Next Article in Journal
Metabolomic and Transcriptomic Profiling Provide Novel Insights into Fruit Ripening and Ripening Disorder Caused by 1-MCP Treatments in Papaya
Next Article in Special Issue
Der f 38 Is a Novel TLR4-Binding Allergen Related to Allergy Pathogenesis from Dermatophagoides farinae
Previous Article in Journal
The Role of Gut Microbiota in an Ischemic Stroke
Previous Article in Special Issue
Immunologic and Non-Immunologic Mechanisms Leading to Airway Remodeling in Asthma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in the Knowledge of the Underlying Airway Remodeling Mechanisms in Chronic Rhinosinusitis Based on the Endotypes: A Review

Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(2), 910; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020910
Submission received: 29 December 2020 / Revised: 12 January 2021 / Accepted: 14 January 2021 / Published: 18 January 2021

Abstract

:
Chronic rhinosinusitis (CRS) is a chronic inflammatory condition of the nasal and paranasal sinus mucosa that affects up to 10% of the population worldwide. CRS is the most representative disease of the upper respiratory tract where airway remodeling occurs, including epithelial damage, thickening of the basement membrane, fibrosis, goblet cell hyperplasia, subepithelial edema, and osteitis. CRS is divided into two phenotypes according to the presence or absence of nasal polyps: CRS with nasal polyp (CRSwNP) and CRS without nasal polyps (CRSsNP). Based on the underlying pathophysiologic mechanism, CRS is also classified as eosinophilic CRS and non-eosinophilic CRS, owing to Type 2 T helper (Th2)-based inflammation and Type 1 T helper (Th1)/Type 17 T helper (Th17) skewed immune response, respectively. Differences in tissue remodeling in CRS are suggested to be based on the clinical phenotype and endotypes; this is because fibrosis is prominent in CRSsNP, whereas edematous changes occur in CRSwNP, especially in the eosinophilic type. This review aims to summarize the latest information on the different mechanisms of airway remodeling in CRS according to distinct endotypes.

1. Introduction

Chronic rhinosinusitis (CRS) is defined as a chronic inflammatory condition of the nasal and paranasal sinus mucosa, which affects approximately 12% of the population worldwide [1]. CRS is classified into the following two phenotypes based on the presence or absence of nasal polyps using endoscopic examination or computed tomographic findings: CRS with nasal polyps (CRSwNP) and CRS without nasal polyps (CRSsNP) [2,3]. Owing to the pathogenesis of CRS, there has been recent emphasis on the importance of classifying CRS based on the endotypes [4,5]. Previously, depending on the phenotype, CRSsNP was known to exhibit neutrophil dominance, whereas CRSwNP is an eosinophil-dominant type 2 inflammation [6]. However, unlike American and European patients, most Asian CRSwNP patients display neutrophil dominance, which accounts for more than 50% of cases. CRSsNP is reported to exhibit a type 2-biased immune response as well as type 1 or type 3 inflammation [7,8,9]. In addition, it has been identified that type 2 immune response is present in CRSsNP as well [10]. Thus, both phenotypes are presently further classified according to the endotypes, based on immune response characterization. In the European Position Paper on Rhinosinusitis and Nasal Polyps (EPOS) 2020, the classification of CRS was defined based on anatomic distribution (localized vs. diffuse) and endotype dominance (type 2 vs. non-type 2) rather than the presence of nasal polyp [1].
Tissue remodeling is defined as the abnormal reconstruction or restitution of the damaged tissue in relation to inflammation or mechanical injury that occurs in any organ. In lower airway diseases, including asthma, chronic obstructive pulmonary disease, and bronchiectasis, the concept of tissue remodeling has been studied in detail; however, only a few studies in relation to the upper airway have been carried out [11]. In asthma, airway remodeling, including goblet cell hyperplasia, subepithelial collagen deposition, thickened basement membrane, increased airway smooth muscle, and angiogenesis is affected by several underlying mechanisms and influences disease prognosis [12]. Allergic rhinitis (AR) and CRS are representative upper airway diseases known to be strongly related to asthma. In AR, tissue remodeling is not a representative feature, and the evidence related to remodeling is conflicting [13]. However, in CRS, tissue remodeling is a prominent feature.
In CRS, tissue remodeling has been reported to include mucosal hypertrophy, basement membrane thickening (BMT), fibrosis, collagen deposition, angiogenesis, and osteitis [14]. However, although classification based on the endotype of the disease is considered important, there is no literature review that summarizes the differences in remodeling patterns. Here, we sought to describe the recent perceptions with regards to the end of CRS and features of the upper airway remodeling according to each endotype.

2. Current Perceptions in CRS Classification

As the importance of precision medicine is emphasized in all medical fields for CRS, it is considered important to divide the spectrum of disease based on the underlying pathomechanism. Immune response is generally categorized into three inflammatory endotypes according to the inflammatory mediators, infiltrating immune cells, and physiologic functions. A distinct subset of CD4+ T cells [T helper (Th)1, Th2, and Th17 cells) plays important roles in the induction of a distinct inflammatory process according to the endotypes [15]. Based on the infiltrating cell types, CRS was initially classified as eosinophilic and non-eosinophilic CRS. Eosinophilic CRS, which is dominant in American and European patients, presents a type 2 immune response, whereas non-eosinophilic CRS, a neutrophil-dominant type prominent in Asian patients, causes either type 1 or type 3 inflammation [16]. Recent studies, including cluster analysis and multi-regional studies, further subdivided the endotypes and suggested that several endotypes could be present in one patient [17,18,19]. Nevertheless, the most fundamental classification for the endotype of CRS is type 2 or non-type 2. In this section, we summarize the pathogenesis of CRS according to either type 2 CRS or non-type 2 CRS, which leads to different clinical features and distinct upper airway remodeling patterns (Table 1) [20,21].

2.1. Type 2 Chronic Rhinosinusitis

Type 2 CRS is characterized by eosinophil-dominant inflammation. Type 2 immune response is triggered by foreign matter stimulation of the nasal epithelium following secretion of thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, IL-1, and IL-33 from epithelial cells [22,23]. TLSP stimulates and activates myeloid dendritic cells (DCs) to present antigen and co-stimulatory signals, thereby differentiating naïve T cells into effector Th2 cells; maturation subsequently occurs under an IL-4 environment, and Th2 cells secrete type 2 cytokines, including IL-4, IL-5, and IL-13, which is a hallmark of type 2 inflammation [24]. TSLP and IL-33 also enhance type 2 cytokine production by stimulating type 2 innate lymphoid cells (ILC2s) [23]. TSLP, IL-33, and IL-1 stimulate mast cells in the nasal mucosa for IL-5 and IL-13 secretion [25].
Antigen and co-stimulatory signals present on Th2 cells induce antibody production in B cells. Furthermore, IL-4 secreted from Th2 cells, dendritic cells, and ILC2s trigger antibody isotype switching to immunoglobulin E (IgE) [26]. IL-5 is known to be a key factor in the recruitment and activation of eosinophils, inducing the production of eosinophilic extracellular traps [27]. In addition, Chemokine (C-C motif) ligand (CCL) 23 produced from eosinophils via IL-5 stimulation recruit macrophages and their differentiation into M2 macrophages, which release eotaxins and CCL18 to recruit eosinophils, Th2 cells, and myeloid DCs [28,29].
The production of Staphylococcus aureus (S. aureus) enterotoxin, which acts as a super antigen either via direct interaction with MHC cells II on T cells or through binding to the TCRβ-chain, is reported to activate T cells [30]. S. aureus enterotoxin also induces the release of Th2 cytokines and enhances eosinophilic inflammation, even independently of enterotoxin activities, by inducing the secretion of IL-33 and TSLP from epithelial cells [31].

2.2. Non-Type 2 Chronic Rhinosinusitis

Non-type 2 CRS is characterized by neutrophilic inflammation in the nasal mucosa, which is triggered by infections or external irritants, such as air pollutants. Th1 cell-based type 1 inflammation and Th17 cell-based type 3 inflammation are mixed in non-type 2 CRS. External pathogen invasion induces the production of IL-6, IL-8, and tumor necrosis factor α (TNF-α) from the nasal epithelium [32]. In addition, the interaction between pathogens and Pathogen-associated molecular pattern/Toll-like receptor (PAMP/TLR) stimulates the production of interferon-γ (IFN-γ) and IL-8 [32]. These cytokines and chemokines activate DCs to differentiate CD4-positive T cells into effector T cells. IFN-γ secretion from epithelial cells after pathogen recognition induces Th1 differentiation, which is followed by the secretion of IFN-γ and IL-2 from Th1 cells, leading to type 1 inflammation [32]. Th17 and Th22 cell differentiation occurs in response to the release of IL-6 from epithelial cells, leading to type 3 inflammation. Th17 cells secrete IL-17 and IL-22, whereas Th22 cells only produce IL-22 [33]. IL-8 secreted from epithelial cells recruits neutrophils, which release inflammatory cytokines, such as IL-1β, IL-6, and IL-18, as well as myeloperoxidase (MPO) [34].
Transforming growth factor-β (TGF-β) stimulation induces regulatory T (Treg) cell differentiation from naïve T cells. Furthermore, in response to the secretion of IL-2 from Th1 cells, Treg maturation occurs [35]. Tregs are key regulators of the immune response as they produce the anti-inflammatory cytokine, IL-10, and reduce the function of Th1 and Th2 [36]. Tregs also secrete TGF-β, which is known to play an important role in tissue remodeling in non-type 2 CRS [37].

3. Tissue Remodeling

Tissue remodeling in CRS is a dynamic process that involves different temporary or permanent changes. Traditionally, differences in remodeling patterns according to the presence of nasal polyps have been reported. In CRSwNP, remodeling is characterized by pseudocyst formation, stromal edema, reduced collagen deposition, and inflammatory cell infiltration [38]. Notably, eosinophilic CRSwNP is known to be more edematous and less fibrotic than non-eosinophilic CRSwNP [20,39]. However, remodeling in CRSsNP is characterized by BMT, fibrosis, and goblet cell hyperplasia, ultimately presenting TGF-β as a critical mediator [40]. In this section, we discuss the pathogenesis of tissue remodeling in CRS and the associated inflammatory type for the induction of epithelial, mucosal, and bone remodeling.

3.1. Epithelial Remodeling

The epithelium serves as the primary barrier for physical, chemical, and immunologic stimuli, ultimately protecting the host from the external environment [41]. Respiratory epithelial cells consist of different cell types [42]. Common cell types include ciliated cells, basal cells, club cells, and mucous-secreting goblet cells, whereas rare cell types include neuroendocrine cells, ionocytes, and solitary chemosensory cells. In the airway, damaged epithelium plays a key role in driving tissue remodeling [43]. Epithelial damage has been considered an important feature owing to bronchoconstriction and ongoing inflammation in lower airway diseases, such as asthma. Epithelial damage in CRS has also been studied previously [44]. Goblet cell hyperplasia and excessive mucin production are important features of CRS. The expression of Mucin 5AC (MUC5AC) and Mucin 5B (MUC5B) were significantly upregulated in the sinus mucosa harvested from CRS patients compared to those from the control [45]. Previous studies identified that MUC5AC played an important role in airway inflammation.
Epithelial–mesenchymal transition (EMT) is defined as the transformation of a polarized epithelial cell into a mesenchymal cell phenotype, inducing loss of typical morphology of each epithelial cell, cell-to-cell junction, and polarity [46]. EMT is known to cause pathological remodeling in lower airway diseases and cancer progression. In CRS, EMT is also considered to be associated with disease recalcitrance [37,47]. During EMT, epithelial markers, such as E-cadherin and sirtuin 1 (SIRT1), are downregulated whereas mesenchymal markers, including N-cadherin, fibronectin, vimentin, alpha-smooth muscle actin (α-SMA), and matrix metalloproteinase (MMP), are upregulated [48]. In this section, we outline the epithelial remodeling in CRS and the associated inflammatory changes.
The graphical summary of epithelial remodeling in CRS is shown in Figure 1. Section 3.1.1 and Section 3.1.2 introduce the effect of inflammation on epithelial remodeling based on the type of immune response (Type 2 versus none-type 2) in CRS.

3.1.1. Type 2 Inflammation in Epithelial Remodeling

Previous studies have attempted to identify the correlation between epithelial disruption and eosinophil infiltration. Ponikau et al. reported that all histologic specimens obtained from 22 patients with refractory CRS had epithelial damage [49]. In the same study, eosinophil infiltration in CRS specimens was also identified; however, the relationship between the degree of epithelial damage and eosinophil infiltration was not assessed. The significant correlation between epithelial damage in eosinophil infiltration was first reported by Saitoh et al., suggesting that eosinophils migrating to the epithelium secrete inflammatory mediators, aggravating epithelial damage [50]. Recently, extracellular eosinophilic trap, a complex meshwork of granule protein DNA fibers produced from eosinophils, has been suggested to be associated with epithelial barrier defects in the nasal mucosa of CRS patients [51].
The effect of cytokines on epithelial integrity in CRS pathogenesis has been evaluated. Moreover, Soyka et al. reported, using air–liquid interface culture of epithelial cells from patients with CRSwNP, that stimulation of IFN-γ and IL-4 diminished epithelial barrier function, whereas IL-17 had no influence on transepithelial resistance [52]. A recent experimental study using nasal biopsies from patients with CRSwNP and primary human nasal epithelial cells reported that IL-13-matured nasal biopsies or cells showed significantly decreased expression of tight junction proteins. Furthermore, rhinovirus-induced epithelial barrier dysfunction was aggravated by IL-13 stimulation [53]. S. aureus enterotoxin B has also been identified as a driving factor for barrier disruption via IL-6 and IL-8 production in nasal epithelial cells harvested from CRSwNP patients [54].
Protease inhibitors are known to protect barrier integrity from environmental protease allergen stimulation. Compared to epithelial cells harvested from healthy patients or non-eosinophilic CRS patients, two endogenous protease inhibitors, namely Serine peptidase inhibitor Kazal Type 5 (SPINK5) and cystatin A showed decreased expression in nasal epithelial cells retrieved from eosinophilic CRS patients, thereby suggesting the role of endogenous protease inhibitor imbalance on the epithelial barrier in type 2 CRS [55,56].
Studies on the regulation of mucin genes in response to cytokines reported that IL-4, IL-13, IL-8 IL-33, and TNF increase the mRNA and protein expression levels of MUC5AC in nasal polyp-derived epithelial cells [57,58]. The increased expression of pendrin, an epithelial anion transporter, was associated with IL-13-induced mucus production in CRSwNP [59].

3.1.2. Non-Type 2 Inflammation in Epithelial Remodeling

TGF-β1 signaling is considered to be a potent stimulator of EMT, promoting the transition of epithelial cells to myofibroblasts. Accordingly, it induces tissue remodeling and the pathogenesis of CRS. TGF-β1 is known to be upregulated in CRSsNP; however, in CRSwNP, it is downregulated [37]. TGF-β1 initially binds to TGF-βR II. Thereafter, TGF-βR II recruits and phosphorylates TGF-βRI, which is followed by the phosphorylation of R-Smad, a complex of Smad2 and Smad3. Phosphorylated R-Smad binds to Smad4, forming Co-Smad, which shuttles into the nucleus to bind the target gene and regulate transcription [60].
Several epigenetic regulations have been reported to be related to TGF-β1-induced EMT. Activation of histone deacetylase (HDAC) 2 and HDAC4 enhances TGF-β1-induced EMT. Inhibitors of these genes suppress the expression of EMT markers in primary nasal epithelial cells [61]. Li et al. also reported that microRNA-21 mediates TGF-β1-induced EMT through the PTEN/Akt pathway in primary nasal epithelial cells of CRSwNP patients [62].
Wnt signaling also upregulates mesenchymal markers, such as α-SMA and vimentin, by increasing β-catenin in CRSwNP [63]. In neutrophil-dominant CRS patients, IFN-γ promotes EMT through pathways, such as the p38 and extracellular signal-regulated kinase (ERK) pathway [64]. Hypoxia was also found to induce EMT via the downregulation of PP2Ac phosphatase and upregulation of pSmad3, contributing to nasal polyposis; this hypoxia-induced EMT was suppressed by hypoxia-inducible factor (HIF)-1α inhibitors [65]. HIF-1α-induced EMT was also inhibited by the activation of SIRT1, a histone deacetylase, in sinonasal specimens harvested from CRS patients as well as in a nasal polypogenesis murine model [66]. HIF-1α was upregulated in CRSsNP and non-eosinophilic CRSwNP compared to that in healthy sinus mucosa and eosinophilic CRSwNP [49].
In nasal epithelial cells harvested from healthy sinus, the stimulation of cytokines, including TGF-β and IFN-β, and neutrophil elastase, also upregulated MUC5AC expression, whereas lipopolysaccharides and platelet activating factor downregulated MUC5AC expression. Among the TGF-β isoforms, TGF-β1 is considered to have the greatest effect on MUC5AC expression via the Smd signal transduction pathway and TGFβ-IIR binding [67]. However, TNF and IL-1β showed conflicting results in normal nasal epithelial cells. The exposed-CRS rabbit model also showed markedly increased MUC5AC expression. IL-17A was reported to induce MUC5AC expression and goblet cell hyperplasia via the act1 signaling pathway in CRSwNP-derived epithelial cells, ultimately suggesting the mechanism of mucin secretion by Th17-dominant sinus inflammation in Asian CRS patients [68]. Neutrophils also induce excessive mucin secretion through pathways, such as the TNF-, TGF-β, epidermal growth factor receptor, and α-convertase. Another study reported that pendrin enhances mucus production by inducing goblet cell hyperplasia and neutrophil infiltration in Chinese patients with CRS. Human neutrophil elastase, a serine protease released from neutrophils, is known to have an effect on goblet cell hyperplasia and excessive mucin secretion in patients with CRS [69,70].
For the epithelial disruption observed in type 1 CRS, Wang et al. reported that TLR2 was highly related to epithelial damage in CRSsNP patients [71]. In the same study, CRSsNP patients had higher expression of Toll-like receptors (TLR)2, TLR4, and TGF-β1 as well as higher collagen deposition than patients with CRSwNP, including neutrophil infiltration. Furthermore, oncostatin M (OSM) produced from neutrophils was highly increased in nasal polyps compared to that in healthy uncinate tissue. Stimulation of OSM was previously found to decrease transepithelial resistance and reduce barrier function in an air–liquid interface culture of nasal epithelial cells.

3.2. Subepithelial Remodeling

Tissue remodeling beneath the epithelium involves BMT, extracellular matrix (ECM) deposition, stromal edema, and glandular hyperplasia (Figure 2). The histology of nasal polyps also includes pseudocyst formation, edematous stroma with albumin deposition, absence of collagen production, and infiltration of inflammatory cells (eosinophils and neutrophils). Studies comparing eosinophilic and non-eosinophilic nasal polyps noted that eosinophilic nasal polyps are often associated with stromal edema and thick BM, whereas non-eosinophilic nasal polyps are related to glandular hypertrophy, fibrosis, and more pseudocysts than eosinophilic nasal polyps.
BMT is known to be the result of collagen deposition, and in CRS, type III and type V collagen are predominant [72]. The duration of CRS symptoms was found to have a positive correlation with BM thickness [73]. Studies comparing the histologies between adult and pediatric CRS patients reported that BMT was more dominant in adult CRS patients, suggesting that the duration of inflammation might be related to BMT [74,75].
ECM accumulation is also a critical process in tissue remodeling, leading to either pathologic reconstruction or normal reconstruction [76]. MMPs are the major proteolytic enzymes responsible for extracellular degradation in inflammation, tumor invasion, and chronic degenerative diseases [77]. MMPs recruit immune cells and alter cellular communication as well as immune responses via the proteolytic process of cytokines, chemokines, and complement components and their receptors. The function of MMPs is regulated by tissue inhibitors of metalloproteinases (TIMPs), and the balance between MMPs and TIMPs is considered to play a critical role in the production and degradation of individual components of the ECM, thereby affecting the pathogenesis of airway inflammation [77]. Among the 26 proteins in the MMP family, MMP-9, MMP-2, and MMP-9 have been studied comprehensively in CRS; however, conflicting results have been reported. MMP-9, which mainly degrades gelatin, aggrecan, and elastin, was considered to be associated with nasal polyp formation and TIMP-1 regulated MMP-9 activity [78].
Besides goblet cells, submucosal gland hyperplasia and hypertrophy are other sources of mucin overproduction in CRS [79]. Moreover, the submucosal glands have been regarded to play a more important role than goblet cells in mucin gene expression and mucous secretion in CRS patients [80]. In addition, mucus expulsion from the submucosal gland in CRS patients causes decreased mucous strand velocity, resulting in defective mucociliary clearance [81]. A microarray study that investigated differences in the expression of genes associated with submucosal gland hyperplasia between healthy sinus mucosa and remodeled sinus mucosa of CRS patients revealed an increase in the expression of Dexmoglein 3 (DSG3) and Parathyroid hormone like hormone (PTHLH) in serous cells and Keratin 14 (KRT14) in myoepithelial cells of CRS mucosa [82].
In Section 3.2.1 and Section 3.2.2, we describe the type 2 immune pathogenesis and the non-type 2 mechanism in the subepithelial remodeling of CRS, respectively.

3.2.1. Type 2 Inflammation in Subepithelial Remodeling

Previous studies have reported that in nasal polyps, a positive correlation exists between eosinophilic infiltration and BM thickness [50,83]. Saitho et al. identified that this correlation between BMT and eosinophils is stronger for the endothelial eosinophils than eosinophils in the subepithelial layer [50]. In addition, a recent study on the impact of histopathological factors on surgical outcomes reported that thickened BM and mucosal eosinophilia are the main factors that affect the poor prognosis of endoscopic sinus surgery [84].
In an experimental study using organ culture of nasal polyps, S. aureus infection promoted the release of MMP-2, MMP-9, TIMP-1, and Th2 cytokines, including IL-5, IL-13, and eotaxin [85]. Another study on the relationship between MMP and cytokines in nasal polyps reported a significant correlation between IL-5 and TIMP-1, suggesting their potential role in the maintenance of homeostasis in nasal polyp [86]. Watelet et al. reported that MMP-9 was increased in both CRSwNP and CRSsNP, whereas MMP-7 was only elevated in CRSwNP compared to that in the controls [87]. The same group suggested that upregulated TIMP-1 in CRSsNP might counteract the activity of MMPs, whereas no definite changes in TIMP-1 activity in CRSwNP could result in edema [88]. However, another study revealed that the increased levels of MMP-2 in nasal polyps elevated MMP-9 expression in CRSsNP, and decreased levels of TIMP-1 in both CRSsNP and CRSwNP were comparable to those in the controls [89].
Tissue edema in nasal polyps was reported to be associated with an imbalance among factors related to coagulation. The increased expression of coagulation factors and decrease in fibrinolytic activity induce fibrin accumulation in the nasal polyp; this is followed by “scaffold” formation, which traps plasma proteins to enhance edema [90]. Coagulation factor XIIIa, which is increased in type 2 nasal polyps, and the tissue plasminogen activator (t-PA) are two factors that are thought to play a role in fibrin accumulation [91]. As Th2 cytokines suppress t-PA in nasal epithelial cells, which play a role in fibrin degradation, fibrin accumulation occurs in a type 2 inflammation environment, thereby leading to edematous eosinophilic nasal polyp [92]. Sejima et al. reported upregulated expression of another fibrinolytic component, urokinase-type plasminogen activator (uPA), in CRSwNP, especially in eosinophils [93]. Their study revealed a significant correlation between uPA and ECP and suggested that excessive uPA expression might interfere with the normal TGF-β activated feedback mechanism of uPA in CRSwNP, resulting in edema in these patients.
In a recent study of mucin secretion in type 2 CRS, the IL-5 positive CRSwNP group showed upregulated expression of IL-4Rα in the submucosal gland as well as the epithelium. Further, MUC5AC and MUC5B secretion were increased following IL-4 and IL-13 stimulation [94].

3.2.2. Non-Type 2 Inflammation in Subepithelial Remodeling

In a study of ECM marker expression in Chinese CRS patients, CRSsNP showed elevated concentrations of TIMP-1, TIMP-4, FOXP3, and collagen deposition compared to those in CRSwNP. The authors claimed that this phenomenon is associated with a relative decrease in TGF-β expression and Treg function in CRSwNP compared to those in CRSsNP, resulting in a lack of collagen deposition in CRSwNP [95]. Recently, another study of CRSsNP in Asians reported that the MMP-9/TIMP-1 ratio had a positive correlation with CRSsNP severity, which is consistent with previous studies on lower respiratory tract disease. Accordingly, this finding suggests the association between MMP-9 and neutrophilic inflammation [96]. Another study from the same group reported that the expression level of MMP-9 was upregulated in non-eosinophilic CRSwNP compared to that in CRSsNP and eosinophilic CRSwNP. However, MMP-1 and MMP-3 showed higher expression levels in the nasal polyps of eosinophilic CRSwNP patients than those with non-eosinophilic nasal polyps [97]. Kostamo et al. reported that MMP-8/TIMP-1 and MMP-9/TIMP-1 ratios are increased in non-eosinophilic CRSwNP but the ratios did not significantly differ in eosinophilic CRSwNP compared to the control [76]. In a study of Chinese CRS, CRSsNP resulted in higher levels of TIMP-1 and TIMP-4 than CRSwNP. However, the levels of MMP-7 and MMP-9 were elevated in both CRSwNP and CRSsNP compared to those in controls [95].
A recent study using transcriptome analysis to assess fibroblasts from CRSsNP compared to those from healthy controls revealed an increase in the expression of the nuclear factor erythroid 2-related factor 3 (NFE2L3), which was known to be upregulated in tissues cultured with Th1 cytokines, such as TNF and IFN-γ [98].

3.3. Bone Remodeling

Osteitis, simply an inflammatory bony change, is characterized by neo-osteogenesis with new woven bone formation and periosteal thickening. Based on radiologic or pathologic findings, osteitis is present in 36–53% of CRS patients [99]. Furthermore, it is known to be associated with recalcitrant cases or disease severity [100]. The histological findings of osteitis in CRS include periosteal thickening, new woven bone formation, bone resorption, and fibrosis [101]. The underlying molecular mechanism of osteitis development is not well established. However, there have been attempts to unravel the molecular mechanism of osteitis in CRS using animal models [82]. In the CRS rabbit model, histopathological findings using the sinus bone showed periosteal fibrosis, neo-osteogenesis, and bony degradation in the disease side, affecting the contralateral side as well [102,103]. In Section 3.3.1 and Section 3.3.2, we would discuss the pathogenesis of bone remodeling in CRS based on immune response type (Figure 3).

3.3.1. Type 2 Inflammation in Bone Remodeling

Several studies have investigated the association between eosinophilic inflammation and osteitis in CRS. Snidvongs et al. reported that serum eosinophilia was associated with osteitis in patients, regardless of them having received surgery priorly [100]. Mehta et al. also proposed that a direct correlation exists between blood eosinophil levels and osteitis in relation to sinus mucosal thickening in CRS patients [104].
P-glycoprotein (P-gp), an adenosine triphosphate (ATP)-dependent efflux pump expressed on different cell types, is known to be highly expressed in eosinophilic CRS and CRSwNP and is associated with multidrug resistance [105,106]. Gunel et al. reported that a higher expression level of P-gp was found in CRS patients with osteitis than in those without osteitis [107]. In a recent microarray analysis of the genes related to osteitis in CRS, growth differentiation factor 5 and exostosin glycosyltransferase 1 were upregulated in the osteitic bone of CRS patients. Furthermore, they displayed a positive correlation with eosinophilic inflammation [108]. Fibroblast growth factor and colony stimulating factor levels were also increased in osteitic bone [108]. In a murine eosinophilic CRS model induced using Aspergillus fumigatus, significant upregulation of bone morphogenetic protein, fibroblast growth factor, and MMP family members was demonstrated [109].
The correlation between Th2 cytokines and osteitis was also previously assessed. In an in vitro study using human osteoblasts, IL-4 and Il-13 stimulation induced osteoblast differentiation activity and increased collagen secretion and mineralization [110]. In an experimental study with paranasal mucosa tissue obtained during endoscopic sinus surgery, both the mRNA and protein expression levels of IL-13 were increased in CRS patients with neo-osteogenesis [111]. In the same study, a positive correlation was found between IL-13 and both the Global Osteitis Scoring Scale (GOSS) and in vitro mineralization by osteoblast [111]. In a murine eosinophilic CRS model induced by the administration of ovalbumin and staphylococcal enterotoxin B, histological and radiographic findings of osteitis were identified, thereby demonstrating the positive correlation among bone thickness, Runt-related transcription factor 2 (RUNX2)-immunoreactive osteoblasts, and IL-13 expression [112]. A higher expression level of RUNX2 was observed in osteoblasts harvested from CRS patients with neo-osteogenesis than in those without neo-osteogenesis. Moreover, IL-13 and IL-17 were found to induce osteoblast differentiation via RUNX2 stimulation [113].

3.3.2. Non-Type 2 Inflammation in Bone Remodeling

Bacteria-induced osteitis has been reported in CRS. In fact, in experimental studies with a rabbit CRS model, bacterial organisms, including S. aureus and Pseudomonas aeruginosa, can induce bony changes, such as periosteal thickening, woven bone formation, hyperplastic osteoblasts, osteoclastic activity, and fibrosis [114,115]. A retrospective study of 90 patients undergoing sinus surgery revealed that P. aeruginosa, and not S. aureus, is an independent predictor of neo-osteogenesis in refractory CRS patients [116]. However, Karampelis et al. reported that neither P. aeruginosa nor S. aureus was associated with neo-osteogenesis in CRS patients with or without cystic fibrosis [117]. Several studies have also provided evidence of the association between bacterial biofilms and osteitis in CRS patients. In fact, a study with 84 CRS patients reported that CRS patients with higher biofilm volume or biofilm score had higher histopathologic bony grade or greater GOSS score than those with a lower biofilm volume or score [118].
Higher expression levels of inflammatory cytokines, such as IL-6, IL-11, and TNF-α, were identified more in the bone tissue obtained from CRS patients with osteitis than in those without osteitis [119]. TGF-β signaling cascades have also been reported to be involved in the bone remodeling process of both CRSsNP and CRSwNP; this is also accompanied by increased expression of TGF-β and TGF-β receptor 1 as well as a higher number of Smad2/3-positive cells [120].

4. Conclusions

Tissue remodeling is a dynamic and complex process. As a key feature of CRS, remodeling involves different layers of sinonasal tissues, including the epithelium, subepithelium, and underlying bone. Tissue remodeling in CRS exhibits distinct clinical features based on the inflammatory pattern of the disease, which is known as “endotype,” and involves different immune cells and inflammatory mediators. In CRS, remodeling is not only a simple consequential change following long periods of chronic inflammation but is also a parallel process that continuously occurs during ongoing inflammation. Remodeling in CRS is known to affect symptom severity, disease prognosis, response to treatment, and recurrence. Thus, to date, different studies have sought to determine whether it is possible to reverse the remodeled tissue in CRS. Steroids, the most widely used medication that exhibits anti-inflammatory activity, could regulate the level of MMPs and TIMPs, induce epithelial repair, and reduce tissue eosinophil infiltration [121,122]. In addition, a remarkably reduced amount of TGF-β1 positive cells and a thinner basement membrane were identified in a steroid-treated CRS group compared to the controls [123]. Doxycycline could have the potential to reduce nasal polyp volume by decreasing MMP-9 levels and reducing MUC5AC levels [124,125]. However, doxycycline did not significantly alter IL-5 levels and a slightly increased level of eotaxin-3 was identified after doxycycline treatment [124]. Clarithromycin was also demonstrated to contribute to the reduction of MMP-9 and TGF-β in an in vitro study; however, this drug did not exhibit any effect when assessed in a clinical setting [126]. Mepolizumab (an IL-5 antagonist), a recently emerging treatment option for recalcitrant type 2 CRS, was reported to reduce ECM protein deposition and decrease tissue eosinophil numbers [127,128,129]. Furthermore, TGF-β1 expression in airway eosinophils and decreased concentration of TGF-β1 in bronchoalveolar lavage fluid were also reduced by Mepolizumab treatment in asthma patients, suggesting the possible role of Mepolizumab in improving tissue remodeling in type 2 CRS [129].
In this review, we sought to discuss the tissue remodeling pattern and the accompanying immune responses that are induced by the inflammatory type of CRS. However, the underlying pathomechanism of tissue remodeling has yet to be elucidated and is, thus, being actively investigated. Efforts to more precisely classify CRS according to immune response and to elucidate the immune mechanisms are also being conducted to apply precision medicine in the management of CRS [130]. Future research should attempt to identify treatment targets to reverse tissue remodeling in CRS according to the underlying immune response.

Funding

This research was supported by the Basic Science Research Program, National Research Foundation of Korea, funded by the Ministry of Science and Technology and the Ministry of Science, ICT and Future Planning (2017R1A2B2003575, NRF-2020R1A2C1006398), the Ministry of Science and ICT (2020R1C1C1012288), Korea, under the ICT Creative Consilience program (IITP-2020–0018190011001) supervised by the IITP (Institute for Information and Communications Technology Planning and Evaluation), and the Korea Health Technology R&D Project (HI17C0387), Korea Health Industry Development Institute (KHIDI), and the Ministry of Health and Welfare. This research was also supported by a Korea University grant and a grant from Korea University Medical Center and Anam Hospital, Seoul, Republic of Korea.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

CRSChronic rhinosinusitis
ARAllergic rhinitis
BMTBasement membrane thickening
CCLChemokine (C-C motif) ligand
CRSsNPChronic rhinosinusitis without nasal polyp
CRSwNPChronic rhinosinusitis with nasal polyp
CXCLChemokine (C-X-C motif) ligand
ECMExtracellular matrix
EMTEpithelial-mesenchymal transition
ERKExtracellular signal-regulated kinase
HDACHistone deacetylase
HIF-1αHypoxia-inducible factor -1α
IFN-γInterferon-γ
IgEImmunoglobulin E
ILInterleukin
ILC1sType 1 innate lymphoid cells
ILC2sType 2 Innate lymphoid cells
ILC3sType 3 innate lymphoid cells
MMPMatrix metalloproteinase
MPOMyeloperoxidase
MUC5ACMucin 5AC
MUC5BMucin 5B
OSMOncostatin
P-gpP-glycoprotein
RUNX2Runt-related transcription factor 2
S.aureusStaphylococcus aureus
SIRT1Sirtuin 1
TGF-βTransforming growth factor-β
Th1Type 1 T helper
Th17Type 17 T helper
Th2Type 2 T helper
TIMPTissue inhibitors of metalloproteinases
TNF-αTumor necrosis factor α
tPATissue plasminogen activator
TregsRegulatory T cells
TSLPThymic stromal lymphopoietin
uPAUrokinase-type plasminogen activator
α-SMASmooth muscle actin

References

  1. Fokkens, W.J.; Lund, V.J.; Hopkins, C.; Hellings, P.W.; Kern, R.; Reitsma, S.; Toppila-Salmi, S.; Bernal-Sprekelsen, M.; Mullol, J.; Alobid, I.; et al. European Position Paper on Rhinosinusitis and Nasal Polyps 2020. Rhinology 2020, 58, 1–464. [Google Scholar] [CrossRef]
  2. McCormick, J.P.; Thompson, H.M.; Cho, D.-Y.; Woodworth, B.A.; Grayson, J.W. Phenotypes in Chronic Rhinosinusitis. Curr. Allergy Asthma Rep. 2020, 20, 1–8. [Google Scholar] [CrossRef] [PubMed]
  3. Meltzer, E.O.; Hamilos, D.L.; Hadley, J.A.; Lanza, D.C.; Marple, B.F.; Nicklas, R.A.; Bachert, C.; Baraniuk, J.; Baroody, F.M.; Benninger, M.S.; et al. Otolaryngology-Head and Neck Surgery. Otolaryngol. Neck Surg. 2004, 131, 1–62. [Google Scholar] [CrossRef]
  4. Akdis, C.A.; Bachert, C.; Cingi, C.; Dykewicz, M.S.; Hellings, P.W.; Naclerio, R.M.; Schleimer, R.P.; Ledford, D.K. 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. [Google Scholar] [CrossRef] [Green Version]
  5. Bachert, C.; Akdis, C.A. Phenotypes and Emerging Endotypes of Chronic Rhinosinusitis. J. Allergy Clin. Immunol. Pr. 2016, 4, 621–628. [Google Scholar] [CrossRef]
  6. Van Zele, T.; Claeys, S.; Gevaert, P.; Van Maele, G.; Holtappels, G.; Van Cauwenberge, P.; Bachert, C. Differentiation of chronic sinus diseases by measurement of inflammatory mediators. Allergy 2006, 61, 1280–1289. [Google Scholar] [CrossRef]
  7. Brescia, G.; Zanotti, C.; Parrino, D.; Barion, U.; Marioni, G. Nasal polyposis pathophysiology: Endotype and phenotype open issues. Am. J. Otolaryngol. 2018, 39, 441–444. [Google Scholar] [CrossRef]
  8. Cao, P.-P.; Li, H.-B.; Wang, B.-F.; Wang, S.-B.; You, X.-J.; Cui, Y.-H.; Wang, D.-Y.; Desrosiers, M.; Liu, Z. Distinct immunopathologic characteristics of various types of chronic rhinosinusitis in adult Chinese. J. Allergy Clin. Immunol. 2009, 124, 478–484.e2. [Google Scholar] [CrossRef]
  9. Cho, S.-W.; Kim, D.W.; Kim, J.-W.; Lee, C.H.; Rhee, C.-S. Classification of chronic rhinosinusitis according to a nasal polyp and tissue eosinophilia: Limitation of current classification system for Asian population. Asia Pac. Allergy 2017, 7, 121–130. [Google Scholar] [CrossRef] [Green Version]
  10. Delemarre, T.; Holtappels, G.; De Ruyck, N.; Zhang, N.; Nauwynck, H.; Bachert, C.; Gevaert, E. Type 2 inflammation in chronic rhinosinusitis without nasal polyps: Another relevant endotype. J. Allergy Clin. Immunol. 2020, 146, 337–343. [Google Scholar] [CrossRef]
  11. Beers, M.F.; Morrisey, E.E. The three R’s of lung health and disease: Repair, remodeling, and regeneration. J. Clin. Investig. 2011, 121, 2065–2073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Hough, K.P.; Curtiss, M.L.; Blain, T.J.; Liu, R.-M.; Trevor, J.; Deshane, J.S.; Thannickal, V.J. Airway Remodeling in Asthma. Front. Med. 2020, 7, 191. [Google Scholar] [CrossRef] [PubMed]
  13. Samitas, K.; Carter, A.; Kariyawasam, H.H.; Xanthou, G. Upper and lower airway remodelling mechanisms in asthma, allergic rhinitis and chronic rhinosinusitis: The one airway concept revisited. Allergy 2017, 73, 993–1002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Bassiouni, A.; Chen, P.G.; Wormald, P.-J. Mucosal remodeling and reversibility in chronic rhinosinusitis. Curr. Opin. Allergy Clin. Immunol. 2013, 13, 4–12. [Google Scholar] [CrossRef]
  15. Husain, Q.; Sedaghat, A. Understanding and clinical relevance of chronic rhinosinusitis endotypes. Clin. Otolaryngol. 2019, 44, 887–897. [Google Scholar] [CrossRef]
  16. Wang, X.; Zhang, N.; Bo, M.; Holtappels, G.; Zheng, M.; Lou, H.; Wang, H.; Zhang, L.; Bachert, C. Diversity of T H cytokine profiles in patients with chronic rhinosinusitis: A multicenter study in Europe, Asia, and Oceania. J. Allergy Clin. Immunol. 2016, 138, 1344–1353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Tan, B.K.; Klingler, A.I.; Poposki, J.A.; Stevens, W.W.; Peters, A.T.; Suh, L.A.; Norton, J.; Carter, R.G.; Hulse, K.E.; Harris, K.E.; et al. Heterogeneous inflammatory patterns in chronic rhinosinusitis without nasal polyps in Chicago, Illinois. J. Allergy Clin. Immunol. 2017, 139, 699–703. [Google Scholar] [CrossRef] [Green Version]
  18. Tomassen, P.; Vandeplas, G.; Van Zele, T.; Cardell, L.O.; Arebro, J.; Olze, H.; Förster-Ruhrmann, U.; Kowalski, M.L.; Olszewska-Ziąber, A.; Holtappels, G.; et al. Inflammatory endotypes of chronic rhinosinusitis based on cluster analysis of biomarkers. J. Allergy Clin. Immunol. 2016, 137, 1449–1456. [Google Scholar] [CrossRef] [Green Version]
  19. Turner, J.H.; Chandra, R.K.; Li, P.; Bonnet, K.; Schlundt, D.G. Identification of clinically relevant chronic rhinosinusitis endotypes using cluster analysis of mucus cytokines. J. Allergy Clin. Immunol. 2018, 141, 1895–1897.e7. [Google Scholar] [CrossRef] [Green Version]
  20. Schleimer, R.P. Immunopathogenesis of Chronic Rhinosinusitis and Nasal Polyposis. Annu. Rev. Pathol. Mech. Dis. 2017, 12, 331–357. [Google Scholar] [CrossRef] [Green Version]
  21. Stevens, W.W.; Peters, A.T.; Tan, B.K.; Klingler, A.I.; Poposki, J.A.; Hulse, K.E.; Grammer, L.C.; Welch, K.C.; Smith, S.S.; Conley, D.B.; et al. Associations Between Inflammatory Endotypes and Clinical Presentations in Chronic Rhinosinusitis. J. Allergy Clin. Immunol. Pr. 2019, 7, 2812–2820. [Google Scholar] [CrossRef]
  22. Boita, M.; Bucca, C.; Riva, G.; Heffler, E.; Rolla, G. Release of Type 2 Cytokines by Epithelial Cells of Nasal Polyps. J. Immunol. Res. 2016, 2016, 1–7. [Google Scholar] [CrossRef]
  23. Licona-Limon, P.; Kim, L.K.; Palm, N.W.; Flavell, R.A. TH2, allergy and group 2 innate lymphoid cells. Nat. Immunol. 2013, 14, 536–542. [Google Scholar] [CrossRef]
  24. Omori, M.; Ziegler, S. Induction of IL-4 expression in CD4(+) T cells by thymic stromal lymphopoietin. J. Immunol. 2007, 178, 1396–1404. [Google Scholar] [CrossRef] [Green Version]
  25. Saluja, R.; Zoltowska, A.; Ketelaar, M.E.; Nilsson, G. IL-33 and Thymic Stromal Lymphopoietin in mast cell functions. Eur. J. Pharmacol. 2016, 778, 68–76. [Google Scholar] [CrossRef]
  26. Punnonen, J.; Yssel, H.; Devries, J. The relative contribution of IL-4 and IL-13 to human IgE synthesis induced by activated CD4 or CD8 T cells. J. Allergy Clin. Immunol. 1997, 100, 792–801. [Google Scholar] [CrossRef]
  27. Hamilos, D.L.; Leung, D.Y.; Wood, R.; Cunningham, L.; Bean, D.K.; Yasruel, Z.; Schotman, E.; Hamid, Q. Evidence for distinct cytokine expression in allergic versus nonallergic chronic sinusitis. J. Allergy Clin. Immunol. 1995, 96, 537–544. [Google Scholar] [CrossRef]
  28. E Peterson, S.; Poposki, J.A.; Nagarkar, D.R.; Chustz, R.T.; Peters, A.T.; Suh, L.A.; Carter, R.; Norton, J.; Harris, K.E.; Grammer, L.C.; et al. Increased expression of CC chemokine ligand 18 in patients with chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 2012, 129, 119–127. [Google Scholar] [CrossRef] [Green Version]
  29. Poposki, J.A.; Uzzaman, A.; Nagarkar, D.R.; Chustz, R.T.; Peters, A.T.; Suh, L.A.; Carter, R.; Norton, J.; Harris, K.E.; Grammer, L.C.; et al. Increased expression of the chemokine CCL23 in eosinophilic chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 2011, 128, 73–81. [Google Scholar] [CrossRef] [Green Version]
  30. Vickery, T.W.; Ramakrishnan, V.R.; Suh, J.D. The Role of Staphylococcus aureus in Patients with Chronic Sinusitis and Nasal Polyposis. Curr. Allergy Asthma Rep. 2019, 19, 21. [Google Scholar] [CrossRef] [PubMed]
  31. Lan, F.; Zhang, N.; Holtappels, G.; De Ruyck, N.; Krysko, O.; Van Crombruggen, K.; Braun, H.; Johnston, S.L.; Papadopoulos, N.G.; Zhang, L.; et al. Staphylococcus aureusInduces a Mucosal Type 2 Immune Response via Epithelial Cell–derived Cytokines. Am. J. Respir. Crit. Care Med. 2018, 198, 452–463. [Google Scholar] [CrossRef] [PubMed]
  32. Hirschberg, A.; Kiss, M.; Kadocsa, E.; Polyanka, H.; Szabo, K.; Razga, Z.; Bella, Z.; Tiszlavicz, L.; Kemeny, L. Different activations of toll-like receptors and antimicrobial peptides in chronic rhinosinusitis with or without nasal polyposis. Eur. Arch. Oto-Rhino-Laryngol. 2016, 273, 1779–1788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Jia, L.; Wu, C. The Biology and Functions of Th22 Cells. Cannabinoids Neuropsychiatr. Disord. 2014, 841, 209–230. [Google Scholar] [CrossRef]
  34. Bachert, C.; Van Kempen, M.J.P.K.; Höpken, K.; Holtappels, G.; Wagenmann, M. Elevated levels of myeloperoxidase, pro-inflammatory cytokines and chemokines in naturally acquired upper respiratory tract infections. Eur. Arch. Oto-Rhino-Laryngol. 2001, 258, 406–412. [Google Scholar] [CrossRef] [PubMed]
  35. Georgiev, P.; Charbonnier, L.-M.; Chatila, T.A. Regulatory T Cells: The Many Faces of Foxp3. J. Clin. Immunol. 2019, 39, 623–640. [Google Scholar] [CrossRef]
  36. Palmer, C.; Mulligan, J.K.; Smith, S.E.; Atkinson, C. The Role of Regulatory T Cells in the Regulation of Upper Airway Inflammation. Am. J. Rhinol. Allergy 2017, 31, 345–351. [Google Scholar] [CrossRef] [Green Version]
  37. Van Bruaene, N.; DeRycke, L.; Perez-Novo, C.A.; Gevaert, P.; Holtappels, G.; De Ruyck, N.; Cuvelier, C.; Van Cauwenberge, P.; Bachert, C. TGF-β signaling and collagen deposition in chronic rhinosinusitis. J. Allergy Clin. Immunol. 2009, 124, 253–259. [Google Scholar] [CrossRef]
  38. Stevens, W.W.; Lee, R.J.; Schleimer, R.P.; Cohen, N.A. Chronic rhinosinusitis pathogenesis. J. Allergy Clin. Immunol. 2015, 136, 1442–1453. [Google Scholar] [CrossRef] [Green Version]
  39. Shi, L.-L.; Xiong, P.; Zhang, L.; Cao, P.-P.; Liao, B.; Lu, X.; Cui, Y.-H.; Liu, Z. Features of airway remodeling in different types of Chinese chronic rhinosinusitis are associated with inflammation patterns. Allergy 2012, 68, 101–109. [Google Scholar] [CrossRef]
  40. Kountakis, S.E.; Arango, P.; Bradley, D.; Wade, Z.K.; Borish, L. Molecular and Cellular Staging for the Severity of Chronic Rhinosinusitis. Laryngoscope 2004, 114, 1895–1905. [Google Scholar] [CrossRef]
  41. Hellings, P.W.; Steelant, B. Epithelial barriers in allergy and asthma. J. Allergy Clin. Immunol. 2020, 145, 1499–1509. [Google Scholar] [CrossRef] [PubMed]
  42. Bankova, L.G.; Barrett, N.A. Epithelial cell function and remodeling in nasal polyposis. Ann. Allergy Asthma Immunol. 2020, 124, 333–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Davies, D.E. The Role of the Epithelium in Airway Remodeling in Asthma. Proc. Am. Thorac. Soc. 2009, 6, 678–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Khalmuratova, R.; Park, J.-W.; Shin, H.-W. Immune Cell Responses and Mucosal Barrier Disruptions in Chronic Rhinosinusitis. Immune Netw. 2017, 17, 60–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kim, D.H.; Chu, H.-S.; Lee, J.Y.; Hwang, S.J.; Lee, S.H.; Lee, H.-M. Up-regulation of MUC5AC and MUC5B Mucin Genes in Chronic Rhinosinusitis. Arch. Otolaryngol. Head Neck Surg. 2004, 130, 747–752. [Google Scholar] [CrossRef] [Green Version]
  46. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
  47. Hackett, T.-L.; Warner, S.M.; Stefanowicz, D.; Shaheen, F.; Pechkovsky, D.V.; Murray, L.A.; Argentieri, R.; Kicic, A.; Stick, S.M.; Bai, T.R.; et al. Induction of Epithelial–Mesenchymal Transition in Primary Airway Epithelial Cells from Patients with Asthma by Transforming Growth Factor-β1. Am. J. Respir. Crit. Care Med. 2009, 180, 122–133. [Google Scholar] [CrossRef]
  48. Thiery, J.P.; Acloque, H.; Huang, R.Y.J.; Nieto, M.A. Epithelial-Mesenchymal Transitions in Development and Disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
  49. Ponikau, J.U.; A Sherris, D.; Kephart, G.M.; Kern, E.B.; A Gaffey, T.; E Tarara, J.; Kita, H. Features of airway remodeling and eosinophilic inflammation in chronic rhinosinusitis. J. Allergy Clin. Immunol. 2003, 112, 877–882. [Google Scholar] [CrossRef]
  50. Saitoh, T.; Kusunoli, T.; Yao, T.; Kawano, K.; Kojima, Y.; Miyahara, K.; Onoda, J.; Yokoi, H.; Ikeda, K. Relationship between epithelial damage or basement membrane thickness and eosinophilic infiltration in nasal polyps with chronic rhinosinusitis. Rhinol. J. 2009, 47, 275–279. [Google Scholar] [CrossRef] [Green Version]
  51. Hwang, C.S.; Park, S.C.; Cho, H.-J.; Park, D.-J.; Yoon, J.-H.; Kim, C.-H. Eosinophil extracellular trap formation is closely associated with disease severity in chronic rhinosinusitis regardless of nasal polyp status. Sci. Rep. 2019, 9, 8061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Soyka, M.B.; Wawrzyniak, P.; Eiwegger, T.; Holzmann, D.; Treis, A.; Wanke, K.; Kast, J.I.; Akdis, C.A. Defective epithelial barrier in chronic rhinosinusitis: The regulation of tight junctions by IFN-γ and IL-4. J. Allergy Clin. Immunol. 2012, 130, 1087–1096. [Google Scholar] [CrossRef] [PubMed]
  53. Huang, Z.-Q.; Liu, J.; Ong, H.H.; Yuan, T.; Zhou, X.-M.; Wang, J.; Tan, K.S.; Chow, V.T.; Yang, Q.-T.; Shi, L.; et al. Interleukin-13 Alters Tight Junction Proteins Expression Thereby Compromising Barrier Function and Dampens Rhinovirus Induced Immune Responses in Nasal Epithelium. Front. Cell Dev. Biol. 2020, 8, 572749. [Google Scholar] [CrossRef] [PubMed]
  54. Martens, K.; Seys, S.F.; Alpizar, Y.A.; Schrijvers, R.; Bullens, D.M.; Breynaert, C.; Lebeer, S.; Steelant, B. Staphylococcus aureus enterotoxin B disrupts nasal epithelial barrier integrity. Clin. Exp. Allergy 2021, 51, 87–98. [Google Scholar] [CrossRef] [PubMed]
  55. Wu, D.; Wei, Y.; Bleier, B.S. Emerging Role of Proteases in the Pathogenesis of Chronic Rhinosinusitis with Nasal Polyps. Front. Cell. Infect. Microbiol. 2018, 7, 538. [Google Scholar] [CrossRef] [Green Version]
  56. Kouzaki, H.; Matsumoto, K.; Kato, T.; Tojima, I.; Shimizu, S.; Shimizu, T. Epithelial Cell-Derived Cytokines Contribute to the Pathophysiology of Eosinophilic Chronic Rhinosinusitis. J. Interf. Cytokine Res. 2016, 36, 169–179. [Google Scholar] [CrossRef]
  57. Jiao, J.; Duan, S.; Meng, N.; Li, Y.; Fan, E.; Zhang, L. Role of IFN-γ, IL-13, and IL-17 on mucociliary differentiation of nasal epithelial cells in chronic rhinosinusitis with nasal polyps. Clin. Exp. Allergy 2016, 46, 449–460. [Google Scholar] [CrossRef]
  58. Ishinaga, H.; Kitano, M.; Toda, M.; D’Alessandro-Gabazza, C.N.; Gabazza, E.; Shah, S.A.; Takeuchi, K. Interleukin-33 induces mucin gene expression and goblet cell hyperplasia in human nasal epithelial cells. Cytokine 2017, 90, 60–65. [Google Scholar] [CrossRef]
  59. Seshadri, S.; Lu, X.; Purkey, M.R.; Homma, T.; Choi, A.W.; Carter, R.; Suh, L.; Norton, J.; Harris, K.E.; Conley, D.B.; et al. Increased expression of the epithelial anion transporter pendrin/SLC26A4 in nasal polyps of patients with chronic rhinosinusitis. J. Allergy Clin. Immunol. 2015, 136, 1548–1558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Weiss, A.; Attisano, L. The TGFbeta Superfamily Signaling Pathway. Wiley Interdiscip. Rev. Dev. Boil. 2013, 2, 47–63. [Google Scholar] [CrossRef]
  61. Park, I.-H.; Kang, J.-H.; Shin, J.-M.; Lee, H.-M. Trichostatin A Inhibits Epithelial Mesenchymal Transition Induced by TGF-β1 in Airway Epithelium. PLoS ONE 2016, 11, e0162058. [Google Scholar] [CrossRef] [PubMed]
  62. Li, X.; Li, C.; Zhu, G.; Yuan, W.; Xiao, Z.-A. TGF-β1 Induces Epithelial-Mesenchymal Transition of Chronic Sinusitis with Nasal Polyps through MicroRNA-21. Int. Arch. Allergy Immunol. 2019, 179, 304–319. [Google Scholar] [CrossRef] [PubMed]
  63. Ms, A.D.; Khalil, S.M.; Lane, A.P. Nasal polyp fibroblasts modulate epithelial characteristics via Wnt signaling. Int. Forum Allergy Rhinol. 2018, 8, 1412–1420. [Google Scholar] [CrossRef]
  64. Lee, M.; Kim, D.W.; Khalmuratova, R.; Shin, S.-H.; Kim, Y.-M.; Han, D.H.; Kim, H.-J.; Kim, D.-Y.; Rhee, C.-S.; Park, J.-W.; et al. The IFN-γ–p38, ERK kinase axis exacerbates neutrophilic chronic rhinosinusitis by inducing the epithelial-to-mesenchymal transition. Mucosal Immunol. 2019, 12, 601–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Shin, H.-W.; Cho, K.; Kim, D.W.; Han, D.H.; Khalmuratova, R.; Kim, S.-W.; Jeon, S.-Y.; Min, Y.-G.; Lee, C.H.; Rhee, C.-S.; et al. Hypoxia-inducible Factor 1 Mediates Nasal Polypogenesis by Inducing Epithelial-to-Mesenchymal Transition. Am. J. Respir. Crit. Care Med. 2012, 185, 944–954. [Google Scholar] [CrossRef]
  66. Lee, M.; Kim, D.W.; Yoon, H.; So, D.; Khalmuratova, R.; Rhee, C.-S.; Park, J.-W.; Shin, H.-W. Sirtuin 1 attenuates nasal polypogenesis by suppressing epithelial-to-mesenchymal transition. J. Allergy Clin. Immunol. 2016, 137, 87–98. [Google Scholar] [CrossRef]
  67. Milara, J.; Morell, A.; Ballester, B.; Armengot, M.; Morcillo, E.; Cortijo, J. MUC4 impairs the anti-inflammatory effects of corticosteroids in patients with chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 2017, 139, 855–862. [Google Scholar] [CrossRef] [Green Version]
  68. Xia, W.; Bai, J.; Wu, X.; Wei, Y.; Feng, S.; Li, L.; Zhang, J.; Xiong, G.; Fan, Y.; Shi, J.; et al. Interleukin-17A Promotes MUC5AC Expression and Goblet Cell Hyperplasia in Nasal Polyps via the Act1-Mediated Pathway. PLoS ONE 2014, 9, e98915. [Google Scholar] [CrossRef]
  69. Yan, D.; Ye, Y.; Zhang, J.; Zhao, J.; Yu, J.; Luo, Q. Human Neutrophil Elastase Induces MUC5AC Overexpression in Chronic Rhinosinusitis Through miR-146a. Am. J. Rhinol. Allergy 2020, 34, 59–69. [Google Scholar] [CrossRef]
  70. Luo, Q.; Zhang, Z.; Liu, D.; Feng, K.; Jin, X.; Zhang, J. Human neutrophil elastase induces MUC5AC overexpression in chronic rhinosinusitis through tumour necrosis factor-α converting enzyme. Acta Oto-Laryngol. 2016, 136, 641–648. [Google Scholar] [CrossRef]
  71. Wang, X.; Zhao, C.; Ji, W.; Xu, Y.; Guo, H. Relationship of TLR2, TLR4 and tissue remodeling in chronic rhinosinusitis. Int. J. Clin. Exp. Pathol. 2015, 8, 1199–1212. [Google Scholar] [PubMed]
  72. Molet, S.M.; Hamid, Q.A.; Hamilos, D.L. IL-11 and IL-17 expression in nasal polyps: Relationship to collagen deposition and suppression by intranasal fluticasone propionate. Laryngoscope 2003, 113, 1803–1812. [Google Scholar] [CrossRef] [PubMed]
  73. Rehl, R.M.; Balla, A.A.; Cabay, R.J.; Hearp, M.L.; Pytynia, K.B.; Joe, S.A. Mucosal Remodeling in Chronic Rhinosinusitis. Am. J. Rhinol. 2007, 21, 651–657. [Google Scholar] [CrossRef] [PubMed]
  74. Sobol, S.E.; Fukakusa, M.; Christodoulopoulos, P.; Manoukian, J.J.; Schloss, M.D.; Frenkiel, S.; Hamid, Q. Inflammation and Remodeling of the Sinus Mucosa in Children and Adults with Chronic Sinusitis. Laryngoscope 2003, 113, 410–414. [Google Scholar] [CrossRef] [PubMed]
  75. Zang, H.-R.; Wang, T.; Li, Y.; Li, P.; Zhang, S.-Z.; Fan, E.-Z.; Li, Y. A histopathological study: Chronic rhinosinusitis in adolescents versus adults. Zhonghua Yi Xue Za Zhi 2009, 89, 1975–1978. [Google Scholar] [PubMed]
  76. Kostamo, K.; Tervahartiala, T.; Sorsa, T.; Richardson, M.; Toskala, E. Metalloproteinase Function in Chronic Rhinosinusitis with Nasal Polyposis. Laryngoscope 2007, 117, 638–643. [Google Scholar] [CrossRef] [PubMed]
  77. Shapiro, S.D.; Senior, R.M. Matrix Metalloproteinases. Am. J. Respir. Cell Mol. Biol. 1999, 20, 1100–1102. [Google Scholar] [CrossRef] [PubMed]
  78. Kahveci, O.K.; Derekoy, F.S.; Yilmaz, M.; Serteser, M.; Altuntas, A. The role of MMP-9 and TIMP-1 in nasal polyp formation. Swiss Med. Wkly. 2008, 138, 684–688. [Google Scholar]
  79. Tos, M.; Mogensen, C. Mucus Production in Chronic Maxillary Sinusitis: A Quantitative Histopathological Study. Acta Oto-Laryngol. 1984, 97, 151–159. [Google Scholar] [CrossRef]
  80. Ali, M.S.; Wilson, J.A.; Bennett, M.; Pearson, J.P. Mucin gene expression in nasal polyps. Acta Oto-Laryngol. 2005, 125, 618–624. [Google Scholar] [CrossRef]
  81. Tipirneni, K.E.; Zhang, S.; Cho, D.-Y.; Grayson, J.; Bs, D.F.S.; Mackey, C.; Moore, L.; Cole, D.; Banks, C.G.; Woodworth, B.A. Submucosal gland mucus strand velocity is decreased in chronic rhinosinusitis. Int. Forum Allergy Rhinol. 2018, 8, 509–512. [Google Scholar] [CrossRef] [PubMed]
  82. Perloff, J.R.; Gannon, F.H.; Bolger, W.E.; Montone, K.T.; Orlandi, R.; Kennedy, D.W. Bone Involvement in Sinusitis: An Apparent Pathway for the Spread of Disease. Laryngoscope 2000, 110, 2095–2099. [Google Scholar] [CrossRef] [PubMed]
  83. Nonaka, M.; Pawankar, R.; Saji, F.; Yagi, T. Distinct expression of RANTES and GM-CSF by lipopolysaccharide in human nasal fibroblasts but not in other airway fibroblasts. Int. Arch. Allergy Immunol. 1999, 119, 314–321. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, F.; Yang, Y.; Wu, Q.; Chen, H. Histopathologic analysis in chronic rhinosinusitis: Impact on quality of life outcomes. Am. J. Otolaryngol. 2019, 40, 423–426. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, J.H.; Kwon, H.J.; Jang, Y.J. Staphylococcus Aureus Increases Cytokine and Matrix Metalloproteinase Expression in Nasal Mucosae of Patients with Chronic Rhinosinusitis and Nasal Polyps. Am. J. Rhinol. Allergy 2010, 24, 422–427. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, Y.-S.; Langhammer, T.; Westhofen, M.; Lorenzen, J. Relationship between matrix metalloproteinases MMP-2, MMP-9, tissue inhibitor of matrix metalloproteinases-1 and IL-5, IL-8 in nasal polyps. Allergy 2007, 62, 66–72. [Google Scholar] [CrossRef]
  87. Watelet, J.B.; Claeys, C.; Perez-Novo, C.; Gevaert, P.; Van Cauwenberge, P.; Bachert, C. Transforming growth factor beta1 in nasal remodeling: Differences between chronic rhinosinusitis and nasal polyposis. Am. J. Rhinol. 2004, 18, 267–272. [Google Scholar] [CrossRef]
  88. Watelet, J.B.; Bachert, C.; Claeys, C.; Van Cauwenberge, P. Matrix metalloproteinases MMP-7, MMP-9 and their tissue inhibitor TIMP-1: Expression in chronic sinusitis vs nasal polyposis. Allergy 2004, 59, 54–60. [Google Scholar] [CrossRef]
  89. Can, I.H.; Ceylan, K.; Caydere, M.; Samim, E.E.; Ustun, H.; Karasoy, D.S. The expression of MMP-2, MMP-7, MMP-9, and TIMP-1 in chronic rhinosinusitis and nasal polyposis. Otolaryngol. Neck Surg. 2008, 139, 211–215. [Google Scholar] [CrossRef]
  90. Kim, D.-Y.; Cho, S.H.; Takabayashi, T.; Schleimer, R.P. Chronic Rhinosinusitis and the Coagulation System. Allergy Asthma Immunol. Res. 2015, 7, 421–430. [Google Scholar] [CrossRef] [Green Version]
  91. Takabayashi, T.; Kato, A.; Peters, A.T.; Hulse, K.E.; Suh, L.A.; Carter, R.; Norton, J.; Grammer, L.C.; Tan, B.K.; Chandra, R.K.; et al. Increased expression of factor XIII-A in patients with chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 2013, 132, 584–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Takabayashi, T.; Kato, A.; Peters, A.T.; Hulse, K.E.; Suh, L.A.; Carter, R.; Norton, J.; Grammer, L.C.; Cho, S.H.; Tan, B.K.; et al. Excessive Fibrin Deposition in Nasal Polyps Caused by Fibrinolytic Impairment through Reduction of Tissue Plasminogen Activator Expression. Am. J. Respir. Crit. Care Med. 2013, 187, 49–57. [Google Scholar] [CrossRef] [Green Version]
  93. Sejima, T.; Holtappels, G.; Bachert, C. The Expression of Fibrinolytic Components in Chronic Paranasal Sinus Disease. Am. J. Rhinol. Allergy 2011, 25, 1–6. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, Y.; DeRycke, L.; Holtappels, G.; Wang, X.D.; Zhang, L.; Bachert, C.; Zhang, N. Th2 cytokines orchestrate the secretion of MUC 5 AC and MUC 5B in IL -5-positive chronic rhinosinusitis with nasal polyps. Allergy 2019, 74, 131–140. [Google Scholar] [CrossRef]
  95. Li, X.; Meng, J.; Qiao, X.; Liu, Y.; Liu, F.; Zhang, N.; Zhang, J.; Holtappels, G.; Luo, B.; Zhou, P.; et al. Expression of TGF, matrix metalloproteinases, and tissue inhibitors in Chinese chronic rhinosinusitis. J. Allergy Clin. Immunol. 2010, 125, 1061–1068. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, D.W.; Eun, K.M.; Roh, E.Y.; Shin, S.; Park, C.H. Chronic Rhinosinusitis without Nasal Polyps in Asian Patients Shows Mixed Inflammatory Patterns and Neutrophil-Related Disease Severity. Mediat. Inflamm. 2019, 2019, 1–9. [Google Scholar] [CrossRef] [PubMed]
  97. Park, C.H.; Eun, K.M.; Kim, M.-K.; Cho, D.; A Han, S.; Han, S.-Y.; Seo, Y.; Lee, D.-H.; Cho, S.-H.; Kim, D.W. Comparison Between Signature Cytokines of Nasal Tissues in Subtypes of Chronic Rhinosinusitis. Allergy Asthma Immunol. Res. 2019, 11, 201–211. [Google Scholar] [CrossRef]
  98. Ball, S.L.; Cockell, S.J.; Wilson, J.A.; Mann, D.A.; Fisher, A.J. Microarray analysis of primary epithelial and fibroblast cells in chronic rhinosinusitis without nasal polyps. Rhinology 2020, 58, 581–587. [Google Scholar] [CrossRef]
  99. Snidvongs, K.; Sacks, R.; Harvey, R.J. Osteitis in Chronic Rhinosinusitis. Curr. Allergy Asthma Rep. 2019, 19, 24. [Google Scholar] [CrossRef]
  100. Snidvongs, K.; McLachlan, R.; Chin, D.; Pratt, E.; Sacks, R.; Earls, P.; Harvey, R.J. Osteitic bone: A surrogate marker of eosinophilia in chronic rhinosinusitis. Rhinol. J. 2012, 50, 299–305. [Google Scholar] [CrossRef] [Green Version]
  101. Cho, S.H.; Min, H.J.; Han, H.X.; Paik, S.S.; Kim, K.R. CT analysis and histopathology of bone remodeling in patients with chronic rhinosinusitis. Otolaryngol. Neck Surg. 2006, 135, 404–408. [Google Scholar] [CrossRef] [PubMed]
  102. De Campos, C.A.C.; Dolci, E.L.L.; Da Silva, L.; Dolci, J.E.L.; De Campos, C.A.H.; Dolci, R.L.L. Osteitis and mucosal inflammation in a rabbit model of sinusitis. Braz. J. Otorhinolaryngol. 2015, 81, 312–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Norlander, T.; Westrin, K.M.; Stierna, P. The Inflammatory Response of the Sinus and Nasal Mucosa During Sinusitis: Implications for Research and Therapy. Acta Oto-Laryngol. 1994, 114, 38–44. [Google Scholar] [CrossRef] [PubMed]
  104. Mehta, V.; Campeau, N.; Kita, H.; Hagan, J.B. Blood and Sputum Eosinophil Levels in Asthma and Their Relationship to Sinus Computed Tomographic Findings. Mayo Clin. Proc. 2008, 83, 671–678. [Google Scholar] [CrossRef]
  105. Feldman, R.E.; Lam, A.C.; Sadow, P.M.; Bleier, B.S. P-glycoprotein is a marker of tissue eosinophilia and radiographic inflammation in chronic rhinosinusitis without nasal polyps. Int. Forum Allergy Rhinol. 2013, 3, 684–687. [Google Scholar] [CrossRef] [PubMed]
  106. Miyake, M.M.; Nocera, A.; Miyake, M.M. P-glycoprotein and chronic rhinosinusitis. World J. Otorhinolaryngol. Head Neck Surg. 2018, 4, 169–174. [Google Scholar] [CrossRef]
  107. Günel, C.; Feldman, R.E.; Bleier, B.S. Osteitis is Associated with P-Glycoprotein Overexpression in Patients with Chronic Sinusitis without Nasal Polyps. Am. J. Rhinol. Allergy 2014, 28, 99–102. [Google Scholar] [CrossRef]
  108. Günel, C.; Bleier, B.S.; Bozkurt, G.; Eliyatkin, N. Microarray analysis of the genes associated with osteitis in chronic rhinosinusitis. Laryngoscope 2017, 127, E85–E90. [Google Scholar] [CrossRef]
  109. Sautter, N.B.; Bs, K.L.D.; A Hausman, F.; Trune, D.R.; Ba, F.A.H. Tissue remodeling gene expression in a murine model of chronic rhinosinusitis. Laryngoscope 2012, 122, 711–717. [Google Scholar] [CrossRef] [Green Version]
  110. Silfverswärd, C.; Penno, H.; Frost, A.; Nilsson, O.; Ljunggren, O. Expression of markers of activity in cultured human osteoblasts: Effects of interleukin-4 and interleukin-13. Scand. J. Clin. Lab. Investig. 2010, 70, 338–342. [Google Scholar] [CrossRef]
  111. Oue, S.; Ramezanpour, M.; Paramasivan, S.; Miljkovic, D.; Cooksley, C.M.; Bassiouni, A.; Ou, J.; Psaltis, A.J.; Wormald, P.-J.; Vreugde, S. Increased IL-13 expression is independently associated with neo-osteogenesis in patients with chronic rhinosinusitis. J. Allergy Clin. Immunol. 2017, 140, 1444–1448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Khalmuratova, R.; Lee, M.; Park, J.-W.; Shin, H.-W. Evaluation of Neo-Osteogenesis in Eosinophilic Chronic Rhinosinusitis Using a Nasal Polyp Murine Model. Allergy, Asthma Immunol. Res. 2020, 12, 306–321. [Google Scholar] [CrossRef] [PubMed]
  113. Khalmuratova, R.; Shin, H.-W.; Kim, D.W.; Park, J.-W. Interleukin (IL)-13 and IL-17A contribute to neo-osteogenesis in chronic rhinosinusitis by inducing RUNX2. EBioMedicine 2019, 46, 330–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Khalid, A.N.; Hunt, J.; Perloff, J.R.; Kennedy, D.W. The Role of Bone in Chronic Rhinosinusitis. Laryngoscope 2002, 112, 1951–1957. [Google Scholar] [CrossRef]
  115. Dong, Y.; Zhou, B.; Wang, X.; Huang, Z.; Wang, M.; Li, Y.; Zang, H.; Fan, E.; Li, Y.; Wang, X. Computed tomography and histopathological evaluation of osteitis in rabbit models with rhinosinusitis. Acta Oto-Laryngol. 2016, 137, 534–540. [Google Scholar] [CrossRef]
  116. Huang, Z.; Hajjij, A.; Li, G.; Nayak, J.V.; Zhou, B.; Hwang, P.H. Clinical predictors of neo-osteogenesis in patients with chronic rhinosinusitis. Int. Forum Allergy Rhinol. 2015, 5, 303–309. [Google Scholar] [CrossRef]
  117. Karempelis, P.; Karp, E.; Rubin, N.; Hunter, R.; Dunitz, J.; Boyer, H. Risk factors for neo-osteogenesis in cystic fibrosis and non‒cystic fibrosis chronic rhinosinusitis. Int. Forum Allergy Rhinol. 2020, 10, 505–510. [Google Scholar] [CrossRef]
  118. Dong, D.; Zhao, Y.; Xiao, W.; HongYan, Z.; Jia, L.; Yan, X.; Jia, W. Correlation between bacterial biofilms and osteitis in patients with chronic rhinosinusitis. Laryngoscope 2013, 124, 1071–1077. [Google Scholar] [CrossRef]
  119. Tuszynska, A.; Krzeski, A.; Postuba, M.; Paczek, L.; Wyczalkowska-Tomasik, A.; Gornicka, B.; Pykalo, R. Inflammatory cytokines gene expression in bone tissue from patients with chronic rhinosinusitis- a preliminary study. Rhinol. J. 2010, 48, 415–419. [Google Scholar]
  120. Wang, M.; Ye, T.; Liang, N.; Huang, Z.; Cui, S.; Li, Y.; Huang, Q.; Zhou, B. Differing Roles for TGF-β/Smad Signaling in Osteitis in Chronic Rhinosinusitis with and without Nasal Polyps. Am. J. Rhinol. Allergy 2015, 29, e152–e159. [Google Scholar] [CrossRef]
  121. Callejas-Díaz, B.; Martínez-Antón, A.; Picado, C.; Alobid, I.; Pujols, L.; Valero, A.; Roca-Ferrer, J.; Mullol, J. Corticosteroid treatment regulates mucosal remodeling in chronic rhinosinusitis with nasal polyps. Laryngoscope 2015, 125, E158–E167. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, C.; Lou, H.; Wang, X.; Wang, Y.; Fan, E.; Li, Y.; Wang, H.; Bachert, C.; Zhang, L. Effect of budesonide transnasal nebulization in patients with eosinophilic chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. 2015, 135, 922–929.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Yu, R.-L.; Zhu, D.-D.; Dong, Z. [Effects of glucocorticoid on tissue remodeling of nasal mucosa of chronic rhinosinusitis with nasal polyposis after endoscopic surgery]. Chin. J. Otorhinolaryngol. Head Neck Surg. 2006, 41, 773–776. [Google Scholar]
  124. Sommer, J.U.; Schultz, J.D.; Grossbaier, J.; Stern-Straeter, J.; Hörmann, K.; Sauter, A. In vitro effects of doxycycline on inflammatory cytokines and gelatinases in chronic rhinosinusitis. In Vivo 2012, 26, 369–374. [Google Scholar]
  125. Huvenne, W.; Zhang, N.; Tijsma, E.; Hissong, B.; Huurdeman, J.; Holtappels, G.; Claeys, S.; Van Cauwenberge, P.; Nelis, H.; Coenye, T.; et al. Pilot study using doxycycline-releasing stents to ameliorate postoperative healing quality after sinus surgery. Wound Repair Regen. 2008, 16, 757–767. [Google Scholar] [CrossRef] [PubMed]
  126. Sassa, K.; Mizushima, Y.; Kobayashi, M. Differential Modulatory Effects of Clarithromycin on the Production of Cytokines by a Tumor. Antimicrob. Agents Chemother. 1999, 43, 2787–2789. [Google Scholar] [CrossRef] [Green Version]
  127. Numata, T.; Nakayama, K.; Utsumi, H.; Kobayashi, K.; Yanagisawa, H.; Hashimoto, M.; Minagawa, S.; Ishikawa, T.; Hara, H.; Araya, J.; et al. Efficacy of mepolizumab for patients with severe asthma and eosinophilic chronic rhinosinusitis. BMC Pulm. Med. 2019, 19, 1–9. [Google Scholar] [CrossRef] [Green Version]
  128. Nagase, H.; Ueki, S.; Fujieda, S. The roles of IL-5 and anti-IL-5 treatment in eosinophilic diseases: Asthma, eosinophilic granulomatosis with polyangiitis, and eosinophilic chronic rhinosinusitis. Allergol. Int. 2020, 69, 178–186. [Google Scholar] [CrossRef]
  129. Flood-Page, P.; Menzies-Gow, A.; Phipps, S.; Ying, S.; Wangoo, A.; Ludwig, M.S.; Barnes, N.; Robinson, D.; Kay, A.B. Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J. Clin. Investig. 2003, 112, 1029–1036. [Google Scholar] [CrossRef] [Green Version]
  130. Bauer, A.M.; Turner, J.H. Personalized Medicine in Chronic Rhinosinusitis. Immunol. Allergy Clin. N. Am. 2020, 40, 281–293. [Google Scholar] [CrossRef]
Figure 1. Pathophysiology of epithelial remodeling (epithelial barrier disruption, goblet cell hyperplasia, and epithelial–mesenchymal transition) in chronic rhinosinusitis according to the type of inflammation (type 2 versus non-type 2) (figure created using Biorender.com).
Figure 1. Pathophysiology of epithelial remodeling (epithelial barrier disruption, goblet cell hyperplasia, and epithelial–mesenchymal transition) in chronic rhinosinusitis according to the type of inflammation (type 2 versus non-type 2) (figure created using Biorender.com).
Ijms 22 00910 g001
Figure 2. Subepithelial remodeling (extracellular matrix deposition, fibrin deposition, basement membrane thickening, and submucosal hypertrophy) according to the type of inflammation in chronic rhinosinusitis.
Figure 2. Subepithelial remodeling (extracellular matrix deposition, fibrin deposition, basement membrane thickening, and submucosal hypertrophy) according to the type of inflammation in chronic rhinosinusitis.
Ijms 22 00910 g002
Figure 3. Bone remodeling based on the type of inflammation (type 2 inflammation versus non-type 2 inflammation) in chronic rhinosinusitis (figure created using Biorender.com).
Figure 3. Bone remodeling based on the type of inflammation (type 2 inflammation versus non-type 2 inflammation) in chronic rhinosinusitis (figure created using Biorender.com).
Ijms 22 00910 g003
Table 1. Differences between Type 2 CRS and Non-type 2 CRS.
Table 1. Differences between Type 2 CRS and Non-type 2 CRS.
Type 2Non-Type 2
External stimuliAllergen, Staphylococcus aureus biofilm/enterotoxin, and FungiPollution and bacteria
Effector cellsEosinophils,
Th2 cells, ILC2s, B cells, basophils, and mast cells
Neutrophils, NK cells, Type 1: CD8+ T cells, Th1 cells, and ILC1s
Type 3: Th17 cells and ILC3s
Primary cytokinesIL-4, IL-5, and IL-13Type 1: IFN-γ and IL-12
Type 3: IL-17 and IL-22
Other mediatorsIL-25, IL-33, TSLP, and IgEIL-1β, IL-36, IL-6, IL-8, TGF-β, CXCL1, and CXCL10
Clinical featuresBilateral disease
Ethmoid sinus > Maxillary sinus
Headache/migraine, anosmia, nasal polyposis, asthma, aspirin-induced respiratory disease
Maxillary sinus > Ethmoid sinus
Purulent nasal discharge
Tissue remodeling featuresBarrier disruption, basal membrane thickening, and stromal edemaBasal membrane thickening, goblet cell hyperplasia, fibrosis, and collagen deposition
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, K.; Tai, J.; Lee, S.H.; Kim, T.H. 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. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020910

AMA Style

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. International Journal of Molecular Sciences. 2021; 22(2):910. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020910

Chicago/Turabian Style

Lee, Kijeong, Junhu Tai, Sang Hag Lee, and Tae Hoon Kim. 2021. "Advances in the Knowledge of the Underlying Airway Remodeling Mechanisms in Chronic Rhinosinusitis Based on the Endotypes: A Review" International Journal of Molecular Sciences 22, no. 2: 910. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020910

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop