2,341
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Identification of Potential Differentially-Methylated/Expressed Genes in Chronic Obstructive Pulmonary Disease

, , , , , , , & show all
Pages 44-54 | Received 31 Aug 2022, Accepted 08 Dec 2022, Published online: 19 Jan 2023

Abstract

Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory lung disease that causes obstructed airflow from the lungs. DNA methylation can regulate gene expression. Understanding the potential molecular mechanism of COPD is of great importance. The aim of this study was to find differentially methylated/expressed genes in COPD. DNA methylation and gene expression profiles in COPD were downloaded from the dataset, followed by functional analysis of differentially-methylated/expressed genes. The potential diagnostic value of these differentially-methylated/expressed genes was determined by receiver operating characteristic (ROC) analysis. Expression validation of differentially-methylated/expressed genes was performed by in vitro experiment and extra online datasets. Totally, 81 hypermethylated-low expression genes and 121 hypomethylated-high expression genes were found in COPD. Among which, 9 core hypermethylated-low expression genes (CD247, CCR7, CD5, IKZF1, SLAMF1, IL2RB, CD3E, CD7 and IL7R) and 8 core hypomethylated-high expression genes (TREM1, AQP9, CD300LF, CLEC12A, NOD2, IRAK3, NLRP3 and LYZ) were identified in the protein-protein interaction (PPI) network. Moreover, these genes had a potential diagnostic utility for COPD. Some signaling pathways were identified in COPD, including T cell receptor signaling pathway, cytokine-cytokine receptor interaction, hematopoietic cell lineage, HTLV-I infection, endocytosis and Jak-STAT signaling pathway. In conclusion, differentially-methylated/expressed genes and involved signaling pathways are likely to be associated with the process of COPD.

Introduction

Chronic obstructive pulmonary disease (COPD), a chronic inflammatory lung disease, causes obstructed airflow from the lungs [Citation1]. COPD is characterized by persistent airway inflammation and airflow limitation. In addition, patients with COPD suffer from other comorbidities/conditions, including weight loss, malnutrition, chronic heart failure, coronary artery disease, interstitial lung disease, pulmonary hypertension, aging, cancer, diabetes, metabolic syndrome, hypertension and hyperlipidemia [Citation2]. COPD is related to chronic inflammation characterized by an increased number of T lymphocytes and innate lymphoid cells recruited from the circulation [Citation3–5]. It is shown that adaptive immunity directed against lung self-antigens plays important roles in perpetuating the immune response [Citation6–9]. Some factors that affect the development of COPD have been identified such as genetic factors, sex, age, lung development, exposure to particles, asthma and airway hyperresponsiveness, socioeconomic status, infections and chronic bronchitis [Citation10–12]. It is noted that cigarette smoke is the most commonly risk factor for COPD [Citation10].

Clinically, the overall 5 year survival for patients with COPD is from 56% to 92%, which depends on the disease severity [Citation13]. Therefore, early diagnosis of COPD is necessary, before the occurrence of disability/irreversible lung structural changes [Citation14]. It is worth mentioning that DNA methylation can regulate gene expression. Characterizing methylation signatures could have important implications as biomarkers for early diagnosis and prognosis of COPD. The hypermethylation of SERPINA1 has been found in COPD, which could represent a potential biomarker for predicting the development of COPD in acute coronary syndrome patients [Citation15]. In addition, some differential methylated sites, closely associated with smoking, have potential significance in COPD susceptibility [Citation16,Citation17]. Many studies have confirmed that DNA methylation is a potential biomarker for diagnosis, prognosis and therapy of COPD [Citation18]. Thus it can be seen that DNA methylation may shed new insights into the pathogenesis of COPD. In the present study, we aimed to find potential differentially expressed genes (DEGs), differentially methylated genes (DMGs) and related signaling pathways in COPD patients by integrating high-throughput transcriptome expression data with DNA methylation data. Our study may be helpful in exploring the pathogenesis of COPD and providing a certain research basis for the prediction and drug development of COPD.

Methods

Filtering of data sets

Keywords of “chronic obstructive pulmonary disease” and “Homo sapiens” [porgn:__txid9606] were searched. Study types of data sets were "Expression profiling by array" and "Methylation Profiling by array". Detailed inclusion criteria of data sets were as follows: (1) the selected data set was genome-wide mRNA transcriptome data and DNA methylation data; (2) these data were obtained from peripheral blood mononuclear cell samples from COPD patients and normal controls; (3) both standardized and raw data sets were considered in this study. After screening, 1 mRNA data set (GSE42057) and 1 methylation data set (GSE118468) were obtained.

Identification of DEGs and DMGs

Data set GSE42057 was downloaded from Gene Expression Omnibus (GEO) database. Probes were corresponding to genes. The mean value of multiple probes corresponding to one gene was taken as the expression level of the gene. Difference analysis of gene was carried out by limMA package of analysis software R-4.0.5. The screening criterion of DEGs was p value < 0.05. CHAMP package was used for differential methylation analysis of genes under the selection criteria of p value < 0.05 and | deltaBeta | > 0.1. Intersection genes between down-regulated genes and hypermethylated genes were regarded as hypermethylated-low expression genes. In addition, intersection genes between up-regulated genes and hypomethylated genes were considered as hypomethylated-high expression genes. The volcano map and the heatmap were drawn using the ggplot2 package in R and the pheatmap package in R, respectively.

Functional analysis of hypermethylated-low expression and hypomethylated-high expression genes

Firstly, to explore the protein interaction between proteins encoded by hypermethylated-low expression/hypomethylated-high expression genes, protein-protein interaction (PPI) network was constructed through online database STRING. The setting in the analysis was combined_score > 0.15 and false discovery rate (FDR) < 0.05. The results obtained in the STRING database were imported Cytoscape software. CytoHubba plug-in was utilized to filter core genes. A total of 4 algorithms were adopted, including Maximal Clique Centrality (MCC), Maximum Neighborhood Component (MNC), Edge Percolated Component (EPC) and Degree. Core genes were screened after the intersection of the first 10 genes of each algorithm. Secondly, in order to study the function of hypermethylated-low expression and hypomethylated-high expression genes, the David database (background set: Homo sapiens) was used for GO and KEGG functional analysis under the screening criteria of p value < 0.05.

Diagnostic analysis of hypermethylated-low expression and hypomethylated-high expression genes

To study whether hypermethylated-low expression and hypomethylated-high expression genes have diagnostic value, the receiver operating characteristic (ROC) curve was used to determine the accuracy of key genes. The area under the curve (AUC), the evaluation index of model performance, under binomial exact confidence interval was calculated. The AUC value ranges 0-1, where 0.7 is acceptable performance and 0.9 is excellent performance. ROC curves were plotted using pROC packages of R language.

Expression validation of hypermethylated-low expression and hypomethylated-high expression genes by RT-PCR

To validate the expression of hypermethylated-low expression and hypomethylated-high expression genes, in vitro RT-PCR was performed. Totally, 14 COPD patients and 16 healthy individuals were enrolled in this study. Detailed inclusion and exclusion criteria of COPD patients were as follows: (1) patients were diagnosed as COPD based on the standard of Global Initiative for Chronic Obstructive Pulmonary Disease (2021 version) and had complete case data; (2) patients aged from 45 to 80 years old; (3) lung function test indicators of forced expiratory volume (FEV1)/forced vital capacity (FVC) < 70%; (4) repeat hospitalized patients were included in the first hospitalization data; (5) patients were conscious and stable with no obvious communication problems. The inclusion criteria for healthy controls were as follows: (1) normal individuals with normal lung function tests; (2) age of healthy individual was matched with COPD patient. The exclusion criteria for healthy controls were as follows: (1) individuals were complicated with other pulmonary diseases including lung cancer, bronchial asthma, pulmonary interstitial disease, active pulmonary tuberculosis, allergic bronchopulmonary mycosis, etc.; (2) individuals had hypertension, diabetes, serious heart, liver, kidney, hematopoietic system and nervous system diseases; (3) individuals had a history of cold, fever, lung or digestive tract infection in the past 1 week; (4) individuals were unable to comply with lung function examination and incomplete medical history. The study was approved by the ethics committee of Qingdao Fuwai Cardiovascular Hospital (2022-QF-005). In addition, all individuals provided the informed consent of the patients and their families.

The blood samples from the above individuals were collected for RT-PCR. The lysate was mixed with blood sample, shaken continuously for 30 s, and incubated at 15–30 °C for 10 min to completely decompose the ribosomes. The mixture was added with chloroform, shaken vigorously for 15 s, placed at room temperature for 5 min, and centrifuged at 12,000 rpm for 10 min at 4 °C. After removing supernatant, 70% of ethanol and buffer was orderly added and centrifuged at 12,000 rpm for 45 s, followed by adding RNase free water. Reverse transcription of the gene was performed using FastKing cDNA first-strand synthesis kit (TIANGEN). The relative quantitative analysis of the data was performed by 2-△△CT method using ABI 7300 fluorescence quantitative PCR instrument. GAPDH and ACTB were used as internal reference.

Expression validation of hypermethylated-low expression and hypomethylated-high expression genes in GSE94916 and GSE56767 datasets

To further validate the expression of hypermethylated-low expression and hypomethylated-high expression genes, GSE94916 dataset (involving peripheral blood mononuclear cell from 6 COPD cases and 6 normal controls) was used for analysis. The expression of these genes is presented as the box plot.

Results

Identification of hypermethylated-low expression and hypomethylated-high expression genes

Totally, 1592 DEGs were identified in COPD, including 479 up-regulated genes (such as heme binding protein 2, HEBP2) and 1113 down-regulated genes (such as nitric oxide synthase interacting protein, NOSIP) (Supplementary Table 1). In addition, a total of 5840 differentially methylated sites were obtained, involving 2634 DMGs (607 hypermethylated genes and 2027 hypomethylated genes) (Supplementary Table 2). The volcano plot and heat map of all DEGs and DMGs is presented in . Thus it can be seen that there is a significant difference of gene expression between COPD and normal controls. Among which, 81 hypermethylated-low expression genes and 121 hypomethylated-high expression genes were respectively identified between down-regulated genes and hypermethylated genes, and between up-regulated genes and hypomethylated genes ().

Figure 1. The volcano plot and heat map of all DEGs and DMGs. (a) The volcano plot of all DEGs; (b) the heat map of all DEGs; and (c) the volcano plot of all DMGs.

Figure 1. The volcano plot and heat map of all DEGs and DMGs. (a) The volcano plot of all DEGs; (b) the heat map of all DEGs; and (c) the volcano plot of all DMGs.

Figure 2. The Venn diagram of 81 hypermethylated-down-regulated genes (a) and 121 hypomethylated-up-regulated genes (b).

Figure 2. The Venn diagram of 81 hypermethylated-down-regulated genes (a) and 121 hypomethylated-up-regulated genes (b).

Table 1. Identification of 9 core hypermethylated-low expression genes.

Table 2. Identification of 8 core hypomethylated-high expression genes.

PPI network of hypermethylated-low expression and hypomethylated-high expression genes

shows the PPI network of 81 hypermethylated-low expression genes, which consists of 373 interacting gene pairs. A total of 9 core genes were identified after the intersection of the first 10 genes of each algorithm ( and ), including CD247 molecule (CD247) (expression value = −0.23729; p value = 0.007209; | deltaBeta | = 0.143968175), C-C motif chemokine receptor 7 (CCR7) (expression value = −0.42712; p value = 0.000283; | deltaBeta | = 0.123951118), CD5 molecule (CD5) (expression value = −0.17527; p value = 0.017409; | deltaBeta | = 0.125307121), IKAROS family zinc finger 1 (IKZF1) (expression value = −0.10353; p value = 0.006298; | deltaBeta | = 0.128153926), signaling lymphocytic activation molecule family member 1 (SLAMF1) (expression value = −0.18131; p value = 0.049751; | deltaBeta | = 0.132532141), interleukin 2 receptor subunit beta (IL2RB) (expression value = −0.2602; p value = 0.01408; | deltaBeta | = 0.121637175), CD3 epsilon subunit of T-cell receptor complex (CD3E) (expression value = −0.17105; p value = 0.017572; | deltaBeta | = 0.138873971), CD7 molecule (CD7) (expression value = −0.22309; p value = 0.004285; | deltaBeta | = 0.110875454) and interleukin 7 receptor (IL7R) (expression value = −0.22648; p value = 0.013863; | deltaBeta | = 0.155605074). shows the PPI network of 121 hypomethylated-high expression genes, which consist of 576 interacting gene pairs. Totally, 8 core genes were identified after the intersection of the first 10 genes of each algorithm (, ), including triggering receptor expressed on myeloid cells 1 (TREM1) (expression value = 0.385208; p value =; | deltaBeta | = −0.11855486), aquaporin 9 (AQP9) (expression value = 0.441979; p value = 9.69E-05; | deltaBeta | = −0.13560176), CD300 molecule like family member f (CD300LF) (expression value = 0.13993; p value = 0.02407; | deltaBeta | = −0.10395052), C-type lectin domain family 12 member A (CLEC12A) (expression value = 0.307716; p value = 0.01503; | deltaBeta | = −0.10492296), nucleotide binding oligomerization domain containing 2 (NOD2) (expression value = 0.145429; p value = 0.046749; | deltaBeta | = −0.1030296), interleukin 1 receptor associated kinase 3 (IRAK3) (expression value = 0.260752; p value = 0.005461; | deltaBeta | = −0.14495124), NLR family pyrin domain containing 3 (NLRP3) (expression value = 0.203008; p value = 0.047256; | deltaBeta | = −0.13672507) and lysozyme (LYZ) (expression value = 0.157934; p value = 0.02912; | deltaBeta | = −0.13213402).

Figure 3. PPI networks of 81 hypermethylated-low expression genes and 121 hypomethylated-high expression genes. (a) PPI network of 81 hypermethylated-low expression genes; (b) PPI network of 9 hypermethylated-low expression genes; (c) PPI network of 121 hypomethylated-high expression genes; (d) PPI network of 8 hypomethylated-high expression genes.

Figure 3. PPI networks of 81 hypermethylated-low expression genes and 121 hypomethylated-high expression genes. (a) PPI network of 81 hypermethylated-low expression genes; (b) PPI network of 9 hypermethylated-low expression genes; (c) PPI network of 121 hypomethylated-high expression genes; (d) PPI network of 8 hypomethylated-high expression genes.

Functional analysis of hypermethylated-low expression and hypomethylated-high expression genes

According to GO analysis of 81 hypermethylated-low expression genes, type I interferon signaling pathway, membrane and protein binding was the most significantly enriched biological process, cytological component and molecular function, respectively (). In the KEGG analysis, CD247 was involved in T cell receptor signaling pathway, CCR7 was involved in cytokine-cytokine receptor interaction, CD5, CD3E, CD7 and IL7R were involved in hematopoietic cell lineage, IL2RB was involved in HTLV-I infection, endocytosis and Jak-STAT signaling pathway (). Based on GO analysis of 121 hypomethylated-high expression genes, blood coagulation, plasma membrane and catalytic activity was the most significantly enriched biological process, cytological component and molecular function, respectively (). It is a pity that no enriched signaling pathways were found in these hypomethylated-high expression genes.

Figure 4. Functional analysis of 81 hypermethylated-low expression genes. (a) GO analysis; (b) KEGG analysis.

Figure 4. Functional analysis of 81 hypermethylated-low expression genes. (a) GO analysis; (b) KEGG analysis.

Figure 5. GO analysis of 121 hypomethylated-high expression genes.

Figure 5. GO analysis of 121 hypomethylated-high expression genes.

Diagnostic analysis of hypermethylated-low expression and hypomethylated-high expression genes

ROC curve analysis was carried out to assess the possible diagnostic utility of CD247, CCR7, CD5, IKZF1, SLAMF1, IL2RB, CD3E, CD7, IL7R, TREM1, AQP9, CD300LF, CLEC12A, NOD2, IRAK3, NLRP3, LYZ, HEBP2 and NOSIP (). The AUC values of these genes were all greater than 0.6, which indicates that they have a potential diagnostic utility for COPD.

Figure 6. The ROC curves of CD247, CCR7, CD5, IKZF1, SLAMF1, IL2RB, CD3E, CD7, IL7R, TREM1, AQP9, CD300LF, CLEC12A, NOD2, IRAK3, NLRP3, LYZ, HEBP2 and NOSIP in COPD.

Figure 6. The ROC curves of CD247, CCR7, CD5, IKZF1, SLAMF1, IL2RB, CD3E, CD7, IL7R, TREM1, AQP9, CD300LF, CLEC12A, NOD2, IRAK3, NLRP3, LYZ, HEBP2 and NOSIP in COPD.

Expression validation of hypermethylated-low expression and hypomethylated-high expression genes by RT-PCR

To validate the expression of randomly selected CD247, CCR7, IKZF1, CD7, AQP9, NOD2 and IRAK3, in vitro RT-PCR was performed in blood samples from 14 COPD patients and 16 healthy individuals (). The primer sequences of these genes are listed in Table 3. The clinical information of these individuals is listed in Supplementary Table 3. AQP9, NOD2 and IRAK3 were significantly up-regulated, CD247, CCR7 and CD7 were remarkably down-regulated. IKZF1 was down-regulated without statistical significance. In addition, further expression validation of CD247, CCR7, CD5, IKZF1, SLAMF1, CD3E, IL7R, AQP9, NOD2, IRAK3, and NOSIP was performed in GSE94916 dataset (). The result showed that AQP9, NOD2 and IRAK3 were up-regulated, CD247, CCR7, CD5, IKZF1, SLAMF1, CD3E, IL7R and NOSIP were down-regulated, which was consistent with bioinformatics analysis.

Figure 7. Expression validations of CD247, CCR7, IKZF1, CD7, AQP9, NOD2 and IRAK3 by RT-PCR. *p value < 0.05; **p value < 0.01; ***p value < 0.001.

Figure 7. Expression validations of CD247, CCR7, IKZF1, CD7, AQP9, NOD2 and IRAK3 by RT-PCR. *p value < 0.05; **p value < 0.01; ***p value < 0.001.

Figure 8. Expression validations of CD247, CCR7, CD5, IKZF1, SLAMF1, CD3E, IL7R, AQP9, NOD2, IRAK3, and NOSIP in GSE94916 dataset. *p value < 0.05; **p value < 0.01; ns: not significant.

Figure 8. Expression validations of CD247, CCR7, CD5, IKZF1, SLAMF1, CD3E, IL7R, AQP9, NOD2, IRAK3, and NOSIP in GSE94916 dataset. *p value < 0.05; **p value < 0.01; ns: not significant.

Discussion

CD247, involved in immune function, are associated with asthma, allergies and systemic sclerosis with pulmonary fibrosis [Citation19,Citation20]. In COPD patients, the expression of CD247 is down-regulated, which results in the immunosuppressive state and defective effector cell function [Citation21,Citation22]. The down-regulation of CD247 may account for the predisposition to recurrent airway infections in COPD patients [Citation23]. CCR7 is significantly down-regulated in pulmonary tuberculosis and COPD patients [Citation24–26]. CD5, a T cell receptor signaling gene, is down-regulated in bronchopulmonary dysplasia and COPD [Citation27,Citation28]. IKZF1, associated with autoimmune and allergic diseases (such as asthma/allergic rhinitis/chronic rhinosinusitis), is down-regulated in bronchopulmonary dysplasia [Citation27,Citation29,Citation30]. A susceptible genotype (rs17634369) near the IKZF1 gene is found in a general Japanese population of COPD [Citation31]. SLAMF1 can be found on activated B, T, and dendritic cells and monocytes [Citation32]. Altered activity of SLAMF1 is found in the dustmite-associated allergic asthma patients [Citation33].

IL2RB, a gene causing a predisposition to asthma, plays roles in the regulation of the epithelial barrier function and the adaptive and innate immune responses [Citation34]. The expression levels of IL2RB are decreased in peripheral blood from patients with COPD [Citation35]. CD3E is involved in T-cell receptor signaling pathway in end stage chronic respiratory diseases [Citation36]. CD7, belongs to the members of immunoglobulin super-family, is expressed in T cells and natural killer cells and plays an important role in the interaction between T cells [Citation37]. It is found that CD7 is down-regulated in peripheral blood mononuclear cells of COPD patients [Citation35]. IL7R, adaptive immunity related gene, is down-regulated in the asthmatic and COPD patients [Citation33,Citation35]. Single nucleotide polymorphism in IL7R is related to lung cancer risk in women with COPD [Citation38]. In this study, we found that CD247, CCR7, CD5, IKZF1, SLAMF1, IL2RB, CD3E, CD7 and IL7R were hypermethylated and down-regulated in COPD, which was in line with previous reports. Moreover, they had the potential diagnostic value for COPD. These genes may be involved in the immune regulation of COPD, which can be regarded as potential immune diagnostic target molecules for COPD.

TREM1 is up-regulated in chronic pulmonary aspergillosis patients and children with nonallergic asthma [Citation39,Citation40]. In fungal asthma, TREM1 plays roles in modulating the immune response directed against A. fumigates [Citation41]. AQP9 mainly expressed and play active roles in neutrophil volume and migration [Citation42]. Increased expression levels of AQP9 have been found in peripheral blood in patients with acute exacerbation of COPD [Citation35]. CD300LF, a gene codes for the inhibitory receptor of the Ig super-family of myeloid cells, is up-regulated in acute respiratory distress syndrome [Citation43]. It is noted that CD300LF is considered as a novel gene for COPD [Citation44]. CLEC12A is related to evaluation of COPD longitudinally [Citation45]. NOD2, expressed in airway epithelial cells, is involved in the clearance of bacterial pathogens [Citation46]. The relationship between NOD2 gene polymorphism results in the advancement of COPD [Citation47,Citation48]. The increases in the prevalence of the NOD2 rs1077861 single nucleotide polymorphism have been found in COPD patients [Citation48]. IRAK3 is involved in viral acute respiratory infections [Citation49]. Expressions of IRAK3 are elevated in patients with frequent exacerbations of COPD [Citation50]. In COPD patients, NLRP3 is over-expressed in the lung, which is associated with airflow obstruction [Citation51]. It is suggested that the activation of NLRP3 results in the occurrence of COPD airway inflammation [Citation52]. In addition, many different types of air pollutants can enhance the risk of COPD via regulating the NLRP3 inflammasome [Citation53]. Therefore, blockade of NLRP3 may be the possible therapeutic strategy for COPD [Citation54]. LYZ, an immune-related gene, has an important antibacterial activity in the airway epithelium. The expression of LYZ is found in allergic asthma [Citation55]. Additionally, LYZ is involved in the inflammatory response biological pathway in COPD [Citation56]. Herein, we found that TREM1, AQP9, CD300LF, CLEC12A, NOD2, IRAK3, NLRP3 and LYZ were hypomethylated and up-regulated in COPD. It is noted that these genes had the potential diagnostic value for COPD patients. Expression dysregulation of these genes may be associated with immune response and airway inflammation in COPD.

Beside above core genes in PPI network, we also found that HEBP2 and NOSIP were respectively one of top 10 up-regulated and down-regulated genes in COPD. HEBP2 is up-regulated in mouse lung telocytes as compared with mesenchymal stem cells and fibroblasts [Citation57]. NOSIP is differentially expressed in bronchopulmonary dysplasia [Citation58]. Decreased expression of NOSIP has been found in COPD [Citation28]. HEBP2 and NOSIP play important roles in COPD. In the KEGG analysis, we found that CD247 was involved in T cell receptor signaling pathway, CCR7 was involved in cytokine-cytokine receptor interaction, CD5, CD3E, CD7 and IL7R were involved in hematopoietic cell lineage, IL2RB was involved in HTLV-I infection, endocytosis and Jak-STAT signaling pathway. It is reported that COPD-specific down-regulated genes are involved in T cell receptor signaling pathway [Citation35]. In addition, down-regulation of T cell receptor results in the inadequate response to infection, causing increased susceptibility to COPD complications [Citation22]. Cytokine-cytokine receptor interaction is associated with fibrotic or pulmonary diseases and idiopathic pulmonary fibrosis [Citation59]. In peripheral blood mononuclear cells of COPD patients, cytokine-cytokine receptor interaction is a significantly enrichment pathway of dysregulated genes [Citation60]. In the lung tissue of COPD smokers, hematopoietic cell lineage is related to activation of metabolic pathways [Citation61]. HTLV-I infected lung epithelial cells produce cytokines and cell adhesion molecules, contributing to the clinical features of HTLV-I-related pulmonary diseases [Citation62]. Interestingly, HTLV-I infection is related to chronic thromboembolic pulmonary hypertension [Citation63]. Cigarette smoke can increase caveolin-mediated endocytosis, which could contribute to lung infection in smokers [Citation64]. The relationship between endocytosis and COPD has been found [Citation65]. In animal models, targeting Jak-STAT signaling pathway reduces airway hyperresponsiveness, leading to efforts to develop inhaled therapeutics targeting the Jak-STAT pathway for COPD [Citation66]. It is indicated that CD247, CCR7, CD5, CD3E, CD7, IL7R and IL2RB were involved in the development of COPD by participating above signaling pathways.

Conclusions

Some core differentially-methylated/expressed genes (CD247, CCR7, CD5, IKZF1, SLAMF1, IL2RB, CD3E, CD7, IL7R, TREM1, AQP9, CD300LF, CLEC12A, NOD2, IRAK3, NLRP3 and LYZ) and involved signaling pathways were identified in COPD. Significantly, the above genes had a potential diagnostic value for COPD. Our study may increase the understanding of the DNA methylation molecular mechanism of COPD. However, there are some limitations to our study. Firstly, the sample and validated gene number in the RT-PCR are small. Larger numbers of samples and more genes are further needed. Secondly, the expression of differentially methylated genes should be validated by bisulfite PCR. Thirdly, the potential molecular mechanism study lacked, and in vitro experiments are further needed.

Author contributions

Conception and design: Zongling Wang.

Administrative support: Zongling Wang.

Supply of materials and samples: Shuyuan An, Lina Dai and Shuo Xu.

Data collection and collation: Dan Liu and Lizhi Wang.

Data analysis and interpretation: Ruixue Zhang and Fengliang Wang.

Article writing: Wen Pan and Zongling Wang.

All authors approve and revise the manuscript.

Consent for publication

Not applicable.

Ethics approval and consent to participate

This study was performed in line with the principles of the Declaration of Helsinki and approved by the Medical Ethics Committee of Qingdao Fuwai Cardiovascular Hospital.

Patient consent for publication

Not applicable.

Supplemental material

Supplemental Material

Download Zip (774.8 KB)

Acknowledgements

None.

Availability of data and materials

Data are available in the article.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Decramer M, Janssens W, Miravitlles M. Chronic obstructive pulmonary disease. Lancet. 2012;379(9823):1341–1351. DOI:10.1016/S0140-6736(11)60968-9
  • Agusti A, Soriano JB. COPD as a systemic disease. COPD. 2008;5(2):133–138. DOI:10.1080/15412550801941349
  • Kurimoto E, Miyahara N, Kanehiro A, et al. IL-17A is essential to the development of elastase-induced pulmonary inflammation and emphysema in mice. Respir Res. 2013;14(1):5. DOI:10.1186/1465-9921-14-5
  • Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J Allergy Clin Immunol. 2016;138(1):16–27. DOI:10.1016/j.jaci.2016.05.011
  • Fujii U, Miyahara N, Taniguchi A, et al. IL-23 is essential for the development of elastase-induced pulmonary inflammation and emphysema. Am J Respir Cell Mol Biol. 2016;55(5):697–707. DOI:10.1165/rcmb.2016-0015OC
  • Cosio MG, Saetta M, Agusti A. Immunologic aspects of chronic obstructive pulmonary disease. N Engl J Med. 2009;360(23):2445–2454. DOI:10.1056/NEJMra0804752
  • Núñez B, Sauleda J, Antó JM, PAC-COPD Investigators, et al. Anti-tissue antibodies are related to lung function in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2011;183(8):1025–1031. DOI:10.1164/rccm.201001-0029OC
  • Bonarius HP, Brandsma CA, Kerstjens HA, et al. Antinuclear autoantibodies are more prevalent in COPD in association with low body mass index but not with smoking history. Thorax. 2011;66(2):101–107. DOI:10.1136/thx.2009.134171
  • Arnson Y, Shoenfeld Y, Amital H. Effects of tobacco smoke on immunity, inflammation and autoimmunity. J Autoimmun. 2010;34(3):J258–J265. DOI:10.1016/j.jaut.2009.12.003
  • Halpin DMG, Criner GJ, Papi A, et al. Global initiative for the diagnosis, management, and prevention of chronic obstructive lung disease. The 2020 GOLD science committee report on COVID-19 and chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2021;203(1):24–36. DOI:10.1164/rccm.202009-3533SO
  • Taniguchi A, Miyahara N, Oda N, et al. Protective effects of bisoprolol against acute exacerbation in moderate-to-severe chronic obstructive pulmonary disease. Acta Med Okayama. 2017;71(5):453–457.
  • Oda N, Miyahara N, Ichikawa H, et al. Long-term effects of beta-blocker use on lung function in Japanese patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2017;12:1119–1124. DOI:10.2147/COPD.S133071
  • Almagro P, Martinez-Camblor P, Soriano JB, et al. Finding the best thresholds of FEV1 and dyspnea to predict 5-year survival in COPD patients: the COCOMICS study. PLoS One. 2014;9(2):e89866. DOI:10.1371/journal.pone.0089866
  • Lowe KE, Regan EA, Anzueto A, et al. COPDGene(®) 2019: redefining the diagnosis of chronic obstructive pulmonary disease. J COPD F. 2019;6(5):384–399. DOI:10.15326/jcopdf.6.5.2019.0149
  • Rotondo JC, Aquila G, Oton-Gonzalez L, et al. Methylation of SERPINA1 gene promoter may predict chronic obstructive pulmonary disease in patients affected by acute coronary syndrome. Clin Epigenetics. 2021;13(1):79.
  • Qiu W, Baccarelli A, Carey VJ, et al. Variable DNA methylation is associated with chronic obstructive pulmonary disease and lung function. Am J Respir Crit Care Med. 2012;185(4):373–381. DOI:10.1164/rccm.201108-1382OC
  • Vucic EA, Chari R, Thu KL, et al. DNA methylation is globally disrupted and associated with expression changes in chronic obstructive pulmonary disease small airways. Am J Respir Cell Mol Biol. 2014;50(5):912–922. DOI:10.1165/rcmb.2013-0304OC
  • Sundar IK, Yin Q, Baier BS, et al. DNA methylation profiling in peripheral lung tissues of smokers and patients with COPD. Clin Epigenetics. 2017;9:38. DOI:10.1186/s13148-017-0335-5
  • Vicente CT, Revez JA, Ferreira MAR. Lessons from ten years of genome-wide association studies of asthma. Clin Transl Immunol. 2017;6(12):e165. DOI:10.1038/cti.2017.54
  • Zhao W, Yue X, Liu K, et al. The status of pulmonary fibrosis in systemic sclerosis is associated with IRF5, STAT4, IRAK1, and CTGF polymorphisms. Rheumatol Int. 2017;37(8):1303–1310. DOI:10.1007/s00296-017-3722-5
  • Kalathil SG, Lugade AA, Pradhan V, et al. T-regulatory cells and programmed death 1+ T cells contribute to effector T-cell dysfunction in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2014;190(1):40–50. DOI:10.1164/rccm.201312-2293OC
  • Grundy S, Plumb J, Lea S, et al. Down regulation of T cell receptor expression in COPD pulmonary CD8 cells. PLoS One. 2013;8(8):e71629. DOI:10.1371/journal.pone.0071629
  • Hurst JR, Vestbo J, Anzueto A, Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) Investigators, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363(12):1128–1138. DOI:10.1056/NEJMoa0909883
  • Li L, Lv J, He Y, et al. Gene network in pulmonary tuberculosis based on bioinformatic analysis. BMC Infect Dis. 2020;20(1):612. DOI:10.1186/s12879-020-05335-6
  • Rogers AV, Adelroth E, Hattotuwa K, et al. Bronchial mucosal dendritic cells in smokers and ex-smokers with COPD: an electron microscopic study. Thorax. 2008;63(2):108–114. DOI:10.1136/thx.2007.078253
  • Liao SX, Ding T, Rao XM, et al. Cigarette smoke affects dendritic cell maturation in the small airways of patients with chronic obstructive pulmonary disease. Mol Med Rep. 2015;11(1):219–225. DOI:10.3892/mmr.2014.2759
  • Shrestha D, Ye GX, Stabley D, et al. Pulmonary immune cell transcriptome changes in double-hit model of BPD induced by chorioamnionitis and postnatal hyperoxia. Pediatr Res. 2021;90(3):565–575. DOI:10.1038/s41390-020-01319-z
  • Zhang J, Liu J, Xu S, et al. Bioinformatics analyses of the pathogenesis and new biomarkers of chronic obstructive pulmonary disease. Medicine (Baltimore). 2021;100(46):e27737. DOI:10.1097/MD.0000000000027737
  • Laulajainen-Hongisto A, Lyly A, Hanif T, et al. Genomics of asthma, allergy and chronic rhinosinusitis: novel concepts and relevance in airway mucosa. Clin Transl Allergy. 2020;10(1):45. DOI:10.1186/s13601-020-00347-6
  • Kreiner E, Waage J, Standl M, et al. Shared genetic variants suggest common pathways in allergy and autoimmune diseases. J Allergy Clin Immunol. 2017;140(3):771–781. DOI:10.1016/j.jaci.2016.10.055
  • Muro S, Tabara Y, Matsumoto H, Nagahama Study Group, et al. Relationship among Chlamydia and Mycoplasma pneumoniae seropositivity, IKZF1 genotype and chronic obstructive pulmonary disease in a general Japanese population: the Nagahama study. Medicine (Baltimore). 2016;95(15):e3371. DOI:10.1097/MD.0000000000003371
  • Lin LT, Richardson CD. The host cell receptors for measles virus and their interaction with the viral hemagglutinin (H) Protein. Viruses. 2016;8(9):250. DOI:10.3390/v8090250
  • Whittle E, Leonard MO, Gant TW, et al. Multi-Method molecular characterisation of human Dust-Mite-associated allergic asthma. Sci Rep. 2019;9(1):8912. DOI:10.1038/s41598-019-45257-1
  • Ridolo E, Pucciarini F, Nizi MC, et al. Mabs for treating asthma: omalizumab, mepolizumab, reslizumab, benralizumab, dupilumab. Hum Vaccin Immunother. 2020;16(10):2349–2356. DOI:10.1080/21645515.2020.1753440
  • Wu X, Sun X, Chen C, et al. Dynamic gene expressions of peripheral blood mononuclear cells in patients with acute exacerbation of chronic obstructive pulmonary disease: a preliminary study. Crit Care. 2014;18(6):508. DOI:10.1186/s13054-014-0508-y
  • Chesné J, Danger R, Botturi K, COLT Consortium, et al. Systematic analysis of blood cell transcriptome in end-stage chronic respiratory diseases. PLoS One. 2014;9(10):e109291. DOI:10.1371/journal.pone.0109291
  • Sangle NA, Agarwal AM, Smock KJ, et al. Diffuse large B-cell lymphoma with aberrant expression of the T-cell antigens CD2 and CD7. Appl Immunohistochem Mol Morphol. 2011;19(6):579–583. DOI:10.1097/PAI.0b013e318221c672
  • Schwartz AG. Genetic epidemiology of cigarette smoke-induced lung disease. Proc Am Thorac Soc. 2012;9(2):22–26. DOI:10.1513/pats.201106-037MS
  • Smith NL, Hankinson J, Simpson A, et al. Reduced expression of TLR3, TLR10 and TREM1 by human macrophages in chronic cavitary pulmonary aspergillosis, and novel associations of VEGFA, DENND1B and PLAT. Clin Microbiol Infect. 2014;20(11):O960–8. DOI:10.1111/1469-0691.12643
  • Raedler D, Ballenberger N, Klucker E, et al. Identification of novel immune phenotypes for allergic and nonallergic childhood asthma. J Allergy Clin Immunol. 2015;135(1):81–91. DOI:10.1016/j.jaci.2014.07.046
  • Buckland KF, Ramaprakash H, Murray LA, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) modulates immune responses to Aspergillus fumigatus during fungal asthma in mice. Immunol Invest. 2011;40(7-8):692–722. DOI:10.3109/08820139.2011.578270
  • Karlsson T, Glogauer M, Ellen RP, et al. Aquaporin 9 phosphorylation mediates membrane localization and neutrophil polarization. J Leukoc Biol. 2011;90(5):963–973. DOI:10.1189/jlb.0910540
  • Grigoryev DN, Cheranova DI, Chaudhary S, et al. Identification of new biomarkers for acute respiratory distress syndrome by expression-based genome-wide association study. BMC Pulm Med. 2015;15:95. DOI:10.1186/s12890-015-0088-x
  • Maghsoudloo M, Azimzadeh Jamalkandi S, Najafi A, et al. Identification of biomarkers in common chronic lung diseases by co-expression networks and drug-target interactions analysis. Mol Med. 2020;26(1):9.
  • McDonald ML, Cho MH, Sørheim IC, Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints and COPDGene Investigators, et al. Common genetic variants associated with resting oxygenation in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2014;51(5):678–687. DOI:10.1165/rcmb.2014-0135OC
  • Uehara A, Fujimoto Y, Fukase K, et al. Various human epithelial cells express functional toll-like receptors, NOD1 and NOD2 to produce anti-microbial peptides, but not proinflammatory cytokines. Mol Immunol. 2007;44(12):3100–3111. DOI:10.1016/j.molimm.2007.02.007
  • Adler J, Rangwalla SC, Dwamena BA, et al. The prognostic power of the NOD2 genotype for complicated Crohn’s disease: a meta-analysis. Am J Gastroenterol. 2011;106(4):699–712. DOI:10.1038/ajg.2011.19
  • Kinose D, Ogawa E, Hirota T, et al. A NOD2 gene polymorphism is associated with the prevalence and severity of chronic obstructive pulmonary disease in a Japanese population. Respirology. 2012;17(1):164–171. DOI:10.1111/j.1440-1843.2011.02069.x
  • Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 2014;42(Database issue):D68–73. DOI:10.1093/nar/gkt1181
  • Baines KJ, Fu JJ, McDonald VM, et al. Airway gene expression of IL-1 pathway mediators predicts exacerbation risk in obstructive airway disease. Int J Chron Obstruct Pulmon Dis. 2017;12:541–550. DOI:10.2147/COPD.S119443
  • Faner R, Sobradillo P, Noguera A, et al. The inflammasome pathway in stable COPD and acute exacerbations. ERJ Open Res. 2016;2(3):00002-2016. DOI:10.1183/23120541.00002-2016
  • Yang Y, Huang L, Tian C, et al. Magnesium isoglycyrrhizinate inhibits airway inflammation in rats with chronic obstructive pulmonary disease. BMC Pulm Med. 2021;21(1):371. DOI:10.1186/s12890-021-01745-7
  • Zhang J, Xu Q, Sun W, et al. New insights into the role of NLRP3 inflammasome in pathogenesis and treatment of chronic obstructive pulmonary disease. JIR. 2021;14:4155–4168. DOI:10.2147/JIR.S324323
  • Yang W, Ni H, Wang H, et al. NLRP3 inflammasome is essential for the development of chronic obstructive pulmonary disease. Int J Clin Exp Pathol. 2015;8(10):13209–13216.
  • Sun X, Hou T, Cheung E, et al. Anti-inflammatory mechanisms of the novel cytokine interleukin-38 in allergic asthma. Cell Mol Immunol. 2020;17(6):631–646. DOI:10.1038/s41423-019-0300-7
  • Sharma A, Kitsak M, Cho MH, et al. Integration of molecular interactome and targeted interaction analysis to identify a COPD disease network module. Sci Rep. 2018;8(1):14439. DOI:10.1038/s41598-018-32173-z
  • Zheng Y, Zhang M, Qian M, et al. Genetic comparison of mouse lung telocytes with mesenchymal stem cells and fibroblasts. J Cell Mol Med. 2013;17(4):567–577. DOI:10.1111/jcmm.12052
  • Pietrzyk JJ, Kwinta P, Wollen EJ, et al. Gene expression profiling in preterm infants: new aspects of bronchopulmonary dysplasia development. PLoS One. 2013;8(10):e78585. DOI:10.1371/journal.pone.0078585
  • Karatzas E, Bourdakou MM, Kolios G, et al. Drug repurposing in idiopathic pulmonary fibrosis filtered by a bioinformatics-derived composite score. Sci Rep. 2017;7(1):12569. DOI:10.1038/s41598-017-12849-8
  • Dang X, Qu X, Wang W, et al. Bioinformatic analysis of microRNA and mRNA regulation in peripheral blood mononuclear cells of patients with chronic obstructive pulmonary disease. Respir Res. 2017;18(1):4. DOI:10.1186/s12931-016-0486-5
  • Bi H, Zhou J, Wu D, et al. Microarray analysis of long non-coding RNAs in COPD lung tissue. Inflamm Res. 2015;64(2):119–126. DOI:10.1007/s00011-014-0790-9
  • Teruya H, Tomita M, Senba M, et al. Human T-cell leukemia virus type I infects human lung epithelial cells and induces gene expression of cytokines, chemokines and cell adhesion molecules. Retrovirology. 2008;5(1):86. DOI:10.1186/1742-4690-5-86
  • Miao R, Dong X, Gong J, et al. Hsa-miR-106b-5p participates in the development of chronic thromboembolic pulmonary hypertension via targeting matrix metalloproteinase 2. Pulm Circ. 2020;10(3):2045894020928300. DOI:10.1177/2045894020928300
  • Duffney PF, Embong AK, McGuire CC, et al. Cigarette smoke increases susceptibility to infection in lung epithelial cells by upregulating caveolin-dependent endocytosis. PLoS One. 2020;15(5):e0232102. DOI:10.1371/journal.pone.0232102
  • Halu A, Liu S, Baek SH, et al. Exploring the cross-phenotype network region of disease modules reveals concordant and discordant pathways between chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Hum Mol Genet. 2019;28(14):2352–2364. DOI:10.1093/hmg/ddz069
  • Barnes PJ. Kinases as novel therapeutic targets in asthma and chronic obstructive pulmonary disease. Pharmacol Rev. 2016;68(3):788–815. DOI:10.1124/pr.116.012518