93
Views
9
CrossRef citations to date
0
Altmetric
Original Research

Interleukin -1β Promotes Lung Adenocarcinoma Growth and Invasion Through Promoting Glycolysis via p38 Pathway

, , , , ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 6491-6509 | Published online: 02 Dec 2021
 

Abstract

Background

There is a close relationship among inflammation, glycolysis, and tumors. The IL-1 family includes important inflammatory cytokines, among which IL-1β has been widely studied. In this study, we focused on the effect of IL-1β on glycolysis of lung adenocarcinoma (LUAD) cells in vivo and in vitro and explored its possible mechanisms.

Methods

A bioinformatic database and quantitative real-time PCR were used to analyze the expression of glycolysis-related enzyme genes and their correlations with IL1β in human LUAD samples. The human LUAD cell line A549 and Lewis lung carcinoma LLC cell line were stimulated with IL-1β. In vitro treatment effects, including glycolysis level, migration, and invasion were evaluated with a glucose assay kit, lactate assay kit, Western blotting, wound healing, and the transwell method. We established a mouse model of subcutaneous tumors using LLC cells pretreated with IL-1β and analyzed in vivo treatment effects through positron-emission tomography-computed tomography and staining. Virtual screening and molecular dynamic simulation were used to screen potential inhibitors of IL-1β.

Results

Our results showed that IL1β was positively correlated with the expression of glycolysis-related enzyme genes in LUAD. Glycolysis, migration, and invasion significantly increased in A549 and LLC stimulated with IL-1β. In vivo, IL-1β increased growth, mean standard uptake value, and pulmonary tumor metastasis, which were inhibited by the glycolysis inhibitor 2-deoxy-D-glucose and p38-pathway inhibitors. Small molecular compound ZINC14610053 was suggested being a potential inhibitor of IL-1β.

Conclusion

IL-1β promotes glycolysis of LUAD cells through p38 signaling, further enhancing tumor-cell migration and invasion. These results show that IL-1β links inflammation to glycolysis in LUAD, and targeting IL-1β and the glycolysis pathway may be a potential therapeutic strategy for lung cancer.

Abbreviations

qRT-PCR, quantitative real-time polymerase chain reaction; LLC, Lewis lung carcinoma; PET-CT, positron-emission tomography–computed tomography; H&E, hematoxylin–eosin; 2DG, 2-deoxy-D-glucose; SUV, standard uptake value; NSCLC, non–small cell lung cancer; GEPIA, gene-expression profiling interactive analysis; TCGA, The Cancer Genome Atlas; GTEx, Genotype-tissue Expression project; EGA, European Genome-Phenome Archive; GEO, Gene Expression Omnibus; HK, hexokinase; PFK, phosphofructokinase; PFKL, phosphofructokinase liver; PK, pyruvate kinase; PKM2, pyruvate kinase M2; LDH, lactate dehydrogenase; LDHA, lactate dehydrogenase A; MCT, monocarboxylate transporter; TIMER, Tumor Immune Estimation Resource; LUAD, lung adenocarcinoma; DFS, disease-free survival; OS, overall survival; RFS, relapse-free survival; RMSD, root-mean-square deviation; RMSF, root-mean-square fluctuation; MDSC, myeloid-derived suppressor cell; TAM, tumor-associated macrophage; TAN, tumor-associated neutrophil.

Ethics Statement

This study was conducted in accordance with the Declaration of Helsinki. The animal study was approved by the Animal Care and Use Committee of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China [(2020) IACUC (S2470)]. All experimental protocols are carried out in accordance with institutional guidelines and national guidelines and regulations of Tongji Medical College, Huazhong University of Science and Technology.

Author Contributions

  1. All authors have made substantial contributions to the concept of the paper and acquisition, analysis, and interpretation of data.

  2. All authors have taken part in drafting and writing the paper and carefully revised and reviewed the article.

  3. All authors have agreed on submission of the paper to the journal.

  4. All authors have reviewed and agreed on the version of the article to be published.

All authors agree to be accountable for all aspects of the work.

Disclosure

The authors declare no conflicts of interest for this study.

Additional information

Funding

This study was supported by grants from the National Major Scientific and Technological Special Project for Significant New Drug Development (2019ZX09301-001), National Science Foundation of China (81770096, 81802113, 81900095), and Key Laboratory Open Fund of Hubei Province (F016.02004.2,0003.082).