- Research Note
- Open access
- Published:
Comprehensive single-cell RNA-sequencing study of Tollip deficiency effect in IL-13-stimulated human airway epithelial cells
BMC Research Notes volume 18, Article number: 194 (2025)
Abstract
Objective
Toll-interacting protein (Tollip) suppresses excessive pro-inflammatory signaling, but its function in airway epithelial responses to IL-13, a key mediator in allergic diseases, remains unclear. This study investigates Tollip knockdown (TKD) effects in primary human airway epithelial cells using single-cell RNA sequencing, providing the first single-cell analysis of TKD and the first exploring its interaction with IL-13.
Results
IL-13 treatment upregulated key genes, including SPDEF, MUC5AC, POSTN, ALOX15, and CCL26, confirming IL-13’s effects and validating our methods. IL-13 reduced TNF-α signaling and epithelial-mesenchymal transition in certain cell types, suggesting a dual role in promoting type 2 inflammation while suppressing Th1-driven inflammation. Tollip deficiency alone significantly amplified TNF-α signaling and inflammatory pathways in goblet, club, and suprabasal cells. Comparisons between TKDIL13 vs IL13 and TKD vs CTR revealed that IL-13 does not substantially alter Tollip deficiency response in most cell types, reinforcing findings in TKD vs CTR. Tollip deficiency alters the response to IL-13 in a cell-type-specific manner, strongly downregulating TNF-α signaling in goblet cells but only weakly in basal and club cells. Tollip deficiency enhances IL-13’s suppression of Th1 inflammatory responses in goblet cells. These novel insights in Tollip-IL-13 interactions offer potential therapeutic targets for asthma and related diseases.
Introduction
Toll-interacting protein (Tollip) is a ubiquitously expressed adaptor protein crucial to innate immunity [1, 2]. Tollip is a negative regulator of toll-like receptor signaling to prevent excessive inflammation [1, 3,4,5]. Reduced Tollip expression is linked to various diseases, including asthma, tuberculosis, idiopathic pulmonary fibrosis, atopic dermatitis, and Alzheimer’s [6,7,8,9,10]. Our prior work demonstrated that the Tollip AG/GG SNP rs5743899 is associated with poor airflow in asthma patients [11]. However, cell type-specific effects of Tollip are yet unclear.
Interleukin 13 (IL-13) is a type 2 cytokine [12], regulating eosinophilic inflammation and enhancing airway hyperresponsiveness [13,14,15]. IL-13 is central to chronic respiratory diseases including asthma and chronic obstructive pulmonary disease (COPD) [16, 17].
This report aimed to determine the individual and combined effects of Tollip knockdown and IL-13 on human airway epithelial cells at the single-cell level. This is the first comprehensive single-cell RNA sequencing (scRNA-seq) study of Tollip-deficient airway epithelial cells and cell type-specific response, and the first exploring the co-influence of Tollip deficiency and IL-13.
Main text
Methods
Primary human tracheobronchial epithelial cells were obtained from a healthy adult donor (National Jewish Health IRB approval #HS-3209). Tollip knockdown was achieved in submerged culture using CRISPR/Cas9 [6]. Verified Tollip knockdown cells were seeded onto collagen-coated Transwell inserts (Corning) in PneumaCult-ALI Medium (STEMCELL Technologies) at air–liquid interface for 21 days to induce mucociliary differentiation. Cells were treated with the following conditions for 3 days:
-
1.
CTR (Control, Tollip-sufficient)
-
2.
IL13 (10 ng/mL, Tollip-sufficient + IL-13)
-
3.
TKD (Tollip-knockdown, no IL-13)
-
4.
TKDIL13 (Tollip-knockdown + IL-13)
Cells were dissociated, loaded into a Chromium Controller (10× Genomics), and processed with Next GEM 3ʹ v3.1 reagents following manufacturer’s protocols. Libraries were sequenced on an Illumina platform (~ 350 million read pairs per sample).
Reads were aligned to GRCh38-2024-A using Cell Ranger (v8.0.0). Quality filters included > 500 genes/cell, < 20% mitochondrial reads, and log10 (UMI counts) within median ± 5 × MAD [18]. CellBender removed ambient RNA [19], and Scrublet filtered doublets [20]. We retained 3114 (CTR), 2148 (IL13), 2447 (TKD), and 1898 (TKDIL13) high-quality cells.
The dataset was aggregated using Cell Ranger’s aggr function to normalize sequencing depth across samples, then processed with the reanalyze function, focusing on barcodes that passed filtering.
Cell types were assigned using Azimuth label transfer against a Human Lung Cell Atlas reference [21,22,23], comprising 584,944 cells from 107 individuals [24], ensuring robust epithelial annotation. Cell-type markers were displayed in Additional file 1 and our previous publication [25].
Harmony was used to integrate single-cell data [26], aligning similar cell types across conditions. Samples were normalized using sctransform v2 [27], merged, and analyzed with 20 principal components. UMAP (reduction = “harmony,” dims = 1:20) and Louvain clustering (resolution 0.5) were performed in Seurat 5 [23] for visualization.
Differential expression for each cell type between conditions was calculated using Loupe Browser 8.0.0, which applies the sSeq test or edgeR for large UMI counts [28]. Pathway analysis used gene set enrichment analysis (GSEA) with FGSEA [29, 30] and hallmark gene sets from MSigDB [31, 32].
Results and discussion
UMAP-based clustering (Fig. 1) identified key airway epithelial cell types: basal, suprabasal, club, goblet, multiciliated, and SMG duct cells. Tollip knockdown (TKD, TKDIL13) consistently reduced multiciliated cells and increased goblet cells compared to CTR and IL13. This finding suggests that Tollip plays a critical role in multiciliated cell development or maintenance, with their loss potentially impairing mucociliary clearance and increasing infection risk [33, 34]. Coupled with heightened inflammatory signaling, this underscores Tollip’s multifaceted role in maintaining airway homeostasis. Cell counts are in Additional file 2, while genes contributing to the significant enrichment of each pathway between conditions are in Additional file 3.
IL-13 vs CTR: validation of IL-13-mediated responses
Comparing IL13 vs CTR revealed how IL-13 reshapes the Tollip-sufficient epithelium. SPDEF and MUC5AC were significantly upregulated in goblet and club cells (Additional file 4). Activated by IL-13 via STAT6, SPDEF induces mucus production and interacts with TTF-1 to exacerbate goblet cell hyperplasia, contributing to airflow obstruction [35]. Targeting the IL-13–SPDEF pathway with IL-13/IL-4Rα blockers can effectively reduce goblet cell hyperplasia and mucus hypersecretion [36]. Elevated MUC5AC, a key mucin, impairs mucociliary clearance and fosters airway obstruction, hallmarks of chronic respiratory diseases like asthma and COPD [37]. Therapeutic monoclonal antibodies against IL-4/IL-13 have been shown to decrease mucus production, highlighting MUC5AC as a principal mucus-obstructive component of asthma and chronic bronchitis [38].
IL-13 elevates POSTN in basal, club, and suprabasal cells (Additional file 5), supporting its involvement in extracellular matrix deposition and airway remodeling such as fibrosis and stiffening in asthma [39, 40]. Across all cell types, IL-13, via JAK–STAT pathways, upregulates ALOX15, which through converting arachidonic acid into 15-HETE, promotes eosinophilic inflammation and exacerbates airway hyperresponsiveness in asthma [41]. ALOX15 represents another potential target for therapies to reduce airway eosinophilic inflammation. We observed that basal cells also increased POSTN and ALOX15 under IL-13 stimulation, implicating the role of differentiating cells in tissue remodeling. While secretory cells respond to IL-13 by producing mucus and chemokines, basal cells respond by secreting factors that prime the airway for structural changes, suggesting IL-13 drives mucus hypersecretion and airway remodeling through different cell populations.
Similarly, IL13-induced CCL26 drives eosinophil recruitment [42], which correlates with more frequent asthma exacerbations. Given the effectiveness of IL-4/13 blockers [38], targeting CCL26 and its receptor CCR3 offers a promising therapeutic strategy [42].
Together, induction of these genes not only confirms IL-13’s known regulatory effects and validates our methods and results but also provides cell-type-specific insights.
TNF-α signaling
GSEA (Fig. 2a) revealed that IL-13 suppressed TNF-α–NF-κB signaling in all cell types, most notably club cells. Key downregulated genes include TLR2, TNFAIP3, CCL20, and IL-18, which supports previous studies indicating IL-13’s inhibition of Th1 cytokine and TNF-α transcription [43,44,45]. Mechanistically, via the phosphoinositide 3-kinase pathway, IL-13 increases IRAK-M, subsequently suppressing TLR2 signaling, NF-κB activation, and innate immunity [46]. Overall, this suggests a dual role of IL-13 in promoting type 2 inflammation (SPDEF, MUC5AC, etc.), while suppressing type 1 inflammation, a shift that promotes Th2-dominant conditions, characteristics of asthma.
A Hallmark (H) pathways significantly downregulated by IL-13 treatment (IL13) compared to control (CTR). The size of the dot represents the Normalized Enrichment Score (NES), indicating the strength of pathway enrichment in a given cell type, with larger dots reflecting stronger enrichment. The color of the dot represents p-adjusted value, indicating the statistical significance of the pathway. B Pathways significantly downregulated in Tollip knockout + IL-13-treated (TKDIL13) compared to Tollip knockout-only (TKD)
Inflammatory response
IL-13 also reduced the overall inflammatory response in suprabasal, basal, club, and multiciliated cells. While many previous studies on IL-13 have focused on its activation of Th2 allergic inflammation [47,48,49]. our research supports IL-13’s emerging role in suppressing Th1 inflammation [45]. Among our significant inflammatory response genes, most are more closely associated with a Th1 immune response than a Th2 response, including CCL20, CXCL8, IL-18, IL-6, and TLR2, all downregulated.
Epithelial-mesenchymal transition (EMT)
IL-13 suppressed the EMT pathway in club and multiciliated cells. This may be attributed to IL-13’s known role in inducing goblet cell metaplasia from club and multiciliated cells [50, 51]. EMT is typically associated with the transition of epithelial cells to more migratory phenotypes [52], contrary to the stationary phenotype of goblet cells. Notable downregulated genes related to the EMT pathway include IL-6, CXCL6, CXCL8, IGFBP3, LAMC1, and LAMC2.
The suppression of EMT suggests a potential protective effect against airway remodeling processes typically associated with fibrosis. This finding, contrasted with the increased expression of mucins and Th2 inflammation-related genes, adds to the growing evidence that IL-13 has both anti-inflammatory and pro-inflammatory effects.
Effect of Tollip deficiency in the absence of IL-13
TNF-α signaling
In TKD cells, the TNF-α signaling pathway is upregulated across all cell types except multiciliated cells (Fig. 3a). Goblet and club cells exhibited the greatest change in TNF-α-signaling-related expression, with significant upregulation of TNF, TNF-αIP2, NFKB1, TRAF1, and RIPK2. This upregulation corroborates Tollip’s known role in inhibiting TNF-α signaling, as its knockdown reduces this suppression [2, 53].
Inflammatory response
The inflammatory response pathway in TKD cells was upregulated across all cell types except multiciliated cells. Once again, goblet and club cells exhibited the greatest change in inflammatory response gene expression. Our previous work found that Tollip inhibits neutrophilic inflammation in response to rhinovirus infection [6]. IL1R1, IRAK2, NFKB1, TNFRSF1B, TLR3, and TLR1, all associated with neutrophilic inflammation, were significantly upregulated in TKD cells, corroborating our previous findings. Multiciliated cells, however, showed no significant change.
IL6-JAK-STAT3 signaling
In TKD cells, the IL6-JAK-STAT3 signaling pathway is upregulated across all cell types, excluding multiciliated cells. Goblet, club, suprabasal, basal, and SMG duct cells showed significant upregulation of related genes, including IL1B, HMOX1, IL6ST, STAT1, and IRF1. This corroborates our earlier research that Tollip deficiency promotes STAT3 activation [54] and that lower Tollip expression is associated with higher IL-6 protein levels [11].
Effect of Tollip deficiency in the presence of IL-13
The TKDIL13 vs TKD results compared to those of IL13 vs CTR show how Tollip deficiency modifies response to IL-13 treatment (Fig. 2b).
TNF-α signaling
Similar to the IL13 vs CTR comparison results, TNF-α signaling via NF-κB is downregulated in TKDIL13 vs TKD across goblet, suprabasal, and multiciliated cells, highlighting IL-13’s role in suppressing pro-inflammatory signaling, even in the absence of Tollip. However, some differences were observed. Goblet cells exhibited a notably stronger downregulation. SMG duct cells, omitted from the IL13 vs CTR comparison due to low cell count, also displayed downregulation of TNF-α signaling in TKDIL13 vs TKD. In contrast, basal and club cells no longer showed any significant enrichment of TNF-α signaling, suggesting that Tollip’s interaction with IL-13 is cell-type-specific.
Inflammatory response
The expression of the inflammatory response pathway differs significantly between IL13 vs CTR and TKDIL13 vs TKD, suggesting that the addition of TKD modifies the response to IL-13 across various cell types. Suprabasal, basal, club, and multiciliated cells all no longer showed significant expression changes in the inflammatory response pathway in the latter comparison. Goblet cells, meanwhile, had many more downregulated genes related to Th1 inflammatory response in TKDIL13 vs TKD compared to IL13 vs CTR, including CSF3, TNFSF15, IL1B, TLR2, CXCL10, and IRF1.
Epithelial–mesenchymal transition
Changes in EMT in multiciliated cells were largely similar between the two comparisons. Notable downregulated genes included SNAI1, TWIST1, SERPINE1, CDH2, and CCL20.
Effect of IL-13 treatment on Tollip-deficient cells
TKDIL13 vs IL13 and TKD vs CTR results were surprisingly similar, suggesting IL-13 does not induce significant changes in response to Tollip deficiency. This similarity validates findings from TKD vs. CTR and elucidates IL-13’s role in modulating the response to Tollip deficiency. Pathway differences were analyzed between TKDIL13 vs. IL13 (Fig. 3b) and TKD vs CTR. In TKDIL13 vs. IL13, suprabasal cells showed the same upregulation in all pathways discussed in TKD vs. CTR. Basal cells also exhibited similar results. However, some differences were observed. Due to low club cell count in TKDIL13, their strong pathway upregulation in TKD vs. CTR was absent in TKDIL13 vs. IL13. Additionally, multiciliated cells experienced upregulation of inflammatory response and TNF-α signaling in TKDIL13 vs IL13, though not in TKD vs CTR. Meanwhile, goblet cells showed slightly weaker upregulation for both pathways in TKDIL13 vs IL13.
Integrated view
Our study provides a detailed analysis of the effects of Tollip deficiency and IL-13 treatment on human airway epithelial cells at the single-cell level. The findings highlight Tollip’s critical role in regulating pro-inflammatory pathways and maintaining multiciliated cell populations while also revealing IL-13’s dual function in activating Th2 inflammation and concurrently suppressing neutrophilic inflammation and EMT-associated remodeling. The complex interactions between Tollip deficiency and IL-13 suggest that therapeutic strategies targeting these pathways will need to be tailored to specific cell types and disease contexts to achieve optimal outcomes.
Future research should focus on elucidating the molecular mechanisms by which Tollip deficiency leads to the upregulation of TNF-α signaling and other key pro-inflammatory pathways, as well as exploring the potential therapeutic benefits of targeting these pathways in asthmatic individuals with reduced Tollip function. Additionally, further studies are needed to investigate broader physiological implications of the observed cell-type-specific responses to Tollip deficiency and IL-13 treatment, particularly in the context of airway remodeling and chronic inflammation.
Limitations
While comparisons between TKDIL13 vs IL13 and TKD vs CTR validate the effect of Tollip knockdown, the relatively low cell count (2000–3000 cells/condition), single-patient data, and in-vitro design limit statistical power and generalizability. Future mechanistic studies with larger sample sizes and in-vivo models could further confirm and expand these findings.
Availability of data and materials
The dataset supporting the conclusions of this article is available in the Gene Expression Omnibus database under accession number GSE279044.
Abbreviations
- CTR:
-
Control sample
- TKD:
-
Tollip knockout-only sample
- IL13:
-
IL-13 treatment-only sample
- TKDIL13:
-
Tollip knockout and IL-13 treated sample
References
Capelluto DG. Tollip: a multitasking protein in innate immunity and protein trafficking. Microbes Infect. 2012;14(2):140–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.micinf.2011.08.018.
Humbert-Claude M, Duc D, Dwir D, et al. Tollip, an early regulator of the acute inflammatory response in the substantia Nigra. J Neuroinflamm. 2016;13(1):303. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-016-0766-5. (Published 2016 Dec 7).
Zhang G, Ghosh S. Negative regulation of toll-like receptor-mediated signaling by Tollip. J Biol Chem. 2002;277(9):7059–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M109537200.
Shah JA, Vary JC, Chau TT, et al. Human TOLLIP regulates TLR2 and TLR4 signaling and its polymorphisms are associated with susceptibility to tuberculosis. J Immunol. 2012;189(4):1737–46. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1103541.
Didierlaurent A, Brissoni B, Velin D, et al. Tollip regulates proinflammatory responses to interleukin-1 and lipopolysaccharide. Mol Cell Biol. 2006;26(3):735–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/MCB.26.3.735-742.2006.
Dakhama A, Al Mubarak R, Pavelka N, et al. Tollip inhibits ST2 Signaling In Airway Epithelial Cells Exposed To Type 2 cytokines and rhinovirus. J Innate Immun. 2020;12(1):103–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000497072.
Li X, Goobie GC, Gregory AD, Kass DJ, Zhang Y. Toll-interacting protein in pulmonary diseases. Abiding by the goldilocks principle. Am J Respir Cell Mol Biol. 2021;64(5):536–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1165/rcmb.2020-0470TR.
Li X, Kim SE, Chen TY, et al. Toll interacting protein protects bronchial epithelial cells from bleomycin-induced apoptosis. FASEB J. 2020;34(8):9884–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.201902636RR.
Schimming TT, Parwez Q, Petrasch-Parwez E, Nothnagel M, Epplen JT, Hoffjan S. Association of toll-interacting protein gene polymorphisms with atopic dermatitis. BMC Dermatol. 2007;7:3. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-5945-7-3. (Published 2007 Mar 16).
Chen K, Yuan R, Geng S, et al. Toll-interacting protein deficiency promotes neurodegeneration via impeding autophagy completion in high-fat diet-fed ApoE-/-mouse model. Brain Behav Immun. 2017;59:200–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbi.2016.10.002.
Huang C, Jiang D, Francisco D, et al. Tollip SNP rs5743899 modulates human airway epithelial responses to rhinovirus infection. Clin Exp Allergy. 2016;46(12):1549–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cea.12793.
Zhu J. T helper 2 (Th2) cell differentiation, type 2 innate lymphoid cell (ILC2) development and regulation of interleukin-4 (IL-4) and IL-13 production. Cytokine. 2015;75(1):14–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cyto.2015.05.010.
Keeler SP, Agapov EV, Hinojosa ME, Letvin AN, Wu K, Holtzman MJ. Influenza A virus infection causes chronic lung disease linked to sites of active viral RNA remnants. J Immunol. 2018;201(8):2354–68. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.1800671.
Gour N, Wills-Karp M. IL-4 and IL-13 signaling in allergic airway disease. Cytokine. 2015;75(1):68–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cyto.2015.05.014.
Wynn TA. IL-13 effector functions. Annu Rev Immunol. 2003;21:425–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev.immunol.21.120601.141142.
Kasaian MT, Miller DK. IL-13 as a therapeutic target for respiratory disease. Biochem Pharmacol. 2008;76(2):147–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bcp.2008.04.002.
Shastri MD, Allam VSRR, Shukla SD, et al. Interleukin-13: a pivotal target against influenza-induced exacerbation of chronic lung diseases. Life Sci. 2021;283: 119871. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2021.119871.
Germain PL, Sonrel A, Robinson MD. pipeComp, a general framework for the evaluation of computational pipelines, reveals performant single cell RNA-seq preprocessing tools. Genome Biol. 2020;21(1):227. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13059-020-02136-7. (Published 2020 Sep 1).
Fleming SJ, Chaffin MD, Arduini A, et al. Unsupervised removal of systematic background noise from droplet-based single-cell experiments using Cell Bender. Nat Methods. 2023;20(9):1323–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41592-023-01943-7.
Wolock SL, Lopez R, Klein AM. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 2019;8(4):281-291.e9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cels.2018.11.005.
Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019;177(7):1888-1902.e21. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2019.05.031.
Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184(13):3573-3587.e29. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2021.04.048.
Hao Y, Stuart T, Kowalski MH, et al. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat Biotechnol. 2024;42(2):293–304. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41587-023-01767-y.
Sikkema L, RamĂrez-Suástegui C, Strobl DC, et al. An integrated cell atlas of the lung in health and disease. Nat Med. 2023;29(6):1563–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41591-023-02327-2.
Crue T, Lee GY, Peng JY, et al. Single cell RNA-sequencing of human precision-cut lung slices: a novel approach to study the effect of vaping and viral infection on lung health. Innate Immun. 2023;29(5):61–70. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/17534259231181029.
Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods. 2019;16(12):1289–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41592-019-0619-0.
Choudhary S, Satija R. Comparison and evaluation of statistical error models for scRNA-seq. Genome Biol. 2022;23(1):27. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13059-021-02584-9. (Published 2022 Jan 18).
Yu D, Huber W, Vitek O. Shrinkage estimation of dispersion in Negative Binomial models for RNA-seq experiments with small sample size. Bioinformatics. 2013;29(10):1275–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/bioinformatics/btt143.
Robinson MD, Smyth GK. Small-sample estimation of negative binomial dispersion, with applications to SAGE data. Biostatistics. 2008;9(2):321–32. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/biostatistics/kxm030.
Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0506580102.
Korotkevich G, Sukhov V, Sergushichev A. Fast gene set enrichment analysis. BioRxiv. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/060012. (Published online June 20).
Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1(6):417–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cels.2015.12.004.
Tilley AE, Walters MS, Shaykhiev R, Crystal RG. Cilia dysfunction in lung disease. Annu Rev Physiol. 2015;77:379–406. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-physiol-021014-071931.
Brooks ER, Wallingford JB. Multiciliated cells. Curr Biol. 2014;24(19):R973–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cub.2014.08.047.
Yu H, Li Q, Kolosov VP, Perelman JM, Zhou X. Interleukin-13 induces mucin 5AC production involving STAT6/SPDEF in human airway epithelial cells. Cell Commun Adhes. 2010;17(4–6):83–92.
Romano GS, Lei L, Yu W, Thurman AL, Gansemer ND, Meyerholz DK, et al. Il-13 induces loss of CFTR in ionocytes and reduces airway epithelial fluid absorption. J Clin Investig. 2024;134: e181995.
Jackson ND, Everman JL, Chioccioli M, et al. Single-cell and population transcriptomics reveal pan-epithelial remodeling in type 2-high asthma. Cell Rep. 2020;32(1): 107872. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.celrep.2020.107872.
Pelaia C, Heffler E, Crimi C, Maglio A, Vatrella A, Pelaia G, et al. Interleukins 4 and 13 in asthma: key pathophysiologic cytokines and druggable molecular targets. Front Pharmacol. 2022;8:13.
Burgess JK, Jonker MR, Berg M, et al. Periostin: contributor to abnormal airway epithelial function in asthma? Eur Respir J. 2021;57(2):2001286. https://doiorg.publicaciones.saludcastillayleon.es/10.1183/13993003.01286-2020. (Published 2021 Feb 17).
Takayama G, Arima K, Kanaji T, et al. Periostin: a novel component of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J Allergy Clin Immunol. 2006;118(1):98–104. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2006.02.046.
Xu X, Li J, Zhang Y, Zhang L. Arachidonic acid 15-lipoxygenase: effects of its expression, metabolites, and genetic and epigenetic variations on airway inflammation. Allergy Asthma Immunol Res. 2021;13(5):684–96. https://doiorg.publicaciones.saludcastillayleon.es/10.4168/aair.2021.13.5.684.
Larose MC, Chakir J, Archambault AS, et al. Correlation between CCL26 production by human bronchial epithelial cells and airway eosinophils: Involvement in patients with severe eosinophilic asthma. J Allergy Clin Immunol. 2015;136(4):904–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2015.02.039.
Manna SK, Aggarwal BB. IL-13 suppresses TNF-induced activation of nuclear factor-kappa B, activation protein-1, and apoptosis. J Immunol. 1998;161(6):2863–72.
Muchamuel T, Menon S, Pisacane P, Howard MC, Cockayne DA. IL-13 protects mice from lipopolysaccharide-induced lethal endotoxemia: correlation with down-modulation of TNF-alpha, IFN-gamma, and IL-12 production. J Immunol. 1997;158(6):2898–903.
Iwaszko M, Biały S, Bogunia-Kubik K. Significance of Interleukin (IL)-4 and IL-13 in Inflammatory Arthritis. Cells. 2021;10(11):3000. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells10113000. (Published 2021 Nov 3).
Wu Q, Jiang D, Smith S, Thaikoottathil J, Martin RJ, Bowler RP, et al. IL-13 dampens human airway epithelial innate immunity through induction of IL-1 receptor–associated kinase M. J Allergy Clin Immunol. 2011;129(3):825-833.e2.
Caucheteux SM, Hu-Li J, Guo L, et al. IL-1β enhances inflammatory TH2 differentiation. J Allergy Clin Immunol. 2016;138(3):898-901.e4. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jaci.2016.02.033.
Wills-Karp M. Interleukin-13 in asthma pathogenesis. Immunol Rev. 2004;202:175–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.0105-2896.2004.00215.x.
Ford JG, Rennick D, Donaldson DD, et al. Il-13 and IFN-gamma: interactions in lung inflammation. J Immunol. 2001;167(3):1769–77. https://doiorg.publicaciones.saludcastillayleon.es/10.4049/jimmunol.167.3.1769.
Kondo M, Tamaoki J, Takeyama K, et al. Elimination of IL-13 reverses established goblet cell metaplasia into ciliated epithelia in airway epithelial cell culture. Allergol Int. 2006;55(3):329–36. https://doiorg.publicaciones.saludcastillayleon.es/10.2332/allergolint.55.329.
Laoukili J, Perret E, Willems T, et al. IL-13 alters mucociliary differentiation and ciliary beating of human respiratory epithelial cells. J Clin Invest. 2001;108(12):1817–24. https://doiorg.publicaciones.saludcastillayleon.es/10.1172/JCI13557.
Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrm3758.
Yan Z, Dai J, Wang J, et al. RNF167-mediated ubiquitination of Tollip inhibits TNF-α-triggered NF-κB and MAPK activation. FASEB J. 2023;37(8): e23089. https://doiorg.publicaciones.saludcastillayleon.es/10.1096/fj.202201839R.
Schaunaman N, Dimasuay KG, Kraft M, Chu HW. Tollip interaction with STAT3: a novel mechanism to regulate human airway epithelial responses to type 2 cytokines. Respir Res. 2022;23(1):31. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12931-022-01941-x. (Published 2022 Feb 16).
Funding
This research was funded by the National Institutes of Health (NIH) U19AI125357, R01AI150082, R01AI152504 and R01AI161296. The NIH does not have a role in any of the stages of this work.
Author information
Authors and Affiliations
Contributions
G.Y.L. conducted the data analysis and interpretation and wrote the manuscript. N.S. and H.R.N. performed the cell culture experiment. M.K. and H.W.C. contributed to the experimental design and manuscript editing.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
This study, protocol #HS-3209, was determined to be not human research by “the Institutional Review Board (IRB) at National Jewish Health”. Our research project used only banked and de-identified donor lung cells. The need for consent to participate was waived by the IRB at National Jewish Health since this was considered not human research.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Additional file 2. Cell count and percentage composition for each cell type and treatment condition.
13104_2025_7255_MOESM3_ESM.xlsx
Additional file 3. The raw data from the Gene Set Enrichment Analysis (GSEA) are presented in this table. The three left-most columns indicate the comparison groups and cell types in which each pathway is significantly enriched, along with the direction of expression change (upregulated or downregulated). The right-most column lists the leading-edge genes, which are the significantly differentially expressed genes that contribute to the pathway’s significance. We discussed some of these genes in our results section. The pathways themselves are illustrated in Figures 2, 3.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Lee, G.Y., Schaunaman, N., Nouri, H.R. et al. Comprehensive single-cell RNA-sequencing study of Tollip deficiency effect in IL-13-stimulated human airway epithelial cells. BMC Res Notes 18, 194 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07255-7
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07255-7