res. (2020) 39:271. doi: 10.1186/s13046-020-01784-8

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Xu J, Shao T, Song M, Xie Y, Zhou J, Yin J, et al. MIR22HG acts as a tumor suppressor via TGFbeta/SMAD signaling and facilitates immunotherapy in colorectal cancer. Mol Cancer. (2020) 19:51. doi: 10.1186/s12943-020-01174-w

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Yan X, Hou J. miR-22 host gene enhances nuclear factor-kappa B activation to aggravate hypoxia-induced injury in AC16 cardiomyocytes. Cell Transplant. (2021) 30:2139983027. doi: 10.1177/0963689721990323

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Voellenkle C, Garcia-Manteiga JM, Pedrotti S, Perfetti A, De Toma I, Da SD, et al. Implication of Long noncoding RNAs in the endothelial cell response to hypoxia revealed by RNA-sequencing. Sci Rep. (2016) 6:24141. doi: 10.1038/srep24141

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Nakata I, Yamashiro K, Yamada R, Gotoh N, Nakanishi H, Hayashi H, et al. Genetic variants in pigment epithelium-derived factor influence response of polypoidal choroidal vasculopathy to photodynamic therapy. Ophthalmology. (2011) 118:1408–15. doi: 10.1016/j.ophtha.2010.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Verbree-Willemsen L, Zhang YN, Ibrahim I, Ooi S, Wang JW, Mazlan MI, et al. Extracellular vesicle Cystatin C and CD14 are associated with both renal dysfunction and heart failure. ESC Heart Fail. (2020) 7:2240–9. doi: 10.1002/ehf2.12699

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Harper AR, Mayosi BM, Rodriguez A, Rahman T, Hall D, Mamasoula C, et al. Common variation neighbouring micro-RNA 22 is associated with increased left ventricular mass. PLoS One. (2013) 8:e55061. doi: 10.1371/journal.pone.0055061

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Xu G, Li T, Chen J, Li C, Zhao H, Yao C, et al. Autosomal dominant retinitis pigmentosa-associated gene PRPF8 is essential for hypoxia-induced mitophagy through regulating ULK1 mRNA splicing. Autophagy. (2018) 14:1818–30. doi: 10.1080/15548627.2018.1501251

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Tilley DG. G protein-dependent and G protein-independent signaling pathways and their impact on cardiac function. Circ Res. (2011) 109:217–30. doi: 10.1161/CIRCRESAHA.110.231225

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Murga C, Arcones AC, Cruces-Sande M, Briones AM, Salaices M, Mayor FJ, et al. Protein-coupled receptor Kinase 2 (GRK2) as a potential therapeutic target in cardiovascular and metabolic diseases. Front Pharmacol. (2019) 10:112. doi: 10.3389/fphar.2019.00112

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Belmonte SL, Blaxall BC. G protein coupled receptor kinases as therapeutic targets in cardiovascular disease. CIRC RES. (2011) 109:309–19. doi: 10.1161/CIRCRESAHA.110.231233

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Liu X, Lin L, Li Q, Ni Y, Zhang C, Qin S, et al. ERK1/2 communicates GPCR and EGFR signaling pathways to promote CTGF-mediated hypertrophic cardiomyopathy upon Ang-II stimulation. BMC Mol Cell Biol. (2019) 20:14. doi: 10.1186/s12860-019-0202-7

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Xie F, Wu D, Huang SF, Cao JG Li HN, He L, et al. The endoplasmic reticulum stress-autophagy pathway is involved in apelin-13-induced cardiomyocyte hypertrophy in vitro. Acta Pharmacol Sin. (2017) 38:1589–600. doi: 10.1038/aps.2017.97

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Nagueh SF, Stetson SJ, Lakkis NM, Killip D, Perez-Verdia A, Entman ML, et al. Decreased expression of tumor necrosis factor-alpha and regression of hypertrophy after nonsurgical septal reduction therapy for patients with hypertrophic obstructive cardiomyopathy. Circulation. (2001) 103:1844–50. doi: 10.1161/01.CIR.103.14.1844

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Ma ZG, Yuan YP, Zhang X, Xu SC, Kong CY, Song P, et al. C1q-tumour necrosis factor-related protein-3 exacerbates cardiac hypertrophy in mice. Cardiovasc Res. (2019) 115:1067–77. doi: 10.1093/cvr/cvy279

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Philpott DJ, Sorbara MT, Robertson SJ, Croitoru K, Girardin SE. NOD proteins: regulators of inflammation in health and disease. Nat Rev Immunol. (2014) 14:9–23. doi: 10.1038/nri3565

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Zong J, Salim M, Zhou H, Bian ZY Dai J, Yuan Y, et al. NOD2 deletion promotes cardiac hypertrophy and fibrosis induced by pressure overload. Lab Invest. (2013) 93:1128–36. doi: 10.1038/labinvest.2013.99

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Li F, Zhang H, Yang L, Yong H, Qin Q, Tan M, et al. NLRP3 deficiency accelerates pressure overload-induced cardiac remodeling via increased TLR4 expression. J Mol Med (Berl). (2018) 96:1189–202. doi: 10.1007/s00109-018-1691-0

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Ho CY, Day SM, Ashley EA, Michels M, Pereira AC, Jacoby D, et al. Genotype and lifetime burden of disease in hypertrophic cardiomyopathy: insights from the sarcomeric human cardiomyopathy registry (SHaRe). Circulation. (2018) 138:1387–98. doi: 10.1161/CIRCULATIONAHA.117.033200

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Cambronero F, Marin F, Roldan V, Hernandez-Romero D, Valdes M, Lip GY. Biomarkers of pathophysiology in hypertrophic cardiomyopathy: implications for clinical management and prognosis. Eur Heart J. (2009) 30:139–51. doi: 10.1093/eurheartj/ehn538

PubMed Abstract | CrossRef Full Text | Google Scholar

Glossary

ADD3, Adducin 3; ALDH6A1, Aldehyde Dehydrogenase 6 Family Member A1; ALG14, ALG14 UDP-N-Acetylglucosaminyltransferase Subunit; AMPD2, Adenosine Monophosphate Deaminase 2; ANP, Atrial natriuretic peptide; ATG5, Autophagy Related 5; ATG16L1, Autophagy Related 16-Like 1; AURKB, Aurora Kinase B; B3GALT5-AS1, B3GALT5 Antisense RNA 1; BNP, Brain Natriuretic Peptide; BP, Biological processes; BTN3A2, Butyrophilin Subfamily 3 Member A2; BTNL9, Butyrophilin Like 9; C16orf91, Chromosome 16 Open Reading Frame 91; C17orf108, LYR Motif Containing 9, LYRM9; CA1, Carbonic Anhydrase 1; CARD9, Caspase Recruitment Domain Family Member 9; CCDC7, Coiled-Coil Domain Containing 7; CCDC154, Coiled-Coil Domain Containing 154; CLCN7, Chloride Voltage-Gated Channel 7; COMMD6, COMM Domain Containing 6; CORIN, Corin, Serine Peptidase; CTC1, CST telomere replication complex component 1; ECHDC1, Ethylmalonyl-CoA Decarboxylase 1; ELK4, ETS Transcription Factor ELK4; ENCODE, Encyclopedia of DNA Elements; EPB42, Erythrocyte Membrane Protein Band 4.2; FAM167A-AS1, FAM167A Antisense RNA 1; FURIN, Furin, Paired Basic Amino Acid Cleaving Enzyme; GEO, Gene Expression Omnibus; GO, Gene Ontology; GNAI1, G Protein Subunit Alpha I1; GNAI2, G Protein Subunit Alpha I2; GNAI3, G Protein Subunit Alpha I3; GNAT2, G Protein Subunit Alpha Transducin 2; GNB1, G Protein Subunit Beta 1; GNG2, G Protein Subunit Gamma 2; GNG12, G Protein Subunit Gamma 12; GNG13, G Protein Subunit Gamma 13; GNGT1, G Protein Subunit Gamma Transducin 1; GNAS, GNAS Complex Locus; GNPTG, N-Acetylglucosamine-1-Phosphate Transferase Subunit Gamma; HCM, Hypertrophic cardiomyopathy; HBG1, Hemoglobin Subunit Gamma 1; HBM, Hemoglobin Subunit Mu; IFIH1, Interferon Induced With Helicase C Domain 1; KEGG, Kyoto Encyclopedia of Genes and Genomes; KRT1, Keratin 1; KCNIP2, Potassium Voltage-Gated Channel Interacting Protein 2; LncRNAs, Long noncoding RNAs; LRR, leucine rich repeat; MF, molecular function; MAVS, Mitochondrial antiviral-signaling protein; MIR22HG, MIR22 Host Gene; MYBPC3, Myosin Binding Protein C3; MYH7, Myosin Heavy Chain 7; MYH14, Myosin Heavy Chain 14; ncRNA, noncoding RNA; NOD, Nucleotide-Binding Oligomerization Domain; NOD1, Nucleotide-Binding Oligomerization Domain Containing Protein 1; NOD2, Nucleotide-Binding Oligomerization Domain Containing 2; NDUFS2, NADH: Ubiquinone Oxidoreductase Core Subunit S2; P2RY14, Phosphodiesterase 4D Interacting Protein; PDE4DIP, Phosphodiesterase 4D Interacting Protein; PERCC1, Proline and Glutamate Rich with Coiled Coil 1; PFAS, Phosphoribosylformylglycinamidine synthase; PFDN5, Prefoldin Subunit 5; PLCH1-AS2, PLCH1 Antisense RNA 2; PPI, Protein–protein interaction; PPP1R35-AS1, PPP1R35 Antisense RNA 1; PCBP2, Poly(RC) Binding Protein 2; PRPF8, pre-mRNA processing factor 8; RCC1, Regulator of Chromosome Condensation 1; RIPK2, Receptor-Interacting Serine/Threonine Kinase 2; RPL26, Ribosomal Protein L26; RPS24, Ribosomal Protein S24; SELENBP1, Selenium Binding Protein 1; SERPINF1, Serpin Family F Member 2; SERPINF2, Serpin Family F Member 2; SLC4A1, Solute Carrier Family 4 Member 1; SELENOI, Selenoprotein I; STXBP5, Syntaxin Binding Protein 5; TAF12, TATA-Box Binding Protein Associated Factor 12; TBK1, TANK Binding Kinase 1; TECRL, Trans-2,3-Enoyl-CoA Reductase Like; TLCD2, TLC Domain Containing 2; TMEM107, Transmembrane Protein 107; TRNAU1AP, TRNA Selenocysteine 1 Associated Protein 1; TSPEAR-AS1, TSPEAR Antisense RNA 1; TSPEAR-AS2, TSPEAR Antisense RNA 2; TNS1, Tensin 1; TROAP, Trophinin Associated Protein; UNKL, Unk Like Zinc Finger; UQCRH, Ubiquinol-Cytochrome C Reductase Hinge Protein; VWF, von Willebrand factor; WDR81, WD Repeat Domain 81; XIST, X Inactive Specific Transcript; ZNF16, Zinc Finger Protein 16.

Keywords: hypertrophic cardiomyopathy, differentially expressed long non-coding RNAs, differentially expressed mRNAs, co-expression network, bioinformatics analysis

Citation: Cao J and Yuan L (2022) Identification of key genes for hypertrophic cardiomyopathy using integrated network analysis of differential lncRNA and gene expression. Front. Cardiovasc. Med. 9:946229. doi: 10.3389/fcvm.2022.946229

Received: 17 May 2022; Accepted: 08 July 2022;
Published: 04 August 2022.

Edited by:

Yan Zhang, Peking University, China

Reviewed by:

Quanyi Zhao, Chinese Academy of Medical Sciences, China
Mao Zhang, Stanford University, United States

Copyright © 2022 Cao and Yuan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Lei Yuan, yuanlei33@csu.edu.cn

Download