[1] Qi XM, Luo Y, Song MY, et al. Pneumoconiosis: current status and future prospects. Chin Med J, 2021; 134, 898−907. doi:  10.1097/CM9.0000000000001461
[2] Wang DM, Liang RY, Yang M, et al. Incidence and disease burden of coal workers' pneumoconiosis worldwide, 1990-2019: evidence from the global burden of disease study 2019. Eur Respir J, 2021; 58, 2101669. doi:  10.1183/13993003.01669-2021
[3] Chong SM, Lee KS, Chung MJ, et al. Pneumoconiosis: comparison of imaging and pathologic findings. RadioGraphics, 2006; 26, 59−77. doi:  10.1148/rg.261055070
[4] Adamcakova J, Mokra D. New insights into pathomechanisms and treatment possibilities for lung silicosis. Int J Mol Sci, 2021; 22, 4162. doi:  10.3390/ijms22084162
[5] Gong GC, Song SR, Su J. Pulmonary fibrosis alters gut microbiota and associated metabolites in mice: an integrated 16s and metabolomics analysis. Life Sci, 2021; 264, 118616. doi:  10.1016/j.lfs.2020.118616
[6] Wu YL, Li YH, Luo YB, et al. Gut microbiome and metabolites: the potential key roles in pulmonary fibrosis. Front Microbiol, 2022; 13, 943791. doi:  10.3389/fmicb.2022.943791
[7] Pang JL, Qi XM, Luo Y, et al. Multi-omics study of silicosis reveals the potential therapeutic targets PGD2 and TXA2. Theranostics, 2021; 11, 2381−94. doi:  10.7150/thno.47627
[8] Wang MY, Zhang Z, Liu JF, et al. Gefitinib and fostamatinib target EGFR and SYK to attenuate silicosis: a multi-omics study with drug exploration. Sig Transduct Target Ther, 2022; 7, 157. doi:  10.1038/s41392-022-00959-3
[9] Zhang N, Liu KL, Wang K, et al. Dust induces lung fibrosis through dysregulated DNA methylation. Environ Toxicol, 2019; 34, 728−41. doi:  10.1002/tox.22739
[10] He JY, Zhang CW, Zhang YK, et al. Polyunsaturated fatty acid concentrations and risk of pneumoconiosis: a two-sample mendelian randomization study. Biomed Environ Sci, 2024; 37, 1328−33.
[11] Sekula P, Del Greco MF, Pattaro C, et al. Mendelian randomization as an approach to assess causality using observational data. J Am Soc Nephrol, 2016; 27, 3253−65. doi:  10.1681/ASN.2016010098
[12] Zheng J, Baird D, Borges MC, et al. Recent developments in mendelian randomization studies. Curr Epidemiol Rep, 2017; 4, 330−45. doi:  10.1007/s40471-017-0128-6
[13] Lopera-Maya EA, Kurilshikov A, van der Graaf A, et al. Effect of host genetics on the gut microbiome in 7, 738 participants of the dutch microbiome project. Nat Genet, 2022; 54, 143−51. doi:  10.1038/s41588-021-00992-y
[14] Ferkingstad E, Sulem P, Atlason BA, et al. Large-scale integration of the plasma proteome with genetics and disease. Nat Genet, 2021; 53, 1712−21. doi:  10.1038/s41588-021-00978-w
[15] Sun BB, Chiou J, Traylor M, et al. Plasma proteomic associations with genetics and health in the UK biobank. Nature, 2023; 622, 329−38. doi:  10.1038/s41586-023-06592-6
[16] Võsa U, Claringbould A, Westra HJ, et al. Large-scale cis- and trans-eqtl analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat Genet, 2021; 53, 1300−10. doi:  10.1038/s41588-021-00913-z
[17] McRae AF, Marioni RE, Shah S, et al. Identification of 55, 000 replicated DNA methylation QTL. Sci Rep, 2018; 8, 17605. doi:  10.1038/s41598-018-35871-w
[18] Kurki MI, Karjalainen J, Palta P, et al. Finngen provides genetic insights from a well-phenotyped isolated population. Nature, 2023; 613, 508−18. doi:  10.1038/s41586-022-05473-8
[19] Chen J, Ruan XX, Sun YH, et al. Multi-omic insight into the molecular networks of mitochondrial dysfunction in the pathogenesis of inflammatory bowel disease. eBioMedicine, 2024; 99, 104934. doi:  10.1016/j.ebiom.2023.104934
[20] Watanabe K, Taskesen E, van Bochoven A, et al. Functional mapping and annotation of genetic associations with FUMA. Nat Commun, 2017; 8, 1826. doi:  10.1038/s41467-017-01261-5
[21] Legault MA, Perreault LPL, Tardif JC, et al. ExPheWas: a platform for cis-Mendelian randomization and gene-based association scans. Nucleic Acids Res, 2022; 50, W305−11. doi:  10.1093/nar/gkac289
[22] Eberhardt J, Santos-Martins D, Tillack AF, et al. AutoDock Vina 1.2. 0: new docking methods, expanded force field, and python bindings. J Chem Inf Model, 2021; 61, 3891−8. doi:  10.1021/acs.jcim.1c00203
[23] Alikhan MM, Lee FEH. Understanding nontypeable haemophilus influenzae and chronic obstructive pulmonary disease. Curr Opin Pulm Med, 2014; 20, 159−64. doi:  10.1097/MCP.0000000000000023
[24] Mahenthiralingam E. Emerging cystic fibrosis pathogens and the microbiome. Paediatr Respir Rev, 2014; 15, 13−5.
[25] Chen SS, Zhang XY, Yang C, et al. Essential role of IL-17 in acute exacerbation of pulmonary fibrosis induced by non-typeable Haemophilus influenzae. Theranostics, 2022; 12, 5125−37. doi:  10.7150/thno.74809
[26] Cai Y, van Putten JPM, Gilbert MS, et al. Galacto-oligosaccharides as an anti-bacterial and anti-invasive agent in lung infections. Biomaterials, 2022; 283, 121461. doi:  10.1016/j.biomaterials.2022.121461
[27] Middleton AM, Dowling RB, Mitchell JL, et al. Haemophilus parainfluenzae infection of respiratory mucosa. Respir Med, 2003; 97, 375−81. doi:  10.1053/rmed.2002.1454
[28] Li QM, Cui Y, Xu BC, et al. Main active components of Jiawei Gegen Qinlian decoction protects against ulcerative colitis under different dietary environments in a gut microbiota-dependent manner. Pharmacol Res, 2021; 170, 105694. doi:  10.1016/j.phrs.2021.105694
[29] Park HJ, Jeong OY, Chun SH, et al. Butyrate improves skin/lung fibrosis and intestinal dysbiosis in bleomycin-induced mouse models. Int J Mol Sci, 2021; 22, 2765. doi:  10.3390/ijms22052765
[30] Li Q, Deng MS, Wang RT, et al. PD-L1 upregulation promotes drug-induced pulmonary fibrosis by inhibiting vimentin degradation. Pharmacol Res, 2023; 187, 106636. doi:  10.1016/j.phrs.2022.106636
[31] Turini S, Bergandi L, Gazzano E, et al. Epithelial to mesenchymal transition in human mesothelial cells exposed to asbestos fibers: role of TGF-β as mediator of malignant mesothelioma development or metastasis via EMT event. Int J Mol Sci, 2019; 20, 150. doi:  10.3390/ijms20010150
[32] Bogdanovic A, Bennett N, Kieffer S, et al. Syntaxin 7, syntaxin 8, vti1 and VAMP7 (vesicle-associated membrane protein 7) form an active SNARE complex for early macropinocytic compartment fusion in Dictyostelium discoideum. Biochem J, 2002; 368, 29−39. doi:  10.1042/bj20020845
[33] Bilan F, Nacfer M, Fresquet F, et al. Endosomal SNARE proteins regulate CFTR activity and trafficking in epithelial cells. Exp Cell Res, 2008; 314, 2199−211. doi:  10.1016/j.yexcr.2008.04.012
[34] Collawn JF, Matalon S. CFTR and lung homeostasis. Am J Physiol Lung Cell Mol Physiol, 2014; 307, L917−23. doi:  10.1152/ajplung.00326.2014
[35] Luo YF, Yi H, Huang XY, et al. Inhibition of macrophage migration inhibitory factor (MIF) as a therapeutic target in bleomycin-induced pulmonary fibrosis rats. Am J Physiol Lung Cell Mol Physiol, 2021; 321, L6−16. doi:  10.1152/ajplung.00288.2020
[36] Calandra T, Roger T. Macrophage migration inhibitory factor: a regulator of innate immunity. Nat Rev Immunol, 2003; 3, 791−800. doi:  10.1038/nri1200
[37] Calandra T, Bernhagen J, Metz CN, et al. MIF as a glucocorticoid-induced modulator of cytokine production. Nature, 1995; 377, 68−71. doi:  10.1038/377068a0
[38] Donnelly SC, Haslett C, Reid PT, et al. Regulatory role for macrophage migration inhibitory factor in acute respiratory distress syndrome. Nat Med, 1997; 3, 320−3. doi:  10.1038/nm0397-320
[39] Onodera S, Nishihira J, Iwabuchi K, et al. Macrophage migration inhibitory factor up-regulates matrix metalloproteinase-9 and -13 in rat osteoblasts. Relevance to intracellular signaling pathways. J Biol Chem, 2002; 277, 7865−74. doi:  10.1074/jbc.M106020200