Next Article in Journal
Deciphering the Association: Critical HDL-C Levels and Their Impact on the Glycation Gap in People Living with HIV
Next Article in Special Issue
miRNA Signatures in Alveolar Macrophages Related to Cigarette Smoke: Assessment and Bioinformatics Analysis
Previous Article in Journal
Preimplantation Genetic Testing (PGT) to Reduce the Risk for GBA-Related Parkinson’s Disease: Expanding the Applications for Embryo Selection
Previous Article in Special Issue
mirSNPs as Potential Colorectal Cancer Biomarkers: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Identification and Evaluation of Hub Long Non-Coding RNAs and mRNAs in PM2.5-Induced Lung Cell Injury

by
Jing Sui
1,2,
Yanni Zhang
1,
Linjie Zhang
1 and
Hui Xia
2,*
1
Research Institute for Environment and Health, Nanjing University of Information Science and Technology, Nanjing 210044, China
2
Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(3), 911; https://doi.org/10.3390/ijms26030911
Submission received: 31 December 2024 / Revised: 17 January 2025 / Accepted: 20 January 2025 / Published: 22 January 2025
(This article belongs to the Special Issue Roles and Mechanisms of Non-Coding RNAs in Human Health and Disease)

Abstract

:
Exposure to air pollution, especially fine particulate matter (PM2.5), is closely linked to various adverse health effects, particularly in the respiratory system. The present study was designed to investigate the lncRNA–mRNA interactions in PM2.5-induced lung cell injury using weighted gene co-expression network analysis (WGCNA). We downloaded the gene expression data of GSE138870 from the Gene Expression Omnibus (GEO) database and screened for differentially expressed lncRNAs and mRNAs. We constructed co-expression modules with WGCNA. Furthermore, functional enrichment analysis was also performed. We also constructed lncRNA–mRNA co-expression networks and lncRNA–mRNA-pathway networks to identify key regulatory relationships. The results revealed several modules significantly correlated with PM2.5-induced lung injury, such as the turquoise and blue modules. Genes within these modules were enriched in pathways related to signal transduction, metabolism, and cancer. Hub lncRNAs in the turquoise module, including LOC100129034 and CROCCP2, were found to be co-expressed with mRNAs involved in apoptosis and proliferation regulation. In the blue module, lnc-CLVS2-2 and GARS1-DT were connected to genes related to cell migration, invasion, and lung injury. These findings contribute novel perspectives to the molecular mechanisms involved in PM2.5-induced lung injury and suggest that WGCNA could be a valuable tool for predicting and understanding this disease process.

1. Introduction

Exposure to air pollution significantly impacts human health, correlating with several health conditions such as immune, respiratory, cardiovascular, nervous, and metabolic diseases [1,2,3,4]. In 2013, IARC listed air pollutants as Class I human carcinogens [5]. Functionally, the lungs are linked directly with the external environment via the upper respiratory tract, with a large respiratory surface area. Research consistently indicates that the respiratory system, as the initial barrier against external environmental threats, shows heightened susceptibility to pollutants, including particulate matter, volatile organic compounds, sulfur dioxide, and nitrogen dioxide [6].
Fine particulate matter (PM2.5) is globally recognized as a major air pollutant that poses a serious threat to public health. Research has indicated that PM2.5 pollution has become the fourth leading risk factor for health, in addition to high blood pressure, smoking, and poor diet [7]. PM2.5, with an aerodynamic diameter of ≤2.5 μm, is susceptible to deposition in the alveoli, inducing oxidative stress, inflammation, epithelial barrier dysfunction, and epigenetic changes [8]. PM2.5 exposure has been correlated with a variety of adverse pulmonary outcomes. Short-term exposure to PM2.5 is known to impair pulmonary expiratory flow rates and induce significant lung inflammation [9,10]. Li et al. [11] demonstrated that short-term exposure to PM2.5 induces inflammation and epithelial alterations, which were subsequently resolved, with macrophages, neutrophils, and type II alveolar epithelial cells playing pivotal roles in the repair process. Furthermore, prolonged exposure to PM2.5 exacerbates the risk of developing severe respiratory disorders, including lung injury, pulmonary fibrosis, asthma exacerbation, chronic obstructive pulmonary disease (COPD), and lung cancer. These chronic conditions significantly elevate the incidence and mortality related to respiratory diseases [12,13,14,15]. Epidemiological studies have confirmed that air pollution raises the incidence of lung cancer, and PM2.5 is an independent risk factor for lung cancer [16,17].
Long non-coding RNAs (lncRNAs), which are longer than 200 nucleotides, play diverse roles in cellular processes [18]. They participate in mRNA splicing, maturation, transport, localization, and stabilization [19]. Functioning as significant multi-dimensional modifiers, lncRNAs contribute extensively to translation regulation, post-transcriptional modifications, and epigenetic modulation [19]. A growing body of research suggests that lncRNAs play a role in critical biological processes associated with various pulmonary diseases [20,21]. lncRNA PCAT1 activates SOX2 to inhibit cGAS/STING signaling-mediated T-cell activation and promote tumorigenesis and immunosuppression [22]. lncRNA H19 alleviates sepsis-induced acute lung injury by downregulating the expression of TNF-α, IL-6, IL-17, caspase-3, caspase-9, and Bax while upregulating Bcl-2 levels, thereby suppressing lung cell apoptosis and inflammation [23]. Studies have demonstrated that predicting lncRNA functions using co-expression networks of lncRNAs and mRNAs is advantageous for advancing research on PM2.5-induced lung injury [24,25].
Weighted gene co-expression network analysis (WGCNA) is an innovative algorithm designed to identify modules of co-expressed genes from gene expression profiling data [26]. This method elucidates the interactive mechanisms of genes, including hub lncRNAs and mRNAs, and establishes correlations among highly co-expressed modules. Despite its application in elucidating genes interactions in lung cancer [27,28,29], there is a notable absence of studies employing WGCNA to construct lncRNA–mRNA co-expression networks for investigating lung injury. Our study aims to investigate lncRNA–mRNA interactions in lung injury induced by PM2.5. The key lncRNAs and mRNAs were identified from Gene Expression Omnibus (GEO) (GSE138870). Employing WGCNA, we extracted modules of co-expressed lncRNAs and mRNAs and predicted their targeted interactions. Following the construction of the lncRNA–mRNA network, we explored lncRNA regulation of mRNAs in pathways. This analysis is expected to enhance our knowledge of lncRNA functions and their potential therapeutic implications in lung injury.

2. Results

2.1. Weighted Co-Expression Network Construction and Key Module Identification

Analysis of nine samples from the GSE138870 dataset revealed 1773 differentially expressed lncRNAs with fold change > 2 and 3381 mRNAs with fold change > 1.5 (Figure 1). The cutoff for lncRNAs was set at fold change > 2 to focus on the most robustly differentially expressed lncRNAs, while a slightly lower cutoff of fold change > 1.5 was used for mRNAs to capture a broader range of differentially expressed genes. These thresholds were chosen to ensure the most effective use of the data, balancing sensitivity and specificity. To examine the expression profiles of all lncRNAs and mRNAs, hierarchical clustering was performed to identify potential outlier samples. The results showed that there was no outlier sample (Figure 2a). Then, hierarchical clustering dendrograms were constructed to identify various modules (Figure 2b). Analysis of scale independence and mean connectivity resulted in the determination of a soft threshold power (β) value of 16 (Figure 2c). We obtained 12 modules with different expression trends (turquoise, blue, brown, yellow, green, red, black, pink, magenta, purple, green-yellow, and grey) (Table 1). The module relationships were visualized using a eigengene adjacency heatmap cluster (Figure 2d).

2.2. Correlation Analysis of Module and Traits

Figure 2e illustrates the association between co-expression modules and PM2.5 interventions. The red module demonstrated the highest positive correlation, with 500 μg/mL PM2.5 (correlation coefficient = 0.87, p-value = 0.002), while the turquoise module displayed the strongest negative correlation (correlation coefficient = −0.99, p-value = 3 × 107). Moreover, we observed that green-yellow module exhibited the strongest positive correlation, with 100 μg/mL PM2.5 (correlation coefficient = 0.67, p-value = 0.05), while the black module showed the most pronounced negative correlation (correlation coefficient = −0.94, p-value = 2 × 104). Based on the correlation between the modular genes and lung injury, as well as the number of lncRNAs and mRNAs, the turquoise, blue, brown, yellow, and green modules were selected for further analysis.

2.3. GO Enrichment Analysis and Pathway Enrichment Analysis

The GO enrichment analysis showed that the differentially expressed genes in the turquoise module were mainly enriched in signal transduction, positive regulation of transcription by RNA polymerase II, regulation of transcription by RNA polymerase II, intracellular signal transduction, and positive regulation of the apoptotic process (Figure 3a and Table 2). The results of GO enrichment analysis in blue modules were mainly enriched in signal transduction, regulation of transcription by RNA polymerase II, chemical synaptic transmission, G protein-coupled receptor signaling pathway, and negative regulation of transcription by RNA polymerase II (Figure 3b and Table 2). The GO enrichment of brown, yellow, and green modules is shown in Supplementary Figure S1.
KEGG pathway enrichment analysis revealed that the differentially expressed genes were primarily involved in metabolic pathways, pathways in cancer, neuroactive ligand–receptor interaction, Huntington disease, and tuberculosis in the turquoise module (Figure 3c and Table 3). The results of pathway enrichment in the blue modules were mainly related to metabolic pathways, calcium signaling pathway, cAMP signaling pathway, neuroactive ligand–receptor interaction, GnRH signaling pathway (Figure 3d and Table 3). The pathway enrichment of the brown, yellow, and green modules is shown in Supplementary Figure S1.

2.4. Construction of lncRNA–mRNA Co-Expression Networks

lncRNA–mRNA co-expression networks were constructed for the functional prediction of lncRNA in the various modules (Figure 4). In the turquoise module, 25 mRNAs and 16 lncRNAs were identified as hub genes (Figure 4a), and 25 mRNAs and 19 lncRNAs as hub genes were detected in blue module (Figure 4b). Hub genes in the brown, yellow, and green modules are presented in Supplementary Figure S2. Previous research has shown that lncRNAs play an important role in disease development [30]. We found that several lncRNAs were closely associated with hub mRNAs in the co-expression network. It can be predicted that the lncRNA of unknown function at one end of the line has a similar function as the gene at the other end of the line. Hence, we can make inferences about the functions of lncRNAs on the basis of the functions of mRNAs.

2.5. Construction of lncRNA–mRNA-Pathway Network

To identify potential mechanisms of lncRNA-mediated regulation of signaling pathways, we constructed the lncRNA-mRNA-pathway network through interaction of significantly different pathways and lncRNA–mRNA co-expression networks (Figure 4 and Supplementary Figure S2). In the turquoise module, there were 10 lncRNAs and 7 mRNAs in the pathway network (Figure 4c). NR_027406 (Alias: LOC100129034, a lncRNA) was linked to three mRNAs (INPP4A, PBX1, VPS37A, TPSAB1, SSX1, and PLIN2) and enriched in metabolic pathways, phosphatidylinositol signaling system, Cushing syndrome, transcriptional mis-regulation in cancer, endocytosis, Influenza A, transcriptional mis-regulation in cancer, and PPAR signaling pathway. uc009vov.2 (Alias: CROCCP2, a lncRNA) was related to six mRNAs (INPP4A, PBX1, PLIN2, VPS37A, BAX, and TPSAB1) and abundant in 54 signaling pathways, such as metabolic pathways, transcriptional mis-regulation in cancer, PPAR signaling pathway, endocytosis, pathways in cancer, apoptosis, non-small-cell lung cancer, EGFR tyrosine kinase inhibitor resistance, non-alcoholic fatty liver disease, endocrine resistance, longevity-regulating pathway, apoptosis—multiple species, small-cell lung cancer, p53 signaling pathway, protein processing in endoplasmic reticulum, and basal cell carcinoma.
In the pathway network of the blue module (Figure 4d), TCONS_00011945 (Alias: lnc-CLVS2-2, a lncRNA) was linked to seven mRNAs (ITGA2, ADH1B, ATG16L1, ISY1, ZNF300, GPR161, and RDH11) and enriched in 20 signaling pathways, such as platelet activation, pathways in cancer, proteoglycans in cancer, focal adhesion, PI3K-Akt signaling pathway, metabolic pathways, glycolysis/gluconeogenesis, chemical carcinogenesis, tyrosine metabolism, autophagy—animal, and NOD-like receptor signaling pathway. uc003tbj.2 (Alias: GARS1-DT, a lncRNA) was also linked to six mRNAs (ITGA2, ADH1B, ZNF300, RDH11, ISY1, and ATG16L1) and abundant in 19 signaling pathways, such as platelet activation, pathways in cancer, proteoglycans in cancer, focal adhesion, phagosome, human papillomavirus infection, PI3K-Akt signaling pathway, dilated cardiomyopathy, regulation of actin cytoskeleton, and metabolic pathways.

2.6. Construction of Protein–Protein Interaction Network

To investigate the interactions among hub genes within each module, we used the STRING database to construct protein–protein interaction (PPI) networks (Figure 5). The PPI network for the turquoise module is shown in Figure 5a, comprising 14 key genes identified through PPI analysis. Similarly, the PPI network for the blue module is presented in Figure 5b, with 10 crucial genes identified within this module based on the PPI analysis.

3. Discussion

Recent studies have provided growing evidence that exposure to PM2.5 is associated with a significant increase in lung injury and a decline in lung function [31]. Genomic studies have identified several genes implicated in PM2.5-induced lung injury [14,32,33]. However, the underlying mechanisms of PM2.5-induced lung injury remain unclear. WGCNA was applied for the first time to explore the relationship between PM2.5 exposure and lung cell injury. Using the GEO database (GSE138870 dataset), we identified a total of 3391 mRNAs and 1773 lncRNAs. Through WGCNA analysis, we categorized these lncRNAs and mRNAs into 12 modules with diverse functionalities, revealing five modules (turquoise, blue, brown, yellow, and green) closely associated with lung injury. Additionally, functional enrichment analysis was performed to investigate the potential biological impacts of lncRNAs and mRNAs in lung cell injury. The lncRNA–mRNA co-expression network provided novel insights into the interplay between lncRNAs and mRNAs. Furthermore, we constructed a lncRNA–mRNA-pathway co-expression network and identified positive associations with metabolic pathways, calcium signaling pathway, pathways in cancer, cAMP signaling pathway, MAPK signaling pathway, and other signaling pathways. These findings suggest that lung cell injuries may arise from disruptions in multiple signaling pathways, underscoring the regulatory role of PM2.5 in lung cell injury.
In contrast to the identification of differentially expressed genes, WGCNA utilizes large-scale gene expression data to delineate co-expression modules and conduct phenotype association studies [34]. Hub genes within WGCNA modules play significant regulatory roles in various biological processes [35,36]. Consequently, investigating hub genes in modules becomes increasingly crucial for elucidating the molecular mechanisms underlying lung cell injury. In our study, we especially examined the hub genes within the turquoise and blue modules. Within these modules, numerous hub mRNAs were explored to positively regulate critical biological processes, such as metabolism, cell cycle regulation, apoptosis, oxidative stress response, and inflammation. These findings suggest that perturbations in metabolic pathways, calcium signaling pathway, pathways in cancer, cAMP signaling pathway, and MAPK signaling pathway, which are mediated by the concerted actions of multiple genes, may contribute to the pathogenesis of lung cell injury.
LncRNAs can interact with proteins, RNA, and even DNA, mediating gene regulation by acting as signals to stimulate or inhibit transcription processes. As lncRNAs post-transcriptionally regulate mRNAs, predicting their interactions and exploring related mechanisms in pathological processes through regulatory networks is essential [19]. Jiang et al. [37] characterized the lncRNA–mRNA regulatory networks in PM2.5-induced lung cancer with expression profile data and identified the potential therapeutic target, the LCPAT1-RCC2 axis. Furthermore, PM2.5 can be internalized by lung cancer cells, resulting in significant increases in reactive oxygen species (ROS) levels, along with upregulation of loc146880 and LC3B expression, thereby promoting autophagy [38]. In the present study, multiple novel PM2.5-regulated lncRNA–mRNA networks were identified, providing valuable insights into the mechanisms underlying PM2.5 exposure and lung cell injury.
In the turquoise module, LOC100129034, CROCCP2, ENST00000424391 (Alias: MYO3B-AS1), TCONS_00002241 (Alias: linc-SRP9-1), and TCONS_00016745 were key lncRNAs linked to hub mRNAs (BAX, INPP4A, PBX1, PLIN2, VPS37A, SSX1, and TPSAB1). BAX (BCL2 associated X) plays a pivotal role as an effector molecule in the process of mitochondria-dependent programed cell death [39,40]. An in vivo study showed that PM2.5 significantly increased the expression of BAX and induced mitochondrial damage and cell apoptosis [32] to enhance lung injury [41]. In sepsis-induced acute lung injury, upregulation of lncRNA H19 inhibited TNF-α, IL-1β, IL-6, IL-10, and BAX to suppress pulmonary apoptosis and inflammation [23,42]. In addition, Xu et al. reported that LINC01089 interacted with the miR-543/BAX axis to inhibit lung adenocarcinoma cell proliferation and promoted apoptosis [43]. Elevated levels of lncRNA GHRLOS significantly inhibited cancer cell proliferation and invasion while promoting apoptosis through the modulation of E-cadherin, N-cadherin, BAX, and Bcl-2 expression in non-small-cell lung cancer [44]. Therefore, existing evidence suggests that INPP4A is correlated with airway inflammation and remodeling and plays a crucial role in maintaining lung homeostasis [45,46]. It has been demonstrated previously that the other hub mRNAs (PBX1, PLIN2, and TPSAB1) are closely related to the progression of lung cancer and COPD by regulating the cell cycle and cell proliferation [47,48,49]. Consequently, we postulated that the key lncRNAs, LOC100129034, CROCCP2, MYO3B-AS1, linc-SRP9-1, and TCONS_00016745, which were co-expressed with BAX, INPP4A, PBX1, PLIN2, VPS37A, SSX1, and TPSAB1 in our WGCNA analysis, might play a role in apoptosis and proliferation modulation during PM2.5-induced lung cell injury.
In the blue module, lnc-CLVS2-2 and GARS1-DT were the core lncRNAs connected to hub genes (ITGA2, ADH1B, ATG16L1, ISY1, ZNF300, GPR161, and RDH11). Recent results have indicated that ITGA2 activated the FAK-RAC1-PAK signaling pathway to participate in the formation of the cytoskeleton in lung adenocarcinoma cells and then promoted migration and invasion [50]. In the process of methotrexate-induced adverse reactions, such as serious lung injury, ITGA2 plays a key role in the epithelial–mesenchymal transition (EMT) [51]. The ATG16L1 protein was found to be associated with lung injury and inflammation levels in mice with Pseudomonas aeruginosa lung infection and LPS-induced acute lung injury [52,53]. Additionally, ADH1B, ZNF300, GPR161, and RDH11 were presented as the novel regulatory genes in lung injury and lung cancer [54,55,56,57]. We predict that lnc-CLVS2-2 and GARS1-DT might interact with ITGA2, ADH1B, ATG16L1, ISY1, ZNF300, GPR161, and RDH11 in the process of PM2.5-induced lung cell injury through the pathways in cancer, proteoglycans in cancer, focal adhesion, PI3K-Akt signaling pathway, metabolic pathways, chemical carcinogenesis, and PI3K-Akt signaling pathways.

4. Materials and Methods

4.1. Data Retrieving and Processing

The expression profiling of GSE138870, which is identified on the GPL16956 platform (Agilent-045997 Arraystar human lncRNA microarray V3 (Probe Name Version)), was downloaded from the GEO database (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE138870, accessed on 14 March 2023). GSE138870 contained nine HBE cell samples from three normal cells, three HBE cells exposed to 100 μg/mL PM2.5, and three HBE cells exposed to 500 μg/mL PM2.5. HIST2 and Stringtie were used to re-align the human standard genome and gene annotation [58]. Significant differentially expressed lncRNAs and mRNAs between the control and exposure groups were screened by R software (version 4.1.1) with the screening criteria (fold change > 2 or >1.5, p < 0.05, false discovery rate (FDR) < 0.05). The flow chart of this research is shown in Figure 6.

4.2. Weighted Gene Co-Expression Network Analysis (WGCNA)

WGCNA is used to find highly co-expressed gene modules and associations between genomes and diseases [59]. We used the R package “WGCNA” to evaluate the availability of genes and construct the gene co-expression network. The Person coefficient was calculated for the strength of correlation between any two genes [60]. Hierarchical cluster trees were created based on the correlation coefficients between genes, with distinct branches representing different gene modules. Genes were classified according to their expression patterns using weighted correlation coefficients, and those with similar patterns were grouped into modules.

4.3. Gene Ontology and Pathway Enrichment Analysis

The functional annotation of mRNAs in each module was analyzed using the Gene Ontology (GO) database. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database is a public pathway database to perform pathway enrichment analysis. Fisher’s exact test and multiple comparison tests were employed to calculate the p-value and FDR. p < 0.05 and FDR < 0.05 indicated significant differences.

4.4. Construction of lncRNA–mRNA Co-Expression Networks

The hub genes were defined by higher connectivity in the module. Hub genes with high connectivity were upstream regulators, while low-connectivity genes were downstream regulators. Co-expression relationships between hub genes were calculated, and lncRNA–mRNA co-expression and mRNA–mRNA co-expression were selected to construct a co-expression network.

4.5. Construction of lncRNA–mRNA-Pathway Co-Expression Network

In order to construct an lncRNA–mRNA pathway net, the regulatory relationship between lncRNAs and mRNAs was detected, as well as the significant pathways involved in mRNA regulation. The primary objective of this study was to reveal lncRNA-regulated signaling pathways to predict potential diseases associated with lncRNAs.

4.6. Protein–Protein Interaction (PPI) Network Construction

PPI network analysis was conducted using the online tool STRING (version 11.5; https://cn.string-db.org/, accessed on 27 December 2024) [61]. In our study, both lncRNAs and mRNAs within the identified modules were subjected to analysis using STRING.

4.7. Statistical Analysis

Statistical analysis was conducted using R software (Version 4.1.1) and SPSS (Version 21.0). Statistical comparisons between groups of normalized data were performed with Student’s t test or one-way analysis of variance. p < 0.05 was considered to be significant.

5. Conclusions

In conclusion, based on WGCNA, we identified PM2.5-induced lung cell injury-related candidate hub lncRNAs and mRNAs, and we constructed both lncRNA–mRNA co-expression networks and lncRNA–mRNA pathway co-expression networks. The results indicated that the hub lncRNAs may regulate the inflammatory, metabolic, and cancer pathways, which are critically involved in the pathogenesis of lung cell injury. Moreover, WGCNA analysis showed potential as a novel predictive tool for PM2.5-induced lung cell injury. Therefore, future research should focus on investigating the regulatory roles of hub lncRNAs and mRNAs in PM2.5-induced lung cell injury.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms26030911/s1.

Author Contributions

Conceptualization, J.S. and H.X.; methodology, J.S.; software, J.S.; validation, Y.Z., L.Z. and H.X.; formal analysis, J.S.; data curation, J.S.; writing—original draft preparation, J.S.; writing—review and editing, H.X.; supervision, H.X.; project administration, H.X.; funding acquisition, H.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Key R&D Program of China (2023YFF1104402), the China Postdoctoral Science Foundation funded project (2022M720714), Innovation and Entrepreneurship Program of Jiangsu Province (JSS-CBS20210472).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available in a publicly accessible repository.

Acknowledgments

As an invited researcher with the Institute of Climate Change and Public Policy, I would like to thank the institute for its constructive comments and support of the authors.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
PM2.5Fine particulate matter
WGCNAWeighted gene co-expression network analysis
GEOGene Expression Omnibus
COPDChronic obstructive pulmonary disease
lncRNAsLong non-coding RNAs
ROSReactive oxygen species
FDRFalse discovery rate

References

  1. Zhang, K.; Brook, R.D.; Li, Y.; Rajagopalan, S.; Kim, J.B. Air Pollution, Built Environment, and Early Cardiovascular Disease. Circ. Res. 2023, 132, 1707–1724. [Google Scholar] [CrossRef]
  2. Sin, D.D.; Doiron, D.; Agusti, A.; Anzueto, A.; Barnes, P.J.; Celli, B.R.; Criner, G.J.; Halpin, D.; Han, M.K.; Martinez, F.J.; et al. Air pollution and COPD: GOLD 2023 committee report. Eur. Respir. J. 2023, 61, 2202469. [Google Scholar] [CrossRef]
  3. Palacios, N. Air pollution and Parkinson’s disease—Evidence and future directions. Rev. Environ. Health 2017, 32, 303–313. [Google Scholar] [CrossRef] [PubMed]
  4. Liu, C.X.; Liu, Y.B.; Peng, Y.; Peng, J.; Ma, Q.L. Causal effect of air pollution on the risk of cardiovascular and metabolic diseases and potential mediation by gut microbiota. Sci. Total Environ. 2024, 912, 169418. [Google Scholar] [CrossRef] [PubMed]
  5. Yousefi, H.; Lak, E.; Mohammadi, M.J.; Shahriyari, H.A. Carcinogenic Risk Assessment among Children and Adult due to Exposure to Toxic Air Pollutants. Environ. Sci. Pollut. Res. Int. 2022, 29, 23015–23025. [Google Scholar] [CrossRef]
  6. Manisalidis, I.; Stavropoulou, E.; Stavropoulos, A.; Bezirtzoglou, E. Environmental and Health Impacts of Air Pollution: A Review. Front. Public Health 2020, 8, 14. [Google Scholar] [CrossRef] [PubMed]
  7. Zhou, M.; Wang, H.; Zeng, X.; Yin, P.; Zhu, J.; Chen, W.; Li, X.; Wang, L.; Wang, L.; Liu, Y.; et al. Mortality, morbidity, and risk factors in China and its provinces, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2019, 394, 1145–1158. [Google Scholar] [CrossRef]
  8. Zhang, L.; He, X.; Xiong, Y.; Ran, Q.; Xiong, A.; Wang, J.; Wu, D.; Niu, B.; Li, G. Transcriptome-wide profiling discover: PM2.5 aggravates airway dysfunction through epithelial barrier damage regulated by Stanniocalcin 2 in an OVA-induced model. Ecotoxicol. Environ. Saf. 2021, 220, 112408. [Google Scholar] [CrossRef] [PubMed]
  9. Yue, W.; Tong, L.; Liu, X.; Weng, X.; Chen, X.; Wang, D.; Dudley, S.C.; Weir, E.K.; Ding, W.; Lu, Z.; et al. Short term Pm2.5 exposure caused a robust lung inflammation, vascular remodeling, and exacerbated transition from left ventricular failure to right ventricular hypertrophy. Redox Biol. 2019, 22, 101161. [Google Scholar] [CrossRef] [PubMed]
  10. Ye, Z.; Wang, B.; Mu, G.; Zhou, Y.; Qiu, W.; Yang, S.; Wang, X.; Zhang, Z.; Chen, W. Short-term effects of real-time individual fine particulate matter exposure on lung function: A panel study in Zhuhai, China. Environ. Sci. Pollut. Res. Int. 2021, 28, 65140–65149. [Google Scholar] [CrossRef]
  11. Li, Y.; Lin, B.; Hao, D.; Du, Z.; Wang, Q.; Song, Z.; Li, X.; Li, K.; Wang, J.; Zhang, Q.; et al. Short-term PM2.5 exposure induces transient lung injury and repair. J. Hazard. Mater. 2023, 459, 132227. [Google Scholar] [CrossRef]
  12. Lin, Y.C.; Shih, H.S.; Lai, C.Y. Long-term nonlinear relationship between PM2.5 and ten leading causes of death. Environ. Geochem. Health 2021, 44, 3967–3990. [Google Scholar] [CrossRef] [PubMed]
  13. Moradi, M.; Mokhtari, A.; Mohammadi, M.J.; Hadei, M.; Vosoughi, M. Estimation of long-term and short-term health effects attributed to PM2.5 standard pollutants in the air of Ardabil (using Air Q + model). Environ. Sci. Pollut. Res. Int. 2022, 29, 21508–21516. [Google Scholar] [CrossRef]
  14. Lin, H.; Chen, M.; Gao, Y.; Wang, Z.; Jin, F. Tussilagone protects acute lung injury from PM2.5 via alleviating Hif-1alpha/NF-kappaB-mediated inflammatory response. Environ. Toxicol. 2022, 37, 1198–1210. [Google Scholar] [CrossRef]
  15. Ning, J.; Du, H.; Zhang, Y.; Liu, Q.; Jiang, T.; Pang, Y.; Tian, X.; Yan, L.; Niu, Y.; Zhang, R. N6-Methyladenosine Modification of CDH1 mRNA Promotes PM2.5-Induced Pulmonary Fibrosis via Mediating Epithelial Mesenchymal Transition. Toxicol. Sci. 2022, 185, 143–157. [Google Scholar] [CrossRef] [PubMed]
  16. Xia, C.; Dong, X.; Li, H.; Cao, M.; Sun, D.; He, S.; Yang, F.; Yan, X.; Zhang, S.; Li, N.; et al. Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin. Med. J. 2022, 135, 584–590. [Google Scholar] [CrossRef] [PubMed]
  17. Guo, H.; Chang, Z.; Wu, J.; Li, W. Air pollution and lung cancer incidence in China: Who are faced with a greater effect? Environ. Int. 2019, 132, 105077. [Google Scholar] [CrossRef]
  18. Ali, T.; Grote, P. Beyond the RNA-dependent function of LncRNA genes. Elife 2020, 9, e60583. [Google Scholar] [CrossRef] [PubMed]
  19. Herman, A.B.; Tsitsipatis, D.; Gorospe, M. Integrated lncRNA function upon genomic and epigenomic regulation. Mol. Cell 2022, 82, 2252–2266. [Google Scholar] [CrossRef]
  20. Teng, P.C.; Liang, Y.; Yarmishyn, A.A.; Hsiao, Y.J.; Lin, T.Y.; Lin, T.W.; Teng, Y.C.; Yang, Y.P.; Wang, M.L.; Chien, C.S.; et al. RNA Modifications and Epigenetics in Modulation of Lung Cancer and Pulmonary Diseases. Int. J. Mol. Sci. 2021, 22, 10592. [Google Scholar] [CrossRef]
  21. Abdi, E.; Latifi-Navid, S.; Panahi, A.; Latifi-Navid, H. LncRNA polymorphisms and lung cancer risk. Per. Med. 2023, 20, 511–522. [Google Scholar] [CrossRef] [PubMed]
  22. Gao, Y.; Zhang, N.; Zeng, Z.; Wu, Q.; Jiang, X.; Li, S.; Sun, W.; Zhang, J.; Li, Y.; Li, J.; et al. LncRNA PCAT1 activates SOX2 and suppresses radioimmune responses via regulating cGAS/STING signalling in non-small cell lung cancer. Clin. Transl. Med. 2022, 12, e792. [Google Scholar] [CrossRef]
  23. Zhou, Y.; Sun, L.; Zhu, M.; Cheng, H. Effects and early diagnostic value of lncRNA H19 on sepsis-induced acute lung injury. Exp. Ther. Med. 2022, 23, 279. [Google Scholar] [CrossRef]
  24. Lee, K.Y.; Ho, S.C.; Sun, W.L.; Feng, P.H.; Lin, C.W.; Chen, K.Y.; Chuang, H.C.; Tseng, C.H.; Chen, T.T.; Wu, S.M. Lnc-IL7R alleviates PM2.5-mediated cellular senescence and apoptosis through EZH2 recruitment in chronic obstructive pulmonary disease. Cell Biol. Toxicol. 2022, 38, 1097–1120. [Google Scholar] [CrossRef] [PubMed]
  25. Guo, H.; Feng, Y.; Yu, H.; Xie, Y.; Luo, F.; Wang, Y. A novel lncRNA, loc107985872, promotes lung adenocarcinoma progression via the notch1 signaling pathway with exposure to traffic-originated PM2.5 organic extract. Environ. Pollut. 2020, 266 Pt 1, 115307. [Google Scholar] [CrossRef] [PubMed]
  26. Langfelder, P.; Horvath, S. WGCNA: An R package for weighted correlation network analysis. BMC Bioinform. 2008, 9, 559. [Google Scholar] [CrossRef] [PubMed]
  27. Zhao, Y.; Ma, S.; Cui, Z.; Li, S.; Chen, Y.; Yin, Y.; Yin, Z. The relationship between LncRNAs and lung adenocarcinoma as well as their ceRNA network. Cancer Biomark. 2021, 31, 165–176. [Google Scholar] [CrossRef]
  28. Qi, G.; Kong, W.; Mou, X.; Wang, S. A new method for excavating feature lncRNA in lung adenocarcinoma based on pathway crosstalk analysis. J. Cell Biochem. 2019, 120, 9034–9046. [Google Scholar] [CrossRef] [PubMed]
  29. Zeng, Z.; Liang, Y.; Shi, J.; Xiao, L.; Tang, L.; Guo, Y.; Chen, F.; Lin, G. Identification and Application of a Novel Immune-Related lncRNA Signature on the Prognosis and Immunotherapy for Lung Adenocarcinoma. Diagnostics 2022, 12, 2891. [Google Scholar] [CrossRef]
  30. Kopp, F.; Mendell, J.T. Functional Classification and Experimental Dissection of Long Noncoding RNAs. Cell 2018, 172, 393–407. [Google Scholar] [CrossRef] [PubMed]
  31. Wang, M.; Aaron, C.P.; Madrigano, J.; Hoffman, E.A.; Angelini, E.; Yang, J.; Laine, A.; Vetterli, T.M.; Kinney, P.L.; Sampson, P.D.; et al. Association Between Long-term Exposure to Ambient Air Pollution and Change in Quantitatively Assessed Emphysema and Lung Function. JAMA 2019, 322, 546–556. [Google Scholar] [CrossRef]
  32. Shan, H.; Li, X.; Ouyang, C.; Ke, H.; Yu, X.; Tan, J.; Chen, J.; Wang, C.; Zhang, L.; Tang, Y.; et al. Salidroside prevents PM2.5-induced BEAS-2B cell apoptosis via SIRT1-dependent regulation of ROS and mitochondrial function. Ecotoxicol. Environ. Saf. 2022, 231, 113170. [Google Scholar] [CrossRef]
  33. Li, M.; Wei, X.; Li, Y.; Feng, T.; Jiang, L.; Zhu, H.; Yu, X.; Tang, J.; Chen, G.; Zhang, J.; et al. PM2.5 in poultry houses synergizes with Pseudomonas aeruginosa to aggravate lung inflammation in mice through the NF-kappaB pathway. J. Vet. Sci. 2020, 21, e46. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, W.; Langfelder, P.; Fuller, T.; Dong, J.; Li, A.; Hovarth, S. Weighted gene coexpression network analysis: State of the art. J. Biopharm. Stat. 2010, 20, 281–300. [Google Scholar] [CrossRef] [PubMed]
  35. Tian, Z.; He, W.; Tang, J.; Liao, X.; Yang, Q.; Wu, Y.; Wu, G. Identification of Important Modules and Biomarkers in Breast Cancer Based on WGCNA. Onco Targets Ther. 2020, 13, 6805–6817. [Google Scholar] [CrossRef]
  36. Nangraj, A.S.; Selvaraj, G.; Kaliamurthi, S.; Kaushik, A.C.; Cho, W.C.; Wei, D.Q. Integrated PPI- and WGCNA-Retrieval of Hub Gene Signatures Shared Between Barrett’s Esophagus and Esophageal Adenocarcinoma. Front. Pharmacol. 2020, 11, 881. [Google Scholar] [CrossRef] [PubMed]
  37. Lin, H.; Zhang, X.; Feng, N.; Wang, R.; Zhang, W.; Deng, X.; Wang, Y.; Yu, X.; Ye, X.; Li, L.; et al. LncRNA LCPAT1 Mediates Smoking/ Particulate Matter 2.5-Induced Cell Autophagy and Epithelial-Mesenchymal Transition in Lung Cancer Cells via RCC2. Cell Physiol. Biochem. 2018, 47, 1244–1258. [Google Scholar] [CrossRef] [PubMed]
  38. Deng, X.; Feng, N.; Zheng, M.; Ye, X.; Lin, H.; Yu, X.; Gan, Z.; Fang, Z.; Zhang, H.; Gao, M.; et al. PM2.5 exposure-induced autophagy is mediated by lncRNA loc146880 which also promotes the migration and invasion of lung cancer cells. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 112–125. [Google Scholar] [CrossRef] [PubMed]
  39. Spitz, A.Z.; Gavathiotis, E. Physiological and pharmacological modulation of BAX. Trends Pharmacol. Sci. 2022, 43, 206–220. [Google Scholar] [CrossRef]
  40. Jensen, K.; WuWong, D.J.; Wong, S.; Matsuyama, M.; Matsuyama, S. Pharmacological inhibition of Bax-induced cell death: Bax-inhibiting peptides and small compounds inhibiting Bax. Exp. Biol. Med. 2019, 244, 621–629. [Google Scholar] [CrossRef] [PubMed]
  41. Xiong, R.; Jiang, W.; Li, N.; Liu, B.; He, R.; Wang, B.; Geng, Q. PM2.5-induced lung injury is attenuated in macrophage-specific NLRP3 deficient mice. Ecotoxicol. Environ. Saf. 2021, 221, 112433. [Google Scholar] [CrossRef] [PubMed]
  42. Hao, X.; Wei, H. LncRNA H19 alleviates sepsis-induced acute lung injury by regulating the miR-107/TGFBR3 axis. BMC Pulm. Med. 2022, 22, 371. [Google Scholar] [CrossRef]
  43. Xu, Y.; Lin, L.; Lv, D.; Yan, S.; He, S.; Ge, H. LncRNA-LINC01089 inhibits lung adenocarcinoma cell proliferation and promotes apoptosis via sponging miR-543. Tissue Cell 2021, 72, 101535. [Google Scholar] [CrossRef]
  44. Ren, K.; Sun, J.; Liu, L.; Yang, Y.; Li, H.; Wang, Z.; Deng, J.; Hou, M.; Qiu, J.; Zhao, W. TP53-Activated lncRNA GHRLOS Regulates Cell Proliferation, Invasion, and Apoptosis of Non-Small Cell Lung Cancer by Modulating the miR-346/APC Axis. Front. Oncol. 2021, 11, 676202. [Google Scholar] [CrossRef]
  45. Khanna, K.; Chaudhuri, R.; Aich, J.; Pattnaik, B.; Panda, L.; Prakash, Y.S.; Mabalirajan, U.; Ghosh, B.; Agrawal, A. Secretory Inositol Polyphosphate 4-Phosphatase Protects against Airway Inflammation and Remodeling. Am. J. Respir. Cell Mol. Biol. 2019, 60, 399–412. [Google Scholar] [CrossRef] [PubMed]
  46. Arvind, M.; Pattnaik, B.; Gheware, A.; Prakash, Y.S.; Srivastava, M.; Agrawal, A.; Bhatraju, N.K. Plausible role of INPP4A dysregulation in idiopathic pulmonary fibrosis. Physiol. Rep. 2024, 12, e16032. [Google Scholar] [CrossRef] [PubMed]
  47. Lin, Z.; Li, Y.; Han, X.; Fu, Z.; Tian, Z.; Li, C. Targeting SPHK1/PBX1 Axis Induced Cell Cycle Arrest in Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2022, 23, 12741. [Google Scholar] [CrossRef]
  48. Wang, Y.; Hu, Y.; Xu, R.; Jin, X.; Jiao, W. Plin2 inhibits autophagy via activating AKT/mTOR pathway in non-small cell lung cancer. Exp. Cell Res. 2024, 435, 113955. [Google Scholar] [CrossRef] [PubMed]
  49. Winter, N.A.; Gibson, P.G.; McDonald, V.M.; Fricker, M. Sputum Gene Expression Reveals Dysregulation of Mast Cells and Basophils in Eosinophilic COPD. Int. J. Chronic Obstr. Pulm. Dis. 2021, 16, 2165–2179. [Google Scholar] [CrossRef]
  50. Chen, Y.; Jin, L.; Ma, Y.; Liu, Y.; Zhu, Q.; Huang, Y.; Feng, W. BACH1 promotes lung adenocarcinoma cell metastasis through transcriptional activation of ITGA2. Cancer Sci. 2023, 114, 3568–3582. [Google Scholar] [CrossRef]
  51. Kawami, M.; Ojima, T.; Yumoto, R.; Takano, M. Role of integrin alpha2 in methotrexate-induced epithelial-mesenchymal transition in alveolar epithelial A549 cells. Toxicol. Res. 2022, 38, 449–458. [Google Scholar] [CrossRef]
  52. Sun, Y.; Li, H.; Zhang, L.; Zhang, J. ATG16L1 regulated IL-22 induced IFN level in Pseudomonas aeruginosa Lung Infection via cGAS signal passage. Neuro Endocrinol. Lett. 2021, 42, 408–416. [Google Scholar]
  53. Nosaka, N.; Martinon, D.; Moreira, D.; Crother, T.R.; Arditi, M.; Shimada, K. Autophagy Protects Against Developing Increased Lung Permeability and Hypoxemia by Down Regulating Inflammasome Activity and IL-1beta in LPS Plus Mechanical Ventilation-Induced Acute Lung Injury. Front. Immunol. 2020, 11, 207. [Google Scholar] [CrossRef] [PubMed]
  54. Yin, D.; Zhang, Y.; Li, H.; Cheng, L. Association of TOP2A and ADH1B with lipid levels and prognosis in patients with lung adenocarcinoma and squamous cell carcinoma. Clin. Respir. J. 2023, 17, 1301–1315. [Google Scholar] [CrossRef] [PubMed]
  55. Yu, S.; Ao, Z.; Wu, Y.; Song, L.; Zhang, P.; Li, X.; Liu, M.; Qian, P.; Zhang, R.; Li, X.; et al. ZNF300 promotes chemoresistance and aggressive behaviour in non-small-cell lung cancer. Cell Prolif. 2020, 53, e12924. [Google Scholar] [CrossRef] [PubMed]
  56. Qiu, F.S.; Wang, J.F.; Guo, M.Y.; Li, X.J.; Shi, C.Y.; Wu, F.; Zhang, H.H.; Ying, H.Z.; Yu, C.H. Rgl-exomiR-7972, a novel plant exosomal microRNA derived from fresh Rehmanniae Radix, ameliorated lipopolysaccharide-induced acute lung injury and gut dysbiosis. Biomed. Pharmacother. 2023, 165, 115007. [Google Scholar] [CrossRef] [PubMed]
  57. Kuznetsova, E.S.; Zinovieva, O.L.; Oparina, N.Y.; Prokofjeva, M.M.; Spirin, P.V.; Favorskaya, I.A.; Zborovskaya, I.B.; Lisitsyn, N.A.; Prassolov, V.S.; Mashkova, T.D. [Abnormal expression of genes that regulate retinoid metabolism and signaling in non-small-cell lung cancer]. Mol. Biol. 2016, 50, 255–265. [Google Scholar] [CrossRef]
  58. Gangaraj, K.P.; Rajesh, M.K. Dataset of dual RNA-sequencing of Phytophthora palmivora infecting coconut (Cocos nucifera L.). Data Brief 2020, 30, 105455. [Google Scholar] [CrossRef] [PubMed]
  59. Rasmussen, A.H.; Kogelman, L.J.A.; Kristensen, D.M.; Chalmer, M.A.; Olesen, J.; Hansen, T.F. Functional gene networks reveal distinct mechanisms segregating in migraine families. Brain J. Neurol. 2020, 143, 2945–2956. [Google Scholar] [CrossRef]
  60. Xiao, K.W.; Li, J.L.; Zeng, Z.H.; Liu, Z.B.; Hou, Z.Q.; Yan, X.; Cai, L. Monocytes affect bone mineral density in pre- and postmenopausal women through ribonucleoprotein complex biogenesis by integrative bioinformatics analysis. Sci. Rep. 2019, 9, 17290. [Google Scholar] [CrossRef]
  61. Szklarczyk, D.; Gable, A.L.; Nastou, K.C.; Lyon, D.; Kirsch, R.; Pyysalo, S.; Doncheva, N.T.; Legeay, M.; Fang, T.; Bork, P.; et al. The STRING database in 2021: Customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021, 49, D605–D612. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Differential expression analysis of lncRNAs and mRNAs under different PM2.5 exposure conditions. (a) lncRNAs: PM2.5 (500 μg/mL) vs. control; (b) lncRNAs: PM2.5 (100 μg/mL) vs. control; (c) lncRNAs: PM2.5 (500 μg/mL) vs. PM2.5 (100 μg/mL); (d) mRNAs: PM2.5 (500 μg/mL) vs. control; (e) mRNAs: PM2.5 (100 μg/mL) vs. control; (f) mRNAs: PM2.5 (500 μg/mL) vs. PM2.5 (100 μg/mL).
Figure 1. Differential expression analysis of lncRNAs and mRNAs under different PM2.5 exposure conditions. (a) lncRNAs: PM2.5 (500 μg/mL) vs. control; (b) lncRNAs: PM2.5 (100 μg/mL) vs. control; (c) lncRNAs: PM2.5 (500 μg/mL) vs. PM2.5 (100 μg/mL); (d) mRNAs: PM2.5 (500 μg/mL) vs. control; (e) mRNAs: PM2.5 (100 μg/mL) vs. control; (f) mRNAs: PM2.5 (500 μg/mL) vs. PM2.5 (100 μg/mL).
Ijms 26 00911 g001
Figure 2. Identification of co-expression modules in different expression genes by WGCNA. (a) no outlier samples were found by cut-off height; (b) the soft threshold power of WGCNA; (c) the cluster dendrogram of WGCNA; (d) heatmap showing the adjacency matrix of eigengenes of various gene modules; (e) the correlation between modularity genes and lung injury.
Figure 2. Identification of co-expression modules in different expression genes by WGCNA. (a) no outlier samples were found by cut-off height; (b) the soft threshold power of WGCNA; (c) the cluster dendrogram of WGCNA; (d) heatmap showing the adjacency matrix of eigengenes of various gene modules; (e) the correlation between modularity genes and lung injury.
Ijms 26 00911 g002
Figure 3. Functional enrichment analysis in the turquoise and blue modules. (a,b) Enriched GO analysis of mRNAs in turquoise and blue modules. (c,d) Enriched KEGG pathway analysis of mRNAs in turquoise and blue modules.
Figure 3. Functional enrichment analysis in the turquoise and blue modules. (a,b) Enriched GO analysis of mRNAs in turquoise and blue modules. (c,d) Enriched KEGG pathway analysis of mRNAs in turquoise and blue modules.
Ijms 26 00911 g003
Figure 4. Module lncRNA–mRNA net of hub genes in the turquoise (a) and blue (b) modules. Module lncRNA–mRNA pathway net of hub genes in the turquoise (c) and blue (d) modules. Circles represent mRNAs, triangles represent lncRNAs, and gray polygons represent pathways. The size of the graph represents the level of intramodular connectivity of hub genes in the network.
Figure 4. Module lncRNA–mRNA net of hub genes in the turquoise (a) and blue (b) modules. Module lncRNA–mRNA pathway net of hub genes in the turquoise (c) and blue (d) modules. Circles represent mRNAs, triangles represent lncRNAs, and gray polygons represent pathways. The size of the graph represents the level of intramodular connectivity of hub genes in the network.
Ijms 26 00911 g004
Figure 5. Construction of protein–protein interaction (PPI) networks in the turquoise (a) and blue (b) modules.
Figure 5. Construction of protein–protein interaction (PPI) networks in the turquoise (a) and blue (b) modules.
Ijms 26 00911 g005
Figure 6. The main procedure of the study. WGCNA, weighted correlation network analysis.
Figure 6. The main procedure of the study. WGCNA, weighted correlation network analysis.
Ijms 26 00911 g006
Table 1. The numbers of lncRNAs and mRNAs in the 12 modules.
Table 1. The numbers of lncRNAs and mRNAs in the 12 modules.
ModuleAll NumberslncRNA NumbersmRNA Numbers
turquoise22217641457
blue1365519846
brown616239377
yellow404161243
green18642144
red1294881
black1123082
pink1014160
magenta751560
purple652144
green-yellow501832
grey202
Table 2. Top 20 significantly changed GOs of differentially expressed genes in turquoise and blue modules.
Table 2. Top 20 significantly changed GOs of differentially expressed genes in turquoise and blue modules.
ModuleGO idTermNo. of Genes−logP
turquoise moduleGO:0007165signal transduction9830.03
GO:0045944positive regulation of transcription by RNA polymerase II8922.21
GO:0006357regulation of transcription by RNA polymerase II10619.79
GO:0035556intracellular signal transduction4517.80
GO:0043065positive regulation of apoptotic process4217.33
GO:0007155cell adhesion5016.87
GO:0007275multicellular organism development4714.99
GO:0000122negative regulation of transcription by RNA polymerase II6414.64
GO:0045893positive regulation of transcription, DNA-templated5414.46
GO:0008284positive regulation of cell proliferation4412.73
GO:0070374positive regulation of ERK1 and ERK2 cascade2812.69
GO:0007596blood coagulation2511.76
GO:0006915apoptotic process4511.47
GO:0043547positive regulation of GTPase activity3210.94
GO:0030335positive regulation of cell migration2710.59
GO:0007156homophilic cell adhesion via plasma membrane adhesion molecules2310.44
GO:0045087innate immune response4310.11
GO:0010628positive regulation of gene expression349.78
GO:0007399nervous system development319.78
GO:0006954inflammatory response349.60
blue moduleGO:0007165signal transduction6019.18
GO:0006357regulation of transcription by RNA polymerase II7317.32
GO:0007268chemical synaptic transmission2513.37
GO:0007186G protein-coupled receptor signaling pathway5312.95
GO:0000122negative regulation of transcription by RNA polymerase II4311.74
GO:0045944positive regulation of transcription by RNA polymerase II4911.20
GO:0044267cellular protein metabolic process2010.66
GO:0045893positive regulation of transcription, DNA-templated349.90
GO:0016567protein ubiquitination299.41
GO:0006915apoptotic process308.92
GO:0043687post-translational protein modification238.63
GO:0043066negative regulation of apoptotic process267.75
GO:0000209protein polyubiquitination197.50
GO:0045892negative regulation of transcription, DNA-templated277.19
GO:0048013ephrin receptor signaling pathway117.01
GO:0007399nervous system development206.95
GO:0008284positive regulation of cell proliferation256.95
GO:0034220ion transmembrane transport166.87
GO:0006468protein phosphorylation246.68
GO:0007155cell adhesion246.41
Table 3. Top 20 significantly changed pathways of differentially expressed genes in turquoise and blue modules.
Table 3. Top 20 significantly changed pathways of differentially expressed genes in turquoise and blue modules.
ModulePathway IDTermNo. of Genes−logP
turquoise module01100Metabolic pathways10723.01
05200Pathways in cancer4713.39
04080Neuroactive ligand–receptor interaction3310.47
05016Huntington disease3110.35
05152Tuberculosis239.71
05171Coronavirus disease—COVID-19269.66
04060Cytokine-cytokine receptor interaction299.37
04630JAK-STAT signaling pathway218.99
05132Salmonella infection268.98
05022Pathways of neurodegeneration—multiple diseases378.98
05020Prion disease278.79
05167Kaposi sarcoma-associated herpesvirus infection228.33
04071Sphingolipid signaling pathway177.95
05014Amyotrophic lateral sclerosis307.86
05163Human cytomegalovirus infection237.78
04020Calcium signaling pathway237.25
05225Hepatocellular carcinoma197.15
05214Glioma137.12
04613Neutrophil extracellular trap formation206.99
04151PI3K-Akt signaling pathway286.96
blue module01100Metabolic pathways6213.08
04020Calcium signaling pathway198.32
04024cAMP signaling pathway188.24
04080Neuroactive ligand–receptor interaction227.94
04912GnRH signaling pathway127.63
05202Transcriptional misregulation in cancer167.33
00565Ether lipid metabolism97.10
04010MAPK signaling pathway196.88
04742Taste transduction105.93
04270Vascular smooth muscle contraction125.88
04724Glutamatergic synapse115.68
04713Circadian entrainment105.43
00564Glycerophospholipid metabolism105.39
04072Phospholipase D signaling pathway125.38
04921Oxytocin signaling pathway125.20
04928Parathyroid hormone synthesis, secretion, and action105.08
04911Insulin secretion94.94
04022cGMP-PKG signaling pathway124.83
05032Morphine addiction94.74
04014Ras signaling pathway144.74
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sui, J.; Zhang, Y.; Zhang, L.; Xia, H. Identification and Evaluation of Hub Long Non-Coding RNAs and mRNAs in PM2.5-Induced Lung Cell Injury. Int. J. Mol. Sci. 2025, 26, 911. https://doi.org/10.3390/ijms26030911

AMA Style

Sui J, Zhang Y, Zhang L, Xia H. Identification and Evaluation of Hub Long Non-Coding RNAs and mRNAs in PM2.5-Induced Lung Cell Injury. International Journal of Molecular Sciences. 2025; 26(3):911. https://doi.org/10.3390/ijms26030911

Chicago/Turabian Style

Sui, Jing, Yanni Zhang, Linjie Zhang, and Hui Xia. 2025. "Identification and Evaluation of Hub Long Non-Coding RNAs and mRNAs in PM2.5-Induced Lung Cell Injury" International Journal of Molecular Sciences 26, no. 3: 911. https://doi.org/10.3390/ijms26030911

APA Style

Sui, J., Zhang, Y., Zhang, L., & Xia, H. (2025). Identification and Evaluation of Hub Long Non-Coding RNAs and mRNAs in PM2.5-Induced Lung Cell Injury. International Journal of Molecular Sciences, 26(3), 911. https://doi.org/10.3390/ijms26030911

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop