Skip to main content
Erschienen in: Inflammation Research 1/2023

01.11.2022 | Original Research Paper

Interleukin-19 upregulates fibronectin and collagen I expression via the NF-κB-Smad2/3 pathway in fibroblasts of patients with chronic rhinosinusitis

verfasst von: Hongwei Bao, Xia Li, Xiaoping Lai, Xiaohong Chen, Yue Li, Zhouzhou Yao, Zizhen Huang, Jiancong Huang, Lihong Chang, Gehua Zhang

Erschienen in: Inflammation Research | Ausgabe 1/2023

Einloggen, um Zugang zu erhalten

Abstract

Background

Tissue remodeling is a prominent characteristic of chronic rhinosinusitis (CRS). Excess deposition of fibronectin (FN) and collagen (Col) I by fibroblasts is crucial for the pathologic tissue remodeling in CRS without nasal polyps (CRSsNP). Increased interleukin (IL)-19 level in patients with CRS had been demonstrated in our previous studies. Here, we aimed to evaluate the role of IL-19 in mediating FN and Col I expression in CRS.

Methods

Nasal mucosal tissue samples were collected from patients with CRS with nasal polyps (CRSwNP), CRSsNP, and controls. The expression of IL-19, vimentin, FN, and Col I were detected using immunohistochemistry and immunofluorescence. Primary human nasal fibroblasts were treated with IL-19, then the activation of Smad2/3, NF-κB and relevant pathways, and the expression of FN and Col I were measured.

Results

Expression levels of vimentin, FN, and Col I were significantly increased in nasal tissues from patients with CRSsNP compared with CRSwNP and control subjects. Moreover, IL-19 co-localized with FN and Col Ι in nasal tissues. IL-19-treated fibroblasts had increased production of FN and Col I, which was associated with the activated Smad2/3 and NF-κB pathways. Moreover, Smad2/3 activation was mediated by the NF-κB pathway in IL-19-treated fibroblasts.

Conclusions

IL-19 promotes FN and Col I production via the activated NF-κB-Smad2/3 pathway in fibroblasts, leading to fibrosis and collagen deposition in patients with CRS.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Fokkens WJ, Lund VJ, Hopkins C, Hellings PW, Kern R, Reitsma S, et al. European position paper on rhinosinusitis and nasal polyps 2020. Rhinology. 2020;58:1–464.CrossRef Fokkens WJ, Lund VJ, Hopkins C, Hellings PW, Kern R, Reitsma S, et al. European position paper on rhinosinusitis and nasal polyps 2020. Rhinology. 2020;58:1–464.CrossRef
2.
Zurück zum Zitat Bhattacharyya N, Orlandi RR, Grebner J, Martinson M. Cost burden of chronic rhinosinusitis: a claims-based study. Otolaryngol Head Neck Surg. 2011;144:440–5.CrossRef Bhattacharyya N, Orlandi RR, Grebner J, Martinson M. Cost burden of chronic rhinosinusitis: a claims-based study. Otolaryngol Head Neck Surg. 2011;144:440–5.CrossRef
3.
Zurück zum Zitat Fokkens WJ, Lund VJ, Mullol J, Bachert C, Alobid I, Baroody F, et al. EPOS 2012: European position paper on rhinosinusitis and nasal polyps 2012. A summary for otorhinolaryngologists. Rhinology. 2012;50:1–12.CrossRef Fokkens WJ, Lund VJ, Mullol J, Bachert C, Alobid I, Baroody F, et al. EPOS 2012: European position paper on rhinosinusitis and nasal polyps 2012. A summary for otorhinolaryngologists. Rhinology. 2012;50:1–12.CrossRef
4.
Zurück zum Zitat Stevens WW, Lee RJ, Schleimer RP, Cohen NA. Chronic rhinosinusitis pathogenesis. J Allergy Clin Immunol. 2015;136:1442–53.CrossRef Stevens WW, Lee RJ, Schleimer RP, Cohen NA. Chronic rhinosinusitis pathogenesis. J Allergy Clin Immunol. 2015;136:1442–53.CrossRef
5.
Zurück zum Zitat Akdis CA, Bachert C, Cingi C, Dykewicz MS, Hellings PW, Naclerio RM, et al. Endotypes and phenotypes of chronic rhinosinusitis: a PRACTALL document of the European Academy of Allergy and Clinical Immunology and the American Academy of Allergy, Asthma & Immunology. J Allergy Clin Immunol. 2013;131:1479–90.CrossRef Akdis CA, Bachert C, Cingi C, Dykewicz MS, Hellings PW, Naclerio RM, et al. Endotypes and phenotypes of chronic rhinosinusitis: a PRACTALL document of the European Academy of Allergy and Clinical Immunology and the American Academy of Allergy, Asthma & Immunology. J Allergy Clin Immunol. 2013;131:1479–90.CrossRef
6.
Zurück zum Zitat Van Bruaene N, Bachert C. Tissue remodeling in chronic rhinosinusitis. Curr Opin Allergy Clin Immunol. 2011;11:8–11.CrossRef Van Bruaene N, Bachert C. Tissue remodeling in chronic rhinosinusitis. Curr Opin Allergy Clin Immunol. 2011;11:8–11.CrossRef
7.
Zurück zum Zitat Bassiouni A, Chen PG, Wormald PJ. Mucosal remodeling and reversibility in chronic rhinosinusitis. Curr Opin Allergy Clin Immunol. 2013;13:4–12.CrossRef Bassiouni A, Chen PG, Wormald PJ. Mucosal remodeling and reversibility in chronic rhinosinusitis. Curr Opin Allergy Clin Immunol. 2013;13:4–12.CrossRef
8.
Zurück zum Zitat Lee K, Tai J, Lee SH, Kim TH. Advances in the knowledge of the underlying airway remodeling mechanisms in chronic rhinosinusitis based on the endotypes: a review. Int J Mol Sci. 2021;22:910.CrossRef Lee K, Tai J, Lee SH, Kim TH. Advances in the knowledge of the underlying airway remodeling mechanisms in chronic rhinosinusitis based on the endotypes: a review. Int J Mol Sci. 2021;22:910.CrossRef
9.
Zurück zum Zitat Filer A, Pitzalis C, Buckley CD. Targeting the stromal microenvironment in chronic inflammation. Curr Opin Pharmacol. 2006;6:393–400.CrossRef Filer A, Pitzalis C, Buckley CD. Targeting the stromal microenvironment in chronic inflammation. Curr Opin Pharmacol. 2006;6:393–400.CrossRef
10.
Zurück zum Zitat de Castro Brás LE, Frangogiannis NG. Extracellular matrix-derived peptides in tissue remodeling and fibrosis. Matrix Biol. 2020;91–92:176–87.CrossRef de Castro Brás LE, Frangogiannis NG. Extracellular matrix-derived peptides in tissue remodeling and fibrosis. Matrix Biol. 2020;91–92:176–87.CrossRef
11.
Zurück zum Zitat Paulin D, Lilienbaum A, Kardjian S, Agbulut O, Li Z. Vimentin: regulation and pathogenesis. Biochimie. 2022;197:96–112.CrossRef Paulin D, Lilienbaum A, Kardjian S, Agbulut O, Li Z. Vimentin: regulation and pathogenesis. Biochimie. 2022;197:96–112.CrossRef
12.
Zurück zum Zitat Van Bruaene N, Derycke L, Perez-Novo CA, Gevaert P, Holtappels G, De Ruyck N, et al. TGF-beta signaling and collagen deposition in chronic rhinosinusitis. J Allergy Clin Immunol. 2009;124(253–9):259.e1-2. Van Bruaene N, Derycke L, Perez-Novo CA, Gevaert P, Holtappels G, De Ruyck N, et al. TGF-beta signaling and collagen deposition in chronic rhinosinusitis. J Allergy Clin Immunol. 2009;124(253–9):259.e1-2.
13.
Zurück zum Zitat Mueller SK, Nocera AL, Dillon ST, Wu D, Libermann TA, Bleier BS. Highly multiplexed proteomic analysis reveals significant tissue and exosomal coagulation pathway derangement in chronic rhinosinusitis with nasal polyps. Int Forum Allergy Rhinol. 2018;8:1438–44.CrossRef Mueller SK, Nocera AL, Dillon ST, Wu D, Libermann TA, Bleier BS. Highly multiplexed proteomic analysis reveals significant tissue and exosomal coagulation pathway derangement in chronic rhinosinusitis with nasal polyps. Int Forum Allergy Rhinol. 2018;8:1438–44.CrossRef
14.
Zurück zum Zitat Shieh JM, Tsai YJ, Chi JC, Wu WB. TGFβ mediates collagen production in human CRSsNP nasal mucosa-derived fibroblasts through Smad2/3-dependent pathway and CTGF induction and secretion. J Cell Physiol. 2019;234:10489–99.CrossRef Shieh JM, Tsai YJ, Chi JC, Wu WB. TGFβ mediates collagen production in human CRSsNP nasal mucosa-derived fibroblasts through Smad2/3-dependent pathway and CTGF induction and secretion. J Cell Physiol. 2019;234:10489–99.CrossRef
15.
Zurück zum Zitat Bignold R, Johnson JR. Effects of cytokine signaling inhibition on inflammation-driven tissue remodeling. Curr Res Pharmacol Drug Discov. 2021;2: 100023.CrossRef Bignold R, Johnson JR. Effects of cytokine signaling inhibition on inflammation-driven tissue remodeling. Curr Res Pharmacol Drug Discov. 2021;2: 100023.CrossRef
16.
Zurück zum Zitat Rutz S, Wang X, Ouyang W. The IL-20 subfamily of cytokines–from host defence to tissue homeostasis. Nat Rev Immunol. 2014;14:783–95.CrossRef Rutz S, Wang X, Ouyang W. The IL-20 subfamily of cytokines–from host defence to tissue homeostasis. Nat Rev Immunol. 2014;14:783–95.CrossRef
17.
Zurück zum Zitat Wang Y, Sun S, Wang K, Zhang M, Li M, Zan Y, et al. Interleukin-19 aggravates pulmonary fibrosis via activating fibroblast through TGF-β/Smad pathway. Mediators Inflamm. 2022;2022:6755407.CrossRef Wang Y, Sun S, Wang K, Zhang M, Li M, Zan Y, et al. Interleukin-19 aggravates pulmonary fibrosis via activating fibroblast through TGF-β/Smad pathway. Mediators Inflamm. 2022;2022:6755407.CrossRef
18.
Zurück zum Zitat Liu J, Shang B, Bai J. IL-22/IL-22R1 promotes proliferation and collagen synthesis of MRC-5 cells via the JAK/STAT3 signaling pathway and regulates airway subepithelial fibrosis. Exp Ther Med. 2020;20:2148–56. Liu J, Shang B, Bai J. IL-22/IL-22R1 promotes proliferation and collagen synthesis of MRC-5 cells via the JAK/STAT3 signaling pathway and regulates airway subepithelial fibrosis. Exp Ther Med. 2020;20:2148–56.
19.
Zurück zum Zitat Chiu YS, Wei CC, Lin YJ, Hsu YH, Chang MS. IL-20 and IL-20R1 antibodies protect against liver fibrosis. Hepatology. 2014;60:1003–14.CrossRef Chiu YS, Wei CC, Lin YJ, Hsu YH, Chang MS. IL-20 and IL-20R1 antibodies protect against liver fibrosis. Hepatology. 2014;60:1003–14.CrossRef
20.
Zurück zum Zitat Mathur R, Alam MM, Zhao XF, Liao Y, Shen J, Morgan S, et al. Induction of autophagy in Cx3cr1(+) mononuclear cells limits IL-23/IL-22 axis-mediated intestinal fibrosis. Mucosal Immunol. 2019;12:612–23.CrossRef Mathur R, Alam MM, Zhao XF, Liao Y, Shen J, Morgan S, et al. Induction of autophagy in Cx3cr1(+) mononuclear cells limits IL-23/IL-22 axis-mediated intestinal fibrosis. Mucosal Immunol. 2019;12:612–23.CrossRef
21.
Zurück zum Zitat Ohnuma K, Hatano R, Aune TM, Otsuka H, Iwata S, Dang NH, et al. Regulation of pulmonary graft-versus-host disease by IL-26+CD26+CD4 T lymphocytes. J Immunol. 2015;194:3697–712.CrossRef Ohnuma K, Hatano R, Aune TM, Otsuka H, Iwata S, Dang NH, et al. Regulation of pulmonary graft-versus-host disease by IL-26+CD26+CD4 T lymphocytes. J Immunol. 2015;194:3697–712.CrossRef
22.
Zurück zum Zitat Li X, Huang J, Chen X, Lai X, Huang Z, Li Y, et al. IL-19 induced by IL-13/IL-17A in the nasal epithelium of patients with chronic rhinosinusitis upregulates MMP-9 expression via ERK/NF-κB signaling pathway. Clin Transl Allergy. 2021;11: e12003.CrossRef Li X, Huang J, Chen X, Lai X, Huang Z, Li Y, et al. IL-19 induced by IL-13/IL-17A in the nasal epithelium of patients with chronic rhinosinusitis upregulates MMP-9 expression via ERK/NF-κB signaling pathway. Clin Transl Allergy. 2021;11: e12003.CrossRef
23.
Zurück zum Zitat Lai X, Li X, Chang L, Chen X, Huang Z, Bao H, et al. IL-19 up-regulates mucin 5AC production in patients with chronic rhinosinusitis via STAT3 pathway. Front Immunol. 2019;10:1682.CrossRef Lai X, Li X, Chang L, Chen X, Huang Z, Bao H, et al. IL-19 up-regulates mucin 5AC production in patients with chronic rhinosinusitis via STAT3 pathway. Front Immunol. 2019;10:1682.CrossRef
24.
Zurück zum Zitat Chen X, Chang L, Li X, Huang J, Yang L, Lai X, et al. Tc17/IL-17A up-regulated the expression of MMP-9 via NF-κB pathway in nasal epithelial cells of patients with chronic rhinosinusitis. Front Immunol. 2018;9:2121.CrossRef Chen X, Chang L, Li X, Huang J, Yang L, Lai X, et al. Tc17/IL-17A up-regulated the expression of MMP-9 via NF-κB pathway in nasal epithelial cells of patients with chronic rhinosinusitis. Front Immunol. 2018;9:2121.CrossRef
25.
Zurück zum Zitat Xu M, Li S, Wang J, Huang S, Zhang A, Zhang Y, et al. Cilomilast ameliorates renal tubulointerstitial fibrosis by inhibiting the TGF-β1-Smad2/3 signaling pathway. Front Med (Lausanne). 2020;7: 626140.CrossRef Xu M, Li S, Wang J, Huang S, Zhang A, Zhang Y, et al. Cilomilast ameliorates renal tubulointerstitial fibrosis by inhibiting the TGF-β1-Smad2/3 signaling pathway. Front Med (Lausanne). 2020;7: 626140.CrossRef
26.
Zurück zum Zitat Khalil H, Kanisicak O, Prasad V, Correll RN, Fu X, Schips T, et al. Fibroblast-specific TGF-β-Smad2/3 signaling underlies cardiac fibrosis. J Clin Invest. 2017;127:3770–83.CrossRef Khalil H, Kanisicak O, Prasad V, Correll RN, Fu X, Schips T, et al. Fibroblast-specific TGF-β-Smad2/3 signaling underlies cardiac fibrosis. J Clin Invest. 2017;127:3770–83.CrossRef
27.
Zurück zum Zitat Xie Y, Du D, Zhang L, Yang Y, Zou Z, Li Z, et al. TJ-M2010-5, A self-developed MyD88 inhibitor, attenuates liver fibrosis by inhibiting the NF-κB pathway. Chem Biol Interact. 2022;354: 109839.CrossRef Xie Y, Du D, Zhang L, Yang Y, Zou Z, Li Z, et al. TJ-M2010-5, A self-developed MyD88 inhibitor, attenuates liver fibrosis by inhibiting the NF-κB pathway. Chem Biol Interact. 2022;354: 109839.CrossRef
28.
Zurück zum Zitat Li H, Duann P, Li Z, Zhou X, Ma J, Rovin BH, et al. The cell membrane repair protein MG53 modulates transcription factor NF-κB signaling to control kidney fibrosis. Kidney Int. 2022;101:119–30.CrossRef Li H, Duann P, Li Z, Zhou X, Ma J, Rovin BH, et al. The cell membrane repair protein MG53 modulates transcription factor NF-κB signaling to control kidney fibrosis. Kidney Int. 2022;101:119–30.CrossRef
29.
Zurück zum Zitat Saber S, Nasr M, Kaddah M, Mostafa-Hedeab G, Cavalu S, Mourad A, et al. Nifuroxazide-loaded cubosomes exhibit an advancement in pulmonary delivery and attenuate bleomycin-induced lung fibrosis by regulating the STAT3 and NF-κB signaling: a new challenge for unmet therapeutic needs. Biomed Pharmacother. 2022;148: 112731.CrossRef Saber S, Nasr M, Kaddah M, Mostafa-Hedeab G, Cavalu S, Mourad A, et al. Nifuroxazide-loaded cubosomes exhibit an advancement in pulmonary delivery and attenuate bleomycin-induced lung fibrosis by regulating the STAT3 and NF-κB signaling: a new challenge for unmet therapeutic needs. Biomed Pharmacother. 2022;148: 112731.CrossRef
30.
Zurück zum Zitat Baghaei K, Mazhari S, Tokhanbigli S, Parsamanesh G, Alavifard H, Schaafsma D, et al. Therapeutic potential of targeting regulatory mechanisms of hepatic stellate cell activation in liver fibrosis. Drug Discov Today. 2022;27:1044–61.CrossRef Baghaei K, Mazhari S, Tokhanbigli S, Parsamanesh G, Alavifard H, Schaafsma D, et al. Therapeutic potential of targeting regulatory mechanisms of hepatic stellate cell activation in liver fibrosis. Drug Discov Today. 2022;27:1044–61.CrossRef
31.
Zurück zum Zitat Radajewski K, Wierzchowska M, Grzanka D, Antosik P, Zdrenka M, Burduk P. Tissue remodelling in chronic rhinosinusitis—review of literature. Otolaryngol Pol. 2019;73:1–4. Radajewski K, Wierzchowska M, Grzanka D, Antosik P, Zdrenka M, Burduk P. Tissue remodelling in chronic rhinosinusitis—review of literature. Otolaryngol Pol. 2019;73:1–4.
32.
Zurück zum Zitat Kendall RT, Feghali-Bostwick CA. Fibroblasts in fibrosis: novel roles and mediators. Front Pharmacol. 2014;5:123.CrossRef Kendall RT, Feghali-Bostwick CA. Fibroblasts in fibrosis: novel roles and mediators. Front Pharmacol. 2014;5:123.CrossRef
33.
Zurück zum Zitat Molet SM, Hamid QA, Hamilos DL. IL-11 and IL-17 expression in nasal polyps: relationship to collagen deposition and suppression by intranasal fluticasone propionate. Laryngoscope. 2003;113:1803–12.CrossRef Molet SM, Hamid QA, Hamilos DL. IL-11 and IL-17 expression in nasal polyps: relationship to collagen deposition and suppression by intranasal fluticasone propionate. Laryngoscope. 2003;113:1803–12.CrossRef
34.
Zurück zum Zitat Hupin C, Gohy S, Bouzin C, Lecocq M, Polette M, Pilette C. Features of mesenchymal transition in the airway epithelium from chronic rhinosinusitis. Allergy. 2014;69:1540–9.CrossRef Hupin C, Gohy S, Bouzin C, Lecocq M, Polette M, Pilette C. Features of mesenchymal transition in the airway epithelium from chronic rhinosinusitis. Allergy. 2014;69:1540–9.CrossRef
35.
Zurück zum Zitat Kuhar HN, Tajudeen BA, Mahdavinia M, Gattuso P, Ghai R, Batra PS. Inflammatory infiltrate and mucosal remodeling in chronic rhinosinusitis with and without polyps: structured histopathologic analysis. Int Forum Allergy Rhinol. 2017;7:679–89.CrossRef Kuhar HN, Tajudeen BA, Mahdavinia M, Gattuso P, Ghai R, Batra PS. Inflammatory infiltrate and mucosal remodeling in chronic rhinosinusitis with and without polyps: structured histopathologic analysis. Int Forum Allergy Rhinol. 2017;7:679–89.CrossRef
36.
Zurück zum Zitat Peng Y, Zi XX, Tian TF, Lee B, Lum J, Tang SA, et al. Whole-transcriptome sequencing reveals heightened inflammation and defective host defence responses in chronic rhinosinusitis with nasal polyps. Eur Respir J. 2019;54:1900732.CrossRef Peng Y, Zi XX, Tian TF, Lee B, Lum J, Tang SA, et al. Whole-transcriptome sequencing reveals heightened inflammation and defective host defence responses in chronic rhinosinusitis with nasal polyps. Eur Respir J. 2019;54:1900732.CrossRef
37.
Zurück zum Zitat Soy FK, Pinar E, Imre A, Calli C, Calli A, Oncel S. Histopathologic parameters in chronic rhinosinusitis with nasal polyposis: impact on quality of life outcomes. Int Forum Allergy Rhinol. 2013;3:828–33.CrossRef Soy FK, Pinar E, Imre A, Calli C, Calli A, Oncel S. Histopathologic parameters in chronic rhinosinusitis with nasal polyposis: impact on quality of life outcomes. Int Forum Allergy Rhinol. 2013;3:828–33.CrossRef
38.
Zurück zum Zitat Do TQ, Barham HP, Earls P, Sacks R, Christensen JM, Rimmer J, et al. Clinical implications of mucosal remodeling from chronic rhinosinusitis. Int Forum Allergy Rhinol. 2016;6:835–40.CrossRef Do TQ, Barham HP, Earls P, Sacks R, Christensen JM, Rimmer J, et al. Clinical implications of mucosal remodeling from chronic rhinosinusitis. Int Forum Allergy Rhinol. 2016;6:835–40.CrossRef
39.
Zurück zum Zitat Zou Y, Chen Z, Li J, Gong W, Zhang L, Xu F, et al. Progestin and AdipoQ receptor 3 upregulates fibronectin and intercellular adhesion molecule-1 in glomerular mesangial cells via activating NF-κB signaling pathway under high glucose conditions. Front Endocrinol (Lausanne). 2018;9:275.CrossRef Zou Y, Chen Z, Li J, Gong W, Zhang L, Xu F, et al. Progestin and AdipoQ receptor 3 upregulates fibronectin and intercellular adhesion molecule-1 in glomerular mesangial cells via activating NF-κB signaling pathway under high glucose conditions. Front Endocrinol (Lausanne). 2018;9:275.CrossRef
40.
Zurück zum Zitat Finnson KW, Almadani Y, Philip A. Non-canonical (non-SMAD2/3) TGF-β signaling in fibrosis: mechanisms and targets. Semin Cell Dev Biol. 2020;101:115–22.CrossRef Finnson KW, Almadani Y, Philip A. Non-canonical (non-SMAD2/3) TGF-β signaling in fibrosis: mechanisms and targets. Semin Cell Dev Biol. 2020;101:115–22.CrossRef
41.
Zurück zum Zitat Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023.CrossRef Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023.CrossRef
42.
Zurück zum Zitat Park SA, Kim MJ, Park SY, Kim JS, Lee SJ, Woo HA, et al. EW-7197 inhibits hepatic, renal, and pulmonary fibrosis by blocking TGF-β/Smad and ROS signaling. Cell Mol Life Sci. 2015;72:2023–39.CrossRef Park SA, Kim MJ, Park SY, Kim JS, Lee SJ, Woo HA, et al. EW-7197 inhibits hepatic, renal, and pulmonary fibrosis by blocking TGF-β/Smad and ROS signaling. Cell Mol Life Sci. 2015;72:2023–39.CrossRef
43.
Zurück zum Zitat Hsing CH, Kwok FA, Cheng HC, Li CF, Chang MS. Inhibiting interleukin-19 activity ameliorates esophageal squamous cell carcinoma progression. PLoS ONE. 2013;8: e75254.CrossRef Hsing CH, Kwok FA, Cheng HC, Li CF, Chang MS. Inhibiting interleukin-19 activity ameliorates esophageal squamous cell carcinoma progression. PLoS ONE. 2013;8: e75254.CrossRef
44.
Zurück zum Zitat Aazami H, Seif F, Ghalehbaghi B, Babaheidarian P, Mohebbi A, Ahmadi A, et al. Local eosinophils are associated with increased IgA subclass levels in the sinonasal mucosa of chronic rhinosinusitis with polyp patients. Allergy Asthma Clin Immunol. 2020;16:30.CrossRef Aazami H, Seif F, Ghalehbaghi B, Babaheidarian P, Mohebbi A, Ahmadi A, et al. Local eosinophils are associated with increased IgA subclass levels in the sinonasal mucosa of chronic rhinosinusitis with polyp patients. Allergy Asthma Clin Immunol. 2020;16:30.CrossRef
45.
Zurück zum Zitat Könnecke M, Burmeister M, Pries R, Böscke R, Bruchhage KL, Ungefroren H, et al. Epithelial-mesenchymal transition in chronic rhinosinusitis: differences revealed between epithelial cells from nasal polyps and inferior turbinates. Arch Immunol Ther Exp (Warsz). 2017;65:157–73.CrossRef Könnecke M, Burmeister M, Pries R, Böscke R, Bruchhage KL, Ungefroren H, et al. Epithelial-mesenchymal transition in chronic rhinosinusitis: differences revealed between epithelial cells from nasal polyps and inferior turbinates. Arch Immunol Ther Exp (Warsz). 2017;65:157–73.CrossRef
Metadaten
Titel
Interleukin-19 upregulates fibronectin and collagen I expression via the NF-κB-Smad2/3 pathway in fibroblasts of patients with chronic rhinosinusitis
verfasst von
Hongwei Bao
Xia Li
Xiaoping Lai
Xiaohong Chen
Yue Li
Zhouzhou Yao
Zizhen Huang
Jiancong Huang
Lihong Chang
Gehua Zhang
Publikationsdatum
01.11.2022
Verlag
Springer International Publishing
Erschienen in
Inflammation Research / Ausgabe 1/2023
Print ISSN: 1023-3830
Elektronische ISSN: 1420-908X
DOI
https://doi.org/10.1007/s00011-022-01634-7

Weitere Artikel der Ausgabe 1/2023

Inflammation Research 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Bei Herzinsuffizienz muss „Eisenmangel“ neu definiert werden!

16.05.2024 Herzinsuffizienz Nachrichten

Bei chronischer Herzinsuffizienz macht es einem internationalen Expertenteam zufolge wenig Sinn, die Diagnose „Eisenmangel“ am Serumferritin festzumachen. Das Team schlägt vor, sich lieber an die Transferrinsättigung zu halten.

Herzinfarkt mit 85 – trotzdem noch intensive Lipidsenkung?

16.05.2024 Hypercholesterinämie Nachrichten

Profitieren nach einem akuten Myokardinfarkt auch Betroffene über 80 Jahre noch von einer intensiven Lipidsenkung zur Sekundärprävention? Um diese Frage zu beantworten, wurden jetzt Registerdaten aus Frankreich ausgewertet.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Erstmanifestation eines Diabetes-Typ-1 bei Kindern: Ein Notfall!

16.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Manifestiert sich ein Typ-1-Diabetes bei Kindern, ist das ein Notfall – ebenso wie eine diabetische Ketoazidose. Die Grundsäulen der Therapie bestehen aus Rehydratation, Insulin und Kaliumgabe. Insulin ist das Medikament der Wahl zur Behandlung der Ketoazidose.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.