Allergy Asthma Immunol Res.  2018 Nov;10(6):698-715. 10.4168/aair.2018.10.6.698.

Differential Hrd1 Expression and B-Cell Accumulation in Eosinophilic and Non-eosinophilic Chronic Rhinosinusitis With Nasal Polyps

Affiliations
  • 1Department of Otolaryngology-Head and Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. yangjun@xinhuamed.com.cn, allergyli@163.com
  • 2Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
  • 3Key Laboratory of Molecular Virology and Immunology, Vaccine Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
  • 4Department of Otolaryngology-Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China.

Abstract

PURPOSE
Hrd1 has recently emerged as a critical regulator of B-cells in autoimmune diseases. However, its role in the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP) remains largely unexplored. This study aimed to examine Hrd1 expression and B-cell accumulation and their possible roles in CRSwNP.
METHODS
Quantitative real-time polymerase chain reaction, immunohistochemistry, enzyme-linked immunosorbent assay and Western blotting were used to assess gene and protein expression in nasal tissue extracts. Cells isolated from nasal tissues and peripheral blood mononuclear cells were characterized by flow cytometry. Local antibody production was measured in tissue extracts with a Bio-Plex assay. Additionally, changes in Hrd1 expression in response to specific inflammatory stimuli were measured in cultured dispersed polyp cells.
RESULTS
Nasal polyps (NPs) from patients with eosinophilic CRSwNP (ECRS) had increased levels of Hrd1, B-cells and plasma cells compared with NPs from patients with non-eosinophilic CRSwNP (non-ECRS) or other control subjects (P < 0.05). The average Hrd1 levels in B-cells in NPs from ECRS patients were significantly higher than those from non-ECRS patients and control subjects (P < 0.05). NPs also contained significantly increased levels of several antibody isotypes compared with normal controls (P < 0.05). Interestingly, Hrd1 expression in cultured polyp cells from ECRS patients, but not non-ECRS patients, was significantly increased by interleukin-1β, lipopolysaccharide and Poly(I:C) stimulation, and inhibited by dexamethasone treatment (P < 0.05).
CONCLUSIONS
Differential Hrd1 expression and B-cell accumulation between the ECRS and non-ECRS subsets suggests that they can exhibit distinct pathogenic mechanisms and play important roles in NP.

Keyword

Hrd1; chronic rhinosinusitis; B-cells; nasal polyps; innate immunity

MeSH Terms

Antibody Formation
Autoimmune Diseases
B-Lymphocytes*
Blotting, Western
Dexamethasone
Enzyme-Linked Immunosorbent Assay
Eosinophils*
Flow Cytometry
Humans
Immunity, Innate
Immunohistochemistry
Nasal Polyps*
Plasma Cells
Polyps
Real-Time Polymerase Chain Reaction
Tissue Extracts
Dexamethasone
Tissue Extracts

Figure

  • Fig. 1 Hrd1 mRNA expression in nasal tissues. Hrd1 (A), CD19 (B), CD20 (C), CD138 (D) and BAFF (E) mRNA levels were significantly enhanced in polyp tissues from patients with ECRS compared with values seen in the other subgroups. Results are expressed as medians (interquartile ranges). BAFF, B-cell activating factor; ECRS, eosinophilic chronic rhinosinusitis with nasal polyp; non-ECRS, non-eosinophilic chronic rhinosinusitis with nasal polyp; CRSsNP, chronic rhinosinusitis without nasal polyps.

  • Fig. 2 Increased Hrd1 protein levels and accumulation of B cells and plasma cells in NPs. Representative IHC staining of Hrd1 (A), CD19 (C), CD20 (E), and CD138 (G) (magnification, × 400) in nasal tissues. Relative positive cells of Hrd1 (B), CD19 (D), CD20 (F), and CD138 (H) were significantly increased in polyp tissues from patients with ECRS relative to those in other subgroups. Representative Western blotting results (I), densitometric analysis (J). Positive correlation between relative Hrd1 protein levels in the polyp tissues and Lund-Mackay CT scores of the matched patients (K). ELISA of BAFF in nasal tissues (L). Positive correlation between BAFF and Hrd1 protein levels in the matched NPs (M). Bars = 20 μm. NP, nasal polyp; ECRS, eosinophilic chronic rhinosinusitis with nasal polyp; non-ECRS, non-eosinophilic chronic rhinosinusitis with nasal polyp; CRSsNP, chronic rhinosinusitis without nasal polyps; HPF, high-power field; CT, computed tomography; ELISA, enzyme-linked immunosorbent assay; BAFF, B-cell activating factor.

  • Fig. 3 Enhanced Hrd1 expression in B-cells from polyp tissues and matched PBMCs. Gating strategy and representative flow plots for nasal tissues (A). MFI levels of Hrd1 in CD19+ B-cells were significantly increased in polyp tissues from patients with ECRS compared with those in in polyps from patients with non-ECRS, middle turbinates from control subjects (B). Gating strategy and representative flow plots for PBMCs (C). MFI levels of Hrd1 in CD19+ B-cells were significantly increased in PBMCs from patients with CRSwNP compared with those in PBMCs from healthy controls (D). PBMC, peripheral blood mononuclear cell; MFI, mean fluorescence intensity; ECRS, eosinophilic chronic rhinosinusitis with nasal polyp; non-ECRS, non-eosinophilic chronic rhinosinusitis with nasal polyp; CRSwNP, chronic rhinosinusitis with nasal polyps.

  • Fig. 4 Increased local immunoglobulin levels in polyp tissues. Protein levels of IgA, IgE, IgM, IgG and IgG subclasses in tissue homogenates from ECRS, non-ECRS, CRSsNP, and control tissue are detected by using the Bio-Plex assay. Positive correlations are found between relative Hrd1 protein levels and IgM, IgG, IgG1, and IgG2. Ig, immunoglobulin; ECRS, eosinophilic chronic rhinosinusitis with nasal polyp; non-ECRS, non-eosinophilic chronic rhinosinusitis with nasal polyp; CRSsNP, chronic rhinosinusitis without nasal polyps.

  • Fig. 5 Hrd1 mRNA and protein levels in cultured DPCs in response to IL-1β, LPS, Poly(I:C) and Dex. mRNA expression and representative western blot results of DPCs lysates from ECRS patients after 20-hour stimulation (A-C). mRNA expression and representative western blot results of DPCs lysates from non-ECRS patients after 20-hour stimulation (D-F). Results represent mean values from 3 independent experiments. Data are expressed as means (standard error of the means). DPC, dispersed polyp cell; IL, interleukin; LPS, lipopolysaccharide; Poly(I:C), polyinosinic-polycytidylic acid; Dex, dexamethasone; ECRS, eosinophilic chronic rhinosinusitis with nasal polyp;non-ECRS, non-eosinophilic chronic rhinosinusitis with nasal polyp. *P < 0.05; †P < 0.01; ‡P < 0.001.


Reference

1. 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.
Article
2. Hsu J, Peters AT. Pathophysiology of chronic rhinosinusitis with nasal polyp. Am J Rhinol Allergy. 2011; 25:285–290.
Article
3. 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; 131:1479–1490.
4. Payne SC, Borish L, Steinke JW. Genetics and phenotyping in chronic sinusitis. J Allergy Clin Immunol. 2011; 128:710–720.
Article
5. Hulse KE. Immune mechanisms of chronic rhinosinusitis. Curr Allergy Asthma Rep. 2016; 16:1.
Article
6. Khalmuratova R, Park JW, Shin HW. Immune cell responses and mucosal barrier disruptions in chronic rhinosinusitis. Immune Netw. 2017; 17:60–67.
Article
7. Hulse KE, Stevens WW, Tan BK, Schleimer RP. Pathogenesis of nasal polyposis. Clin Exp Allergy. 2015; 45:328–346.
Article
8. Dörner T, Radbruch A, Burmester GR. B-cell-directed therapies for autoimmune disease. Nat Rev Rheumatol. 2009; 5:433–441.
Article
9. Kato A, Hulse KE, Tan BK, Schleimer RP. B-lymphocyte lineage cells and the respiratory system. J Allergy Clin Immunol. 2013; 131:933–957.
Article
10. Kato A, Peters A, Suh L, Carter R, Harris KE, Chandra R, et al. Evidence of a role for B cell-activating factor of the TNF family in the pathogenesis of chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2008; 121:1385–1392. 1392.e1–1382.
Article
11. Song J, Wang H, Zhang YN, Cao PP, Liao B, Wang ZZ, et al. Ectopic lymphoid tissues support local immunoglobulin production in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2018; 141:927–937.
Article
12. Van Zele T, Gevaert P, Watelet JB, Claeys G, Holtappels G, Claeys C, et al. Staphylococcus aureus colonization and IgE antibody formation to enterotoxins is increased in nasal polyposis. J Allergy Clin Immunol. 2004; 114:981–983.
Article
13. Tan BK, Li QZ, Suh L, Kato A, Conley DB, Chandra RK, et al. Evidence for intranasal antinuclear autoantibodies in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2011; 128:1198–1206.e1.
Article
14. Guerriero CJ, Brodsky JL. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev. 2012; 92:537–576.
Article
15. Amano T, Yamasaki S, Yagishita N, Tsuchimochi K, Shin H, Kawahara K, et al. Synoviolin/Hrd1, an E3 ubiquitin ligase, as a novel pathogenic factor for arthropathy. Genes Dev. 2003; 17:2436–2449.
Article
16. Gao B, Lee SM, Chen A, Zhang J, Zhang DD, Kannan K, et al. Synoviolin promotes IRE1 ubiquitination and degradation in synovial fibroblasts from mice with collagen-induced arthritis. EMBO Rep. 2008; 9:480–485.
Article
17. Toh ML, Marotte H, Blond JL, Jhumka U, Eljaafari A, Mougin B, et al. Overexpression of synoviolin in peripheral blood and synoviocytes from rheumatoid arthritis patients and continued elevation in nonresponders to infliximab treatment. Arthritis Rheum. 2006; 54:2109–2118.
Article
18. Xu Y, Zhao F, Qiu Q, Chen K, Wei J, Kong Q, et al. The ER membrane-anchored ubiquitin ligase Hrd1 is a positive regulator of T-cell immunity. Nat Commun. 2016; 7:12073.
Article
19. Kong S, Yang Y, Xu Y, Wang Y, Zhang Y, Melo-Cardenas J, et al. Endoplasmic reticulum-resident E3 ubiquitin ligase Hrd1 controls B-cell immunity through degradation of the death receptor CD95/Fas. Proc Natl Acad Sci U S A. 2016; 113:10394–10399.
Article
20. Wen W, Liu W, Zhang L, Bai J, Fan Y, Xia W, et al. Increased neutrophilia in nasal polyps reduces the response to oral corticosteroid therapy. J Allergy Clin Immunol. 2012; 129:1522–1528.e5.
Article
21. Wei Y, Xia W, Ye X, Fan Y, Shi J, Wen W, et al. The antimicrobial protein short palate, lung, and nasal epithelium clone 1 (SPLUNC1) is differentially modulated in eosinophilic and noneosinophilic chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2014; 133:420–428.e10.
Article
22. Ba L, Du J, Liu F, Yang F, Han M, Liu S, et al. Distinct inflammatory profiles in atopic and nonatopic patients with chronic rhinosinustis accompanied by nasal polyps in Western china. Allergy Asthma Immunol Res. 2015; 7:346–358.
Article
23. Lindqvist M, van Lunzen J, Soghoian DZ, Kuhl BD, Ranasinghe S, Kranias G, et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J Clin Invest. 2012; 122:3271–3280.
Article
24. Hulse KE, Norton JE, Suh L, Zhong Q, Mahdavinia M, Simon P, et al. Chronic rhinosinusitis with nasal polyps is characterized by B-cell inflammation and EBV-induced protein 2 expression. J Allergy Clin Immunol. 2013; 131:1075–1083. 1083.e1–1077.
Article
25. Dilidaer , Zheng Y, Liu Z, Hu X, Zhang J, Hu L, et al. Increased BAFF expression in nasal polyps is associated with local IgE production, Th2 response and concomitant asthma. Eur Arch Otorhinolaryngol. 2017; 274:1883–1890.
Article
26. Feldman S, Kasjanski R, Poposki J, Hernandez D, Chen JN, Norton JE, et al. Chronic airway inflammation provides a unique environment for B cell activation and antibody production. Clin Exp Allergy. 2017; 47:457–466.
Article
27. Chu VT, Fröhlich A, Steinhauser G, Scheel T, Roch T, Fillatreau S, et al. Eosinophils are required for the maintenance of plasma cells in the bone marrow. Nat Immunol. 2011; 12:151–159.
Article
28. Jacobsen EA, Helmers RA, Lee JJ, Lee NA. The expanding role(s) of eosinophils in health and disease. Blood. 2012; 120:3882–3890.
Article
29. Zhang YN, Song J, Wang H, Wang H, Zeng M, Zhai GT, et al. Nasal IL-4(+)CXCR5(+)CD4(+) T follicular helper cell counts correlate with local IgE production in eosinophilic nasal polyps. J Allergy Clin Immunol. 2016; 137:462–473.
Article
30. Schleimer RP. Immunopathogenesis of chronic rhinosinusitis and nasal polyposis. Annu Rev Pathol. 2017; 12:331–357.
Article
31. Hamilos DL. Host-microbial interactions in patients with chronic rhinosinusitis. J Allergy Clin Immunol. 2014; 133:640–653.e4.
Article
32. Ishii KJ, Koyama S, Nakagawa A, Coban C, Akira S. Host innate immune receptors and beyond: making sense of microbial infections. Cell Host Microbe. 2008; 3:352–363.
Article
33. Lund FE, Randall TD. Effector and regulatory B cells: modulators of CD4+ T cell immunity. Nat Rev Immunol. 2010; 10:236–247.
Article
34. Yang H, Qiu Q, Gao B, Kong S, Lin Z, Fang D. Hrd1-mediated BLIMP-1 ubiquitination promotes dendritic cell MHCII expression for CD4 T cell priming during inflammation. J Exp Med. 2014; 211:2467–2479.
Article
Full Text Links
  • AAIR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr