Next Article in Journal
The Advances in Epigenetics for Cancer Radiotherapy
Next Article in Special Issue
Deep Association between Transglutaminase 1 and Tissue Eosinophil Infiltration Leading to Nasal Polyp Formation and/or Maintenance with Fibrin Polymerization in Chronic Rhinosinusitis with Nasal Polyps
Previous Article in Journal
TNF-α Induces Mitophagy in Rheumatoid Arthritis Synovial Fibroblasts, and Mitophagy Inhibition Alleviates Synovitis in Collagen Antibody-Induced Arthritis
Previous Article in Special Issue
Therapeutic Strategies of Biologics in Chronic Rhinosinusitis: Current Options and Future Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Distinct Gene Set Enrichment Profiles in Eosinophilic and Non-Eosinophilic Chronic Rhinosinusitis with Nasal Polyps by Bulk RNA Barcoding and Sequencing

Department of Otorhinolaryngology, Head and Neck Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Kasumi 1-2-3, Hiroshima 734-8551, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(10), 5653; https://doi.org/10.3390/ijms23105653
Submission received: 26 April 2022 / Revised: 14 May 2022 / Accepted: 16 May 2022 / Published: 18 May 2022
(This article belongs to the Special Issue Chronic Rhinosinusitis: Aetiology, Immunology and Treatment)

Abstract

:
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a chronic inflammatory disease with a high symptom burden, including nasal congestion and smell disorders. This study performed a detailed transcriptomic analysis in CRSwNP classified as eosinophilic CRS (ECRS), nonECRS according to the Japanese Epidemiological Survey of Refractory Eosinophilic Chronic Rhinosinusitis (JESREC) criteria, and a group of ECRS with comorbid aspirin intolerant asthma (Asp). Gene expression profiles of nasal polyps and the uncinate process in CRSwNP patients and normal subjects (controls) were generated by bulk RNA barcoding and sequencing (BRB-seq). A differentially expressed genes (DEGs) analysis was performed using DESeq2 software in iDEP to clarify any relationship between gene expression and disease backgrounds. A total of 3004 genes were identified by DEGs analysis to be associated with ECRS vs control, nonECRS vs control, and Asp vs control. A pathway analysis showed distinct profiles between the groups. A Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) showed distinct phenotype-specific pathways of expressed genes. In the specific pathway of “cytokine–cytokine receptor interaction”, the differentially expressed genes were widely distributed. This study indicates that transcriptome analysis using BRB-seq may be a valuable tool to explore the pathogenesis of type 2 inflammation in CRSwNP.

1. Introduction

Chronic rhinosinusitis (CRS) is a chronic inflammatory upper airway disease with a high symptom burden, including nasal congestion, smell disorders, nasal discharge, and facial pain/pressure [1,2]. Based on the presence or absence of nasal polyps, CRS can be classified as without nasal polyps (CRSsNP) or with nasal polyps (CRSwNP), which have distinct inflammatory profiles and pathogenesis. CRS can be subclassified into further categories by its underlying cellular and molecular pathophysiologic mechanisms [3], and CRSwNP can also be subclassified into ECRS and nonECRS in terms of the degree of eosinophilic infiltration of sinonasal tissue [4]. Compared with nonECRS, which is supposed to be characterized by Th1-dominant inflammation, ECRS is a phenotype of CRSwNP characterized by Th2-dominant inflammation, which is difficult-to-treat and leads to poorer surgical outcomes [4,5]. Additionally, ECRS patients with comorbid aspirin intolerant asthma (AIA) are thought to have more difficult-to-treat CRS due to more severe type 2 inflammation [4,6]. Therefore, the type of inflammation in CRSwNP is thought to be a key factor in classifying CRSwNP as a difficult-to-treat CRS. Currently, JESREC criteria are used to classify ECRS into mild, moderate, and severe ECRS [4]. From a transcriptome analysis of nasal polyps, it was certified that nasal polyps can be segregated into two major endotypes showing a type 2 or non-type 2 endotype [7]. However, a previous study using a transcriptome analysis with three ECRS, three nonECRS, and three control subjects showed no distinct differentiation of expressed mRNAs between ECRS and nonECRS [8]. Given the discrepancy between the inflammatory differences of CRSwNP and differentiation of expressed mRNAs in transcriptome analysis, we presume that the severity of eosinophilic infiltration does not enhance the severity of type 2 inflammation or pathological endotype. This study performed a detailed transcriptomic analysis among ECRS and nonECRS based on the JESREC criteria and a group of ECRS with comorbid AIA (Asp) to reveal whether the diagnosis of ECRS and/or Asp correlates with the severity of type 2 inflammation or pathological endotype. Because RNA-seq, a method for transcriptomic analysis, is still too laborious and expensive, we used bulk RNA barcoding and sequencing (BRB-seq), which is a comparatively novel method for RNA sequencing and uses early multiplexing to produce 3′ cDNA libraries for dozens of samples [9]. BRB-seq has a comparable performance to the standard TruSeq approach while showing greater tolerance for lower RNA quality and being up to 25 times cheaper [9]. In this study, we applied unsupervised clustering methods from comprehensive RNA gene expression data using BRB-seq and analyzed the difference between these three groups of CRSwNP to gain insights into the molecular profiles and endotype categorization of CRSwNP.

2. Results

2.1. Gene Expression Profiles of NPs between ECRS, NonECRS, and Asp

PCA, Heatmap of the Correlation Matrix, and Hierarchical Cluster Analysis

PCA was performed among all groups and revealed that the control group was clearly segregated from the other groups, but ECRS, nonECRS, and Asp were not segregated from each other (Figure 1A). A heatmap of the correlation matrix also clarified the difficulty of the segregation among the three CRSwNP groups (Figure 1B).
Hierarchical cluster analysis with a cutoff Z score of 4 revealed no distinct segregation among the three CRSwNP groups, and some ECRS (ECRS1) and nonECRS (nonECRS5 and 7) patients were clustered with the control groups (Figure 2). DEGs were identified as both upregulated and downregulated genes in ECRS vs control (ECRS-Ctrl) of 677 upregulated and 479 downregulated, nonECRS vs control (nonECRS-Ctrl) of 695 and 310, Asp vs control (Asp-Ctrl) of 380 and 463, Asp vs nonECRS (Asp-nonECRS) of 3 and 16, and Asp vs ECRS (Asp-ECRS) of 1 and 9. However, no DEGs were identified in ECRS vs nonECRS (ECRS-nonECRS) (Figure 3).
However, a Venn diagram of all CRSwNP groups vs control showed the existence of specific DEGs in each CRSwNP group (Figure 4).
Pathway analysis with the GO molecular function with DEGs in ECRS-Ctrl, nonECRS-Ctrl, and Asp-Ctrl showed the existence of significant up and downregulated pathways (Figure 5A–C), but Asp-nonECRS and Asp-ECRS showed no significant up and downregulated pathways.
To clarify the detailed differences in the pathology of ECRS-Ctrl, nonECRS-Ctrl, and Asp-Ctrl in the common KEGG pathways, we analyzed the upregulated DEGs using DAVID. Genes with disease-associated terms such as “cytokine–cytokine receptor interaction”, “hematopoietic cell lineage”, “chemokine signaling pathway”, “complement and coagulation cascade”, “osteoclast differentiation”, “Staphylococcus aureus infection”, and “viral protein interaction with cytokine and cytokine receptor” were commonly upregulated in all CRSwNP groups vs control (Table 1).
The term “cytokine–cytokine receptor interaction” was commonly high-ranked in terms of the gene count in all CRSwNP groups vs control. DEGs in “Cytokine–Cytokine Receptor Interaction” showed that CCL13, CCL18, CCL26, TNFRSF18, INHBB, IL1RL1, and IL2RA were more upregulated in ECRS-Ctrl than nonECRS-Ctrl; CCL2, CCL8, CCL20, CCR5, CXCL1, CXCL6, CXCR2, FAS, TNFRSF1B, TNFSF13B, CSF3, IL2RG, IL6R, IL20RB, and IL23A were more upregulated in nonECRS-Ctrl than ECRS-Ctrl (Table 2). The upregulated genes in Asp-Ctrl were similar to the DEGs in ECRS-Ctrl and nonECRS-Ctrl. (Table 2).
The KEGG pathway analysis with the downregulated DEGs of ECRS-Ctrl, nonECRS-Ctrl, and Asp-Ctrl also revealed that genes associated with the terms “metabolic pathways”, “salivary secretion”, “mucin type O-glycan biosynthesis”, and “glycine, serine and threonine metabolism” were commonly downregulated (Table 3). The gene count of “metabolic pathways” showed the highest count among the all CRSwNP groups vs Ctrl, and the gene counts were widely distributed as ECRS-Ctrl of 55, nonECRS-Ctrl of 36, and Asp-Ctrl of 65.

2.2. Segregation All CRSwNPs into Two Types of Clusters by Hierarchic Cluster Analysis

Due to the close association and similar clusters between ECRS, nonECRS, and Asp, we segregated these CRSwNPs into two clusters (cluster1 and 2) using the hierarchical cluster analysis with all CRSwNPs (Figure 6). The two clusters were not correlated with the comorbid of asthma. Additionally, height, weight, BMI, sex, age, comorbid of olfactory disturbance, otitis media, smoking, blood eosinophil, total IgE, nasal polyp score, CT score, tissue eosinophil, JESREC score, Oral FeNO, and Nasal FeNO were not correlated with the segregation (data not shown).
The new clusters and control showed clear separation in the PCA analysis and DEGs (Figure 7).
The cluster-specific upregulated and downregulated DEGs (Table S1) were identified between cluster1 vs cluster2, and the KEGG pathway analysis by DAVID with upregulated DEGs revealed that the disease-associated terms “cytokine–cytokine receptor interaction” was commonly highly ranked between cluster1 vs control (cluster1-Ctrl) and cluster2 vs control (cluster2-Ctrl) (Table S2).
Compared to cluster1-Ctrl and cluster2-Ctrl, cluster2 was more upregulated in genes of CC subfamily, CXC subfamily, gamma-chain utilizing, IL-4-like, IL6/12-like, IL-1-like cytokine, and TNF families associated with “cytokine–cytokine receptor interaction” (Figure 8).

3. Discussion

CRS has been categorized using the pathological molecular evidence and syndromic phenotypes, and currently it is believed that strong pathological associations between endotype and phenotypes would be the key factors to resolve the clinical problems of CRS therapies [3]. ECRS involving Asp is known as the most difficult-to-treat type of CRSwNP because of the easy relapse of NPs after sinus surgery and resistance to medical therapy except for oral systemic corticosteroids; nonECRS is known as an easy-to-treat type of CRSwNP with good response to ESS and ordinal macrolide therapy [10]. The pathological differences between ECRS and nonECRS were recognized by the type of inflammation: whether type 2-dependent with eosinophil infiltration or type 1-dependent.
Currently, the criteria for categorizing ECRS have been readily accepted, especially in terms of the disease severity [11,12,13]. To detect clear differences in the type of inflammation and other factors, we performed the transcriptome analysis for ECRS, nonECRS, and Asp. The PCA and heatmap of the correlation matrix showed clear segregation between the control group and the others, but the others were not segregated. Additionally, hierarchical cluster analysis of the DEGs revealed no distinct segregation among the three CRSwNP groups, and some ECRS and nonECRS patients were segregated in a similar cluster as a control group. These results suggested that CRSwNP was widely distributed, but endotype separation using the current criteria for ECRS, which mainly use a count of eosinophil infiltration in NP and blood eosinophil count or positive of comorbid asthma and/or AIA, could not well-segregate CRSwNP into the pathological endotypes. Furthermore, these results also suggested that new criteria for the segregation of CRSwNPs to enhance the pathological endotypes should be proposed to analyze the effectiveness of various therapies.
In our study, a difference in significant enrichment pathways among the CRSwNP groups could not be detected using the DEGs because of no or a low number of DEGs. However, a difference in category-oriented significant enrichment pathways, which came from the comparison between each CRSwNP groups vs the control, could be identified, and “cytokine–cytokine receptor interaction” was mainly upregulated. Additionally, the DEGs associated with “cytokine–cytokine receptor interaction” among the three comparison groups were widely distributed, and the upregulated genes of CCL13 and CCL26 (eotaxin-3) in ECRS and Asp and CCL18 in ECRS were identified in either ECRS or Asp compared with nonECRS. In addition, CCL11(eotaxin) was upregulated in ECRS and nonECRS, and CCL24 (eotaxin-2) was not identified in any CRSwNPs. Previous reports mentioned that type 2 inflammatory transcript expression of CCL13, CCL18, and CCL26 were upregulated in the type 2 endotype [7] and ECRS compared with nonECRS [8]. It is known that CCL11, CCL13, and CCL26 recruit eosinophils, basophils, mast cells, and CD4(+) T helper 2 cells [14], and CCL18 recruits naive T cells, B cells, immature dendritic cells, and activate fibroblasts [15]. CCL18 is thought to be involved in T(H)2-related inflammatory diseases, including asthma and atopic dermatitis, and the number of CCL18(+) cells was significantly increased within NPs [15]. Production of CCL26 is induced by IL4/13 in epithelial cells [16], and CCL26 induces not only eosinophil infiltration but also NK cell migration, which contributes to trafficking NK cells to the allergic upper airway mediated by CX3CL1 and the tyrosine kinase pathway [17]. Higher expression of type 2 inflammatory molecules (CCL13, IGHE, CCL18, CCL23, CCR3, and CLC) along with lower levels of LACRT, PPDPFL, DES, C6, MUC5B, and SCGB3A1 are related to a stronger clinical response to glucocorticoids [18]. In terms of the above-mentioned evidence, these differences would explain the difference of each phenotype as to whether they mainly have type 2 inflammation or not. From the upregulation of these gene expressions, we concluded that ECRS have higher type 2 inflammation with the upregulation of CCL13, CCL18, and CCL26 than nonECRS, and eosinophil infiltration in ECRS would be mainly induced by CCL13 and CCL26 rather than CCL11 and CCL24.
Furthermore, the upregulated expression of TNFRSF18, IL1RL1, and IL2RA in ECRS and Asp, INHBB in ECRS, and TNFRSF10D, IL6, IL11, and IL31RA in Asp is also associated with “cytokine–cytokine receptor interaction”. IL1RL1 is the receptor of IL33, which induces T helper 2-type inflammatory responses. IL1RL1-positive cells in the inflammatory cells of the subepithelial layer are significantly higher, and the expression of IL1RL1 in polyps is significantly increased in the ECRS compared with controls. Many IL1RA1-positive eosinophils are observed only in the mucosa of ECRS but not of nonECRS [19]. Therefore, overexpression of IL1RA1 in ECRS and Asp suggested that higher type 2 inflammation would be induced by IL33 and IL1RA1 with CD4(+) T helper 2 cells recruited by CCL11, CCL13, and CCL26. Furthermore, TNFRSF10D is expressed by macrophages/monocytes and promotes eosinophil survival via inhibiting the spontaneous apoptosis [20]. IL11 is selectively expressed in eosinophils and epithelial cells in patients with moderate and severe asthma, and correlates directly with disease severity. IL11 also causes nodular mononuclear infiltrates [21]. IL31 and IL31RA induce MUC5AC gene expression in human airway epithelial cells and play important roles in mucus overproduction [22]. These findings suggested that eosinophil accumulation caused by prolonged survival of eosinophil with TNFRSF10D, nodular mononuclear infiltration with IL11, and mucus overexpression with MUC5AC via IL31RA would be the characteristic features of Asp. In the same way as IL11 enhances the disease severity in asthma, overexpression of IL6 and IL11 in Asp may reflect the severity of IL6-mediated inflammation and type 2 inflammation enhancing the disease severity in CRS.
Overexpressions of TNFRSF18, IL2RA, and INHBB in either ECRS or Asp groups may also reflect the disease severity and pathology of the CRSwNP phenotype via interference of cytokine–cytokine receptor interaction despite poor reports of pathological analyses between the overexpressions of these genes and the phenotype of ECRS and/or Asp.
However, the KEGG pathway analysis for the downregulated DEGs of ECRS-Ctrl, nonECRS-Ctrl, and Asp-Ctrl also revealed that “metabolic pathways” would be another factor for the CRSwNP phenotype. The gene count differences of “metabolic pathways”, namely ECRS-Ctrl of 55, nonECRS-Ctrl of 36, and Asp-Ctrl of 65, also pave the way to elucidate the differences in each phenotype associated with disease severity.
Hierarchical cluster analysis using the DEGs revealed no distinct segregation among the three CRSwNP groups, but it did suggest that these CRSwNPs segregate into two clusters in terms of the similarity of up and downregulated DEGs. Two clusters (cluster 1 and 2) among all CRSwNPs, after modifying a hierarchical cluster analysis, showed clear separation in the PCA analysis. The cluster-specific upregulated DEGs and the KEGG pathway analysis by DAVID of two clusters and the control group revealed that the disease-associated term “cytokine–cytokine receptor interaction” was commonly highly ranked. Compared to cluster1-Ctrl and cluster2-Ctrl in the DEGs of “cytokine–cytokine receptor interaction”, cluster2 was more upregulated in the genes of the CC subfamily and CXC subfamily in “chemokines”, gamma-chain utilizing, and IL-4 like and IL-6/12-like genes in “the class I helical cytokines”, “IL1-like cytokines”, and “TNF family”. In the IL4-like genes in “the class I helical cytokines”, a comparison between cluster1-Ctrl and cluster2-Ctrl revealed that cluster2 was more upregulated in the expression of CSF2RB and IL4R than cluster1. These genes are deeply associated with type 2 inflammation via IL4, IL5, and IL13 signaling [23,24]. Previously, dupilumab, which is a dual inhibitor of IL4 and IL13 signaling and controls the type 2 inflammation, was shown to decrease several symptoms of CRSwNP despite the diversity in the degree of eosinophilic infiltration of sinonasal tissue by classifying CRSwNP into the categories from nonECRS to severe ECRS [25]. Given the result of the effectiveness of dupilumab against all CRSwNP, all CRSwNP groups should have type 2 inflammation, which could differ in degree regardless of the criteria for ECRS. The upregulated expression of CSF2RB and IL4R in cluster2 is expected to be strongly associated with the effect of dupilumab by blocking this signaling. In addition, the segregation of DEGs into cluster1 and cluster2 will elucidate the broad effectiveness of dupilumab in all CRSwNPs in spite of the phenotypes of nonECRS, which are suggested to be deeply associated with type 1 inflammation. The segregation of DEGs into cluster1 and 2 will become the criteria for true endotype segregation, and further studies are needed to categorize CRSwNP enhancement by/of IL4, IL5, and IL13 signaling.
There are some limitations to our study. First, due to the small sample size in Asp, we could not demonstrate more clear differences in DEGs between ECRS and nonECRS, and the tendency as to whether Asp could be classified as more cluster1 or 2. Second, this study only included Japanese patients; hence, caution should be taken when extrapolating our results to other ethnic groups. Third, we could not measure expression protein levels. A previous report mentioned that the protein expression levels of CCL13 and CCL26 were minimal or unchanged compared with the control despite the overexpression of these mRNAs [26]. Further studies are needed to analyze the correlation among the CRS phenotype, protein expression levels, and transcriptional overexpression of CCL13 and CCL26. Additionally, some protein expression levels may also be minimal or unchanged despite transcriptomic overexpression. Further studies are necessary to explore the correlation between the DEGs detected in BRB-seq and protein expression levels.

4. Materials and Methods

4.1. Patient Recruitment

Patients with or without CRS, who underwent endoscopic sinus surgery in Hiroshima Medical University Hospital between October 2016 and October 2021, were enrolled in this study. The diagnosis of CRS was based on computed tomography (CT) scanning, patient history, clinical symptoms, and endoscopic findings. The inclusion criteria for CRSwNP were as follows: treatment without oral/nasal steroids within 4 weeks before surgery, no improvement in continuous nasal drip, post-nasal drip, and nasal congestion after medical treatment including low-dose macrolide therapy. Patients with CRSwNP were clinically diagnosed as ECRS or nonECRS based on the diagnostic criteria of the JESREC study [4]. The diagnostic criteria for ECRS (JESREC score) include (1) side affected: both sides, 3 points; (2) with nasal polyps, 2 points; (3) CT changes: ethmoid/maxillary ≥ 1, 2 points; (4) peripheral blood eosinophil count (%): 2 < and ≤ 5%, 4 points; 5 < and ≤ 10%, 8 points; 10%<, 10 points. Eosinophil infiltration into the NPs was diagnosed by calculating the mean cell count of the 3 most dense areas of eosinophils in the hematoxylin and eosin (HE) stained sections under ×400 magnification. A diagnosis of ECRS was made by both the total JESREC score of 11 points or higher and eosinophil infiltration into the NPs of 70 or more [4]. The group of ECRS with comorbid aspirin intolerant asthma was independently classified as Asp (N = 3), and the others were classified as ECRS (N = 9) and nonECRS (N = 8). Control patients (Ctrl) (N = 6) were also diagnosed based on the anatomical abnormalities but showed no inflammatory mucosal change or bacterial infection around the uncinate process. The exclusion criteria for CRSwNP were as follows: treatment with oral/nasal steroids within 4 weeks before surgery, fungal/allergic fungal rhinosinusitis, and primary ciliary dyskinesia. Demographics and clinical characteristics were obtained from the medical records. The detailed patient demography is shown in Table S3.

4.2. RNA-Seq Using BRB-Seq

BRB-seq [9] was performed for library preparation with the following modifications. Barcoded oligo-dT-based primer (5′-GCCGGTAATACGACTCACTATAGGGAGTTCTACAGTCCGACGATCNNNNNNNNNNCCCCCCCCCTTTTTTTTTTTTTTTTTTTTTTTTV -3′; 10bp “N” = UMI, 9bp “C” = cell barcode) was used for single-stranded synthesis and Second Strand Synthesis Module (New England Biolabs, Ipswich, MA, USA, #E6111) was used for double-stranded cDNA synthesis. In-house MEDS-B Tn5 transposase [27,28] was used for tagmentation, and libraries were amplified for 10 cycles of PCR using Phusion High-Fidelity DNA Polymerase (Thermo Scientific, Waltham, MA, USA, #M0530) with the following primers (5′ -AATGATACGGCGACCACCGAGATCTACACindexGTTCAGAGTTCTACAGTCCGA-3′,5′-CAAGCAGAAGACGGCATACGAGATindexGTCTCGTGGGCTCGGAGATGT-3′). An Illumina NovaSeq6000 (Illumina, San Diego, CA, USA) was used to obtain 15 bp of barcode read (Read1) and 81 bp of insert read (Read2).

4.3. Data Processing of BRB-Seq

Read1 (barcode read) was extracted by using UMI-tools (ver.1.1.1) with the command “umi_tools extract -I read1.fastq --read2-in=read2.fastq --bc-pattern=NNNNNNNNNNCCCCCCCCC --read2-stdout”. Adaptor sequences and low-quality sequences were removed and read length below 20 bp were discarded using Trim Galore (ver.0.6.7). Reads were mapped to the GRCh38 reference using HISAT2 (ver.2.2.1). Read counts for each gene were obtained by feature Counts (ver.2.0.1), and UMI duplication was removed by UMI-tools with the command “umi_tools count --method=unique --per-gene --per-cell --gene-tag=XT”. Normalized read count value was obtained by using DESeq2 (ver.1.34.0) (Table S4). Gene-level expression data (read counts) were processed by using the web portal for integrated differential expression and pathway analysis (iDEP.95; http://bioinformatics.sdstate.edu/idep, accessed on 25 April 2022). iDEP.95 was used for principal component analysis (PCA), hierarchical clustering with a heatmap, identification of differentially expressed genes (DEGs), and pathway analysis. DEGs were extracted with an FDR cutoff of 0.1 and min-fold change of 2. Pathway analysis was performed using GAGE with the genesets of GO Molecular Function. Geneset size was set at minimum 5 and maximum 2000, and pathway significance cutoff (FDR) was set at 0.2. To ascertain the functions of the differential expressed mRNAs, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was used with the Database for Annotation, Visualization, and Integrated Discovery (DAVID; https://david.ncifcrf.gov/, accessed on 25 April 2022).

5. Conclusions

This study revealed and shed light on the future gene expression targets for endotype categorization. Additionally, this study also revealed that BRB-seq was useful to elucidate the differences in CRSwNP. BRB-seq allowed us to perform a transcriptome analysis for as many as 26 patients because of the low cost of the sequencing. A greater number of patients could help us to segregate into new clusters and elucidate the differences between them. This method may provide a valuable tool for further studies to analyze the differences in CRSwNPs in terms of the phenotype and endotype categorization.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23105653/s1.

Author Contributions

Conceptualization, T.I.; methodology, T.I.; software, T.I.; validation, T.I., N.C., and T.K.; formal analysis, T.T.; investigation, S.T.; resources, Y.H.; data curation, K.T., M.N., T.O., K.Y., and T.H.; writing—original draft preparation, T.I.; writing—review and editing, S.T.; visualization, T.I.; supervision, S.T.; project administration, T.U.; funding acquisition, S.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded partly by the Japan Society for the Promotion of Science KAKENHI grant: 22K09668, the Health Labor Sciences Research grant: 21FC1013, and Research Collaboration Fund with Universal Sound Design Inc.: 0G20KA7383.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and was approved by the Ethics Committee of Hiroshima University Hospital, Hiroshima University, Hiroshima, Japan (Hi-136; August.11.2014 as date of approval).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge Y.O. for the technical support.

Conflicts of Interest

The authors declare no conflict of interest.

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] [PubMed]
  2. Orlandi, R.R.; Kingdom, T.T.; Smith, T.L.; Bleier, B.; DeConde, A.; Luong, A.U.; Poetker, D.M.; Soler, Z.; Welch, K.C.; Wise, S.K.; et al. International consensus statement on allergy and rhinology: Rhinosinusitis 2021. Int. Forum Allergy Rhinol. 2021, 11, 213–739. [Google Scholar] [CrossRef] [PubMed]
  3. Grayson, J.W.; Hopkins, C.; Mori, E.; Senior, B.; Harvey, R.J. Contemporary Classification of Chronic Rhinosinusitis Beyond Polyps vs. No Polyps: A Review. JAMA Otolaryngol. Head Neck Surg. 2020, 146, 831–838. [Google Scholar] [CrossRef] [PubMed]
  4. Tokunaga, T.; Sakashita, M.; Haruna, T.; Asaka, D.; Takeno, S.; Ikeda, H.; Nakayama, T.; Seki, N.; Ito, S.; Murata, J.; et al. Novel scoring system and algorithm for classifying chronic rhinosinusitis: The JESREC Study. Allergy 2015, 70, 995–1003. [Google Scholar] [CrossRef] [Green Version]
  5. Matsuwaki, Y.; Ookushi, T.; Asaka, D.; Mori, E.; Nakajima, T.; Yoshida, T.; Kojima, J.; Chiba, S.; Ootori, N.; Moriyama, H. Chronic rhinosinusitis: Risk factors for the recurrence of chronic rhinosinusitis based on 5-year follow-up after endoscopic sinus surgery. Int. Arch. Allergy Immunol. 2008, 146, 77–81. [Google Scholar] [CrossRef]
  6. Batra, P.S.; Tong, L.; Citardi, M.J. Analysis of comorbidities and objective parameters in refractory chronic rhinosinusitis. Laryngoscope 2013, 123, S1–S11. [Google Scholar] [CrossRef]
  7. Nakayama, T.; Lee, I.T.; Le, W.; Tsunemi, Y.; Borchard, N.A.; Zarabanda, D.; Dholakia, S.S.; Gall, P.A.; Yang, A.; Kim, D.; et al. Inflammatory molecular endotypes of nasal polyps derived from White and Japanese populations. J. Allergy Clin. Immunol. 2022, 149, 1296–1308. [Google Scholar] [CrossRef]
  8. Wang, W.; Gao, Z.; Wang, H.; Li, T.; He, W.; Lv, W.; Zhang, J. Transcriptome Analysis Reveals Distinct Gene Expression Profiles in Eosinophilic and Noneosinophilic Chronic Rhinosinusitis with Nasal Polyps. Sci. Rep. 2016, 6, 26604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Alpern, D.; Gardeux, V.; Russeil, J.; Mangeat, B.; Meireles-Filho, A.C.A.; Breysse, R.; Hacker, D.; Deplancke, B. BRB-seq: Ultra-affordable high-throughput transcriptomics enabled by bulk RNA barcoding and sequencing. Genome Biol. 2019, 20, 71. [Google Scholar] [CrossRef] [Green Version]
  10. Yamada, T.; Fujieda, S.; Mori, S.; Yamamoto, H.; Saito, H. Macrolide treatment decreased the size of nasal polyps and IL-8 levels in nasal lavage. Am. J. Rhinol. 2000, 14, 143–148. [Google Scholar] [CrossRef]
  11. Kim, D.H.; Kim, S.W.; Basurrah, M.A.; Hwang, S.H. Clinical and laboratory features for various criteria of eosinophilic chronic rhinosinusitis: A systematic review and meta-analysis. Clin. Exp. Otorhinolaryngol. 2022. [Google Scholar] [CrossRef] [PubMed]
  12. Matsumoto-Sasaki, M.; Simizu, K.; Suzuki, M.; Suzuki, M.; Kimura, H.; Nakamaru, Y.; Ito, Y.M.; Honma, A.; Konno, S. Clinical Characteristics of Patients and Factors Associated with Switching Biologics in Asthma. J. Asthma Allergy 2022, 15, 187–195. [Google Scholar] [CrossRef] [PubMed]
  13. Ahn, S.H.; Lee, E.J.; Ha, J.G.; Hwang, C.S.; Yoon, J.H.; Kim, C.H.; Cho, H.J. Comparison of olfactory and taste functions between eosinophilic and non-eosinophilic chronic rhinosinusitis. Auris Nasus Larynx 2020, 47, 820–827. [Google Scholar] [CrossRef] [PubMed]
  14. Morokata, T.; Suzuki, K.; Masunaga, Y.; Taguchi, K.; Morihira, K.; Sato, I.; Fujii, M.; Takizawa, S.; Torii, Y.; Yamamoto, N.; et al. A novel, selective, and orally available antagonist for CC chemokine receptor 3. J. Pharmacol. Exp. Ther. 2006, 317, 244–250. [Google Scholar] [CrossRef]
  15. 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]
  16. Yuan, Q.; Campanella, G.S.; Colvin, R.A.; Hamilos, D.L.; Jones, K.J.; Mathew, A.; Means, T.K.; Luster, A.D. Membrane-bound eotaxin-3 mediates eosinophil transepithelial migration in IL-4-stimulated epithelial cells. Eur. J. Immunol. 2006, 36, 2700–2714. [Google Scholar] [CrossRef]
  17. El-Shazly, A.E.; Doloriert, H.C.; Bisig, B.; Lefebvre, P.P.; Delvenne, P.; Jacobs, N. Novel cooperation between CX3CL1 and CCL26 inducing NK cell chemotaxis via CX3CR1: A possible mechanism for NK cell infiltration of the allergic nasal tissue. Clin. Exp. Allergy 2013, 43, 322–331. [Google Scholar] [CrossRef]
  18. Zhu, Z.; Wang, W.; Zha, Y.; Wang, X.; Wang, L.; Han, J.; Zhang, J.; Lv, W. Transcriptomic and Lipidomic Profiles in Nasal Polyps of Glucocorticoid Responders and Non-Responders: Before and After Treatment. Front. Pharmacol. 2022, 12, 814953. [Google Scholar] [CrossRef]
  19. Baba, S.; Kondo, K.; Kanaya, K.; Suzukawa, K.; Ushio, M.; Urata, S.; Asakage, T.; Kakigi, A.; Suzukawa, M.; Ohta, K.; et al. Expression of IL-33 and its receptor ST2 in chronic rhinosinusitis with nasal polyps. Laryngoscope 2014, 124, E115–E122. [Google Scholar] [CrossRef]
  20. Robertson, N.M.; Zangrilli, J.G.; Steplewski, A.; Hastie, A.; Lindemeyer, R.G.; Planeta, M.A.; Smith, M.K.; Innocent, N.; Musani, A.; Pascual, R.; et al. Differential expression of TRAIL and TRAIL receptors in allergic asthmatics following segmental antigen challenge: Evidence for a role of TRAIL in eosinophil survival. J. Immunol. 2002, 169, 5986–5996. [Google Scholar] [CrossRef] [Green Version]
  21. Zheng, T.; Zhu, Z.; Wang, J.; Homer, R.J.; Elias, J.A. IL-11: Insights in asthma from overexpression transgenic modeling. J. Allergy Clin. Immunol. 2001, 108, 489–496. [Google Scholar] [CrossRef] [PubMed]
  22. Shah, S.A.; Ishinaga, H.; Hou, B.; Okano, M.; Takeuchi, K. Effects of interleukin-31 on MUC5AC gene expression in nasal allergic inflammation. Phamacology 2013, 91, 158–164. [Google Scholar] [CrossRef]
  23. Jarjour, N.N.; Bradstreet, T.R.; Schwarzkopf, E.A.; Cook, M.E.; Lai, C.W.; Huang, S.C.; Taneja, R.; Stappenbeck, T.S.; Dyken, S.J.; Urban, J.F., Jr.; et al. BHLHE40 Promotes TH2 Cell-Mediated Antihelminth Immunity and Reveals Cooperative CSF2RB Family Cytokines. J. Immunol. 2020, 204, 923–932. [Google Scholar] [CrossRef] [PubMed]
  24. Sunadome, H.; Matsumoto, H.; Petrova, G.; Kanemitsu, Y.; Tohda, Y.; Horiguchi, T.; Kita, H.; Kuwabara, K.; Tomii, K.; Otsuka, K.; et al. IL4Rα and ADAM33 as genetic markers in asthma exacerbations and type-2 inflammatory endotype. Clin. Exp. Allergy 2017, 47, 998–1006. [Google Scholar] [CrossRef] [PubMed]
  25. Fujieda, S.; Matsune, S.; Takeno, S.; Ohta, N.; Asako, M.; Bachert, C.; Inoue, T.; Takahashi, Y.; Fujita, H.; Deniz, Y.; et al. Dupilumab efficacy in chronic rhinosinusitis with nasal polyps from SINUS-52 is unaffected by eosinophilic status. Allergy 2022, 77, 186–196. [Google Scholar] [CrossRef] [PubMed]
  26. Workman, A.D.; Nocera, A.L.; Mueller, S.K.; Otu, H.H.; Libermann, T.A.; Bleier, B.S. Translating transcription: Proteomics in chronic rhinosinusitis with nasal polyps reveals significant discordance with messenger RNA expression. Int. Forum Allergy Rhinol. 2019, 9, 776–786. [Google Scholar] [CrossRef]
  27. Picelli, S. Full-Length Single-Cell RNA Sequencing with Smart-seq2. Methods Mol. Biol. 2019, 1979, 25–44. [Google Scholar] [CrossRef]
  28. Sato, S.; Arimura, Y.; Kujirai, T.; Harada, A.; Maehara, K.; Nogami, J.; Ohkawa, Y.; Kurumizaka, H. Biochemical analysis of nucleosome targeting by Tn5 transposase. Open Biol. 2019, 9, 190116. [Google Scholar] [CrossRef] [Green Version]
Figure 1. PCA and heatmap of the correlation matrix performed among all groups. The control group was clearly segregated from the other groups. ECRS, nonECRS, and Asp were not segregated from each other (A). The heatmap of the correlation matrix also showed the difficulty of the segregation among the three CRSwNP groups (B).
Figure 1. PCA and heatmap of the correlation matrix performed among all groups. The control group was clearly segregated from the other groups. ECRS, nonECRS, and Asp were not segregated from each other (A). The heatmap of the correlation matrix also showed the difficulty of the segregation among the three CRSwNP groups (B).
Ijms 23 05653 g001
Figure 2. Hierarchical cluster analysis with cutoff Z score of 4. No distinct segregation among the three CRSwNP groups was seen and some ECRS (ECRS1) and nonECRS (nonECRS5 and 7) patients were in a similar cluster to the control group.
Figure 2. Hierarchical cluster analysis with cutoff Z score of 4. No distinct segregation among the three CRSwNP groups was seen and some ECRS (ECRS1) and nonECRS (nonECRS5 and 7) patients were in a similar cluster to the control group.
Ijms 23 05653 g002
Figure 3. DEGs in ECRS vs. control (ECRS-Ctrl), 677 upregulated and 479 downregulated; nonECRS vs Ctrl (nonECRS-Ctrl), 695 and 310; Asp vs Ctrl (Asp-Ctrl), 380 and 463; Asp vs nonECRS (Asp-nonECRS), 3 and 16; and Asp vs ECRS (Asp-ECRS), 1 and 9. No DEGs were identified in ECRS vs nonECRS (ECRS-nonECRS).
Figure 3. DEGs in ECRS vs. control (ECRS-Ctrl), 677 upregulated and 479 downregulated; nonECRS vs Ctrl (nonECRS-Ctrl), 695 and 310; Asp vs Ctrl (Asp-Ctrl), 380 and 463; Asp vs nonECRS (Asp-nonECRS), 3 and 16; and Asp vs ECRS (Asp-ECRS), 1 and 9. No DEGs were identified in ECRS vs nonECRS (ECRS-nonECRS).
Ijms 23 05653 g003
Figure 4. Venn diagram of all CRSwNP groups vs control. Specific DEGs were identified in each CRSwNP group as ECRS-Ctrl of 338, nonECRS-Ctrl of 271, and Asp-Ctrl of 355.
Figure 4. Venn diagram of all CRSwNP groups vs control. Specific DEGs were identified in each CRSwNP group as ECRS-Ctrl of 338, nonECRS-Ctrl of 271, and Asp-Ctrl of 355.
Ijms 23 05653 g004
Figure 5. Pathway analysis with the GO molecular function with DEGs. ECRS-Ctrl (A), nonECRS-Ctrl (B), and Asp-Ctrl (C) showed the existence of significant enrichment pathways. Two pathways (nodes) are connected if they share 30% or more genes. Green and red represents down and up regulated pathways, respectively. Darker nodes show more significantly enriched gene sets. Bigger nodes represent larger gene sets. Thicker edges represent more overlapping genes.
Figure 5. Pathway analysis with the GO molecular function with DEGs. ECRS-Ctrl (A), nonECRS-Ctrl (B), and Asp-Ctrl (C) showed the existence of significant enrichment pathways. Two pathways (nodes) are connected if they share 30% or more genes. Green and red represents down and up regulated pathways, respectively. Darker nodes show more significantly enriched gene sets. Bigger nodes represent larger gene sets. Thicker edges represent more overlapping genes.
Ijms 23 05653 g005aIjms 23 05653 g005bIjms 23 05653 g005c
Figure 6. Hierarchic cluster analysis with all CRSwNPs segregated into two clusters (cluster1 and 2) among these CRSwNPs. Segregation of two clusters and comorbid of asthma were not correlated.
Figure 6. Hierarchic cluster analysis with all CRSwNPs segregated into two clusters (cluster1 and 2) among these CRSwNPs. Segregation of two clusters and comorbid of asthma were not correlated.
Ijms 23 05653 g006
Figure 7. PCA analysis and DEGs of new clusters. (A) The new clusters showed clear separation from the control in PCA. (B) DEGs were identified not only for both cluster1-Ctrl and cluster2-Ctrl but also for cluster1-cluster2.
Figure 7. PCA analysis and DEGs of new clusters. (A) The new clusters showed clear separation from the control in PCA. (B) DEGs were identified not only for both cluster1-Ctrl and cluster2-Ctrl but also for cluster1-cluster2.
Ijms 23 05653 g007
Figure 8. KEGG pathway analysis in “cytokine–cytokine receptor interaction” by DAVID for upregulated DEGs. Red stars represent the upregulated DEGs. Compared to cluster1-Ctrl (A) and cluster2-Ctrl (B), cluster2 was more upregulated for genes of the CC subfamily, CXC subfamily, gamma-chain utilizing, IL-4-like, IL6/12-like, IL-1-like cytokine, and TNF family.
Figure 8. KEGG pathway analysis in “cytokine–cytokine receptor interaction” by DAVID for upregulated DEGs. Red stars represent the upregulated DEGs. Compared to cluster1-Ctrl (A) and cluster2-Ctrl (B), cluster2 was more upregulated for genes of the CC subfamily, CXC subfamily, gamma-chain utilizing, IL-4-like, IL6/12-like, IL-1-like cytokine, and TNF family.
Ijms 23 05653 g008
Table 1. KEGG pathways using the upregulated DEGs into DAVID. Genes with the disease-associated terms “cytokine–cytokine receptor interaction”, “hematopoietic cell lineage”, “chemokine signaling pathway”, “complement and coagulation cascade”, “osteoclast differentiation”, “Staphylococcus aureus infection”, and “viral protein interaction with cytokine and cytokine receptor” were commonly upregulated in all CRSwNP groups vs control.
Table 1. KEGG pathways using the upregulated DEGs into DAVID. Genes with the disease-associated terms “cytokine–cytokine receptor interaction”, “hematopoietic cell lineage”, “chemokine signaling pathway”, “complement and coagulation cascade”, “osteoclast differentiation”, “Staphylococcus aureus infection”, and “viral protein interaction with cytokine and cytokine receptor” were commonly upregulated in all CRSwNP groups vs control.
ComparisonTermCount%p-ValueBenjamini
ECRS-
Ctrl UP
hsa05200:Pathways in cancer355.26.7 × 10−42.6 × 10−2
hsa04060:Cytokine–cytokine receptor interaction253.71.4 × 10−41.0 × 10−2
hsa04080:Neuroactive ligand–receptor interaction223.21.5 × 10−22.5 × 10−1
hsa04151:PI3K-Akt signaling pathway192.88.5 × 10−27.3 × 10−1
hsa05166:Human T-cell leukemia virus 1 infection182.72.6 × 10−37.0 × 10−2
hsa04020:Calcium signaling pathway182.75.6 × 10−31.0 × 10−1
hsa04640:Hematopoietic cell lineage172.53.8 × 10−71.0 × 10−4
hsa04145:Phagosome162.43.5 × 10−41.9 × 10−2
hsa04062:Chemokine signaling pathway162.43.7 × 10−37.8 × 10−2
hsa04610:Complement and coagulation cascades152.21.6 × 10−62.2 × 10−4
hsa04380:Osteoclast differentiation142.16.5 × 10−42.6 × 10−2
hsa05202:Transcriptional misregulation in cancer142.12.1 × 10−22.8 × 10−1
hsa05150:Staphylococcus aureus infection131.91.5 × 10−41.0 × 10−2
hsa05152:Tuberculosis131.92.8 × 10−23.4 × 10−1
hsa04061:Viral protein interaction with cytokine and cytokine receptor121.88.7 × 10−43.0 × 10−2
hsa05142:Chagas disease111.63.5 × 10−37.8 × 10−2
hsa05146:Amoebiasis111.63.5 × 10−37.8 × 10−2
hsa04514:Cell adhesion molecules111.64.2 × 10−24.2 × 10−1
hsa05140:Leishmaniasis101.51.6 × 10−35.0 × 10−2
hsa04659:Th17 cell differentiation101.51.5 × 10−22.5 × 10−1
hsa04658:Th1 and Th2 cell differentiation91.31.7 × 10−22.6 × 10−1
hsa04611:Platelet activation91.37.7 × 10−26.8 × 10−1
hsa04512:ECM-receptor interaction81.23.8 × 10−24.0 × 10−1
hsa04750:Inflammatory mediator regulation of TRP channels81.26.2 × 10−25.6 × 10−1
hsa04066:HIF-1 signaling pathway81.29.6 × 10−28.0 × 10−1
hsa04978:Mineral absorption71.02.0 × 10−22.8 × 10−1
hsa00590:Arachidonic acid metabolism71.02.2 × 10−22.8 × 10−1
hsa05321:Inflammatory bowel disease71.02.8 × 10−23.4 × 10−1
hsa04664:Fc epsilon RI signaling pathway71.03.5 × 10−23.8 × 10−1
hsa05133:Pertussis71.05.5 × 10−25.3 × 10−1
hsa05310:Asthma60.94.5 × 10−38.8 × 10−2
hsa05219:Bladder cancer50.75.8 × 10−25.5 × 10−1
hsa00532:Glycosaminoglycan biosynthesis—chondroitin sulfate/dermatan sulfate40.63.3 × 10−23.8 × 10−1
nonECRS-Ctrl UPhsa04060:Cytokine–cytokine receptor interaction334.71.5 × 10−82.0 × 10−6
hsa05200:Pathways in cancer334.72.4 × 10−32.7 × 10−2
hsa04080:Neuroactive ligand–receptor interaction233.37.8 × 10−37.6 × 10−2
hsa04062:Chemokine signaling pathway223.14.5 × 10−61.7 × 10−4
hsa05166:Human T-cell leukemia virus 1 infection213.01.3 × 10−42.2 × 10−3
hsa04061:Viral protein interaction with cytokine and cytokine receptor192.71.2 × 10−82.0 × 10−6
hsa05150:Staphylococcus aureus infection182.63.9 × 10−83.4 × 10−6
hsa05152:Tuberculosis182.62.3 × 10−43.6 × 10−3
hsa04640:Hematopoietic cell lineage172.43.7 × 10−72.4 × 10−5
hsa05417:Lipid and atherosclerosis172.44.3 × 10−34.5 × 10−2
hsa05323:Rheumatoid arthritis162.38.8 × 10−74.6 × 10−5
hsa04145:Phagosome162.33.4 × 10−44.7 × 10−3
hsa05171:Coronavirus disease—COVID-19162.31.9 × 10−21.3 × 10−1
hsa04659:Th17 cell differentiation152.12.8 × 10−57.3 × 10−4
hsa04380:Osteoclast differentiation152.11.8 × 10−43.0 × 10−3
hsa05167:Kaposi sarcoma-associated herpesvirus infection152.19.6 × 10−38.5 × 10−2
hsa05169:Epstein–Barr virus infection152.11.3 × 10−21.1 × 10−1
hsa05163:Human cytomegalovirus infection152.13.1 × 10−21.9 × 10−1
hsa05321:Inflammatory bowel disease131.92.6 × 10−61.1 × 10−4
hsa05140:Leishmaniasis131.91.6 × 10−54.7 × 10−4
hsa04658:Th1 and Th2 cell differentiation131.99.7 × 10−52.0 × 10−3
hsa04630:JAK-STAT signaling pathway131.91.3 × 10−21.1 × 10−1
hsa05164:Influenza A131.91.9 × 10−21.3 × 10−1
hsa05142:Chagas disease121.79.9 × 10−41.2 × 10−2
hsa04668:TNF signaling pathway121.72.1 × 10−32.5 × 10−2
hsa05322:Systemic lupus erythematosus121.79.3 × 10−38.5 × 10−2
hsa04936:Alcoholic liver disease121.71.3 × 10−21.1 × 10−1
hsa04514:Cell adhesion molecules121.71.8 × 10−21.3 × 10−1
hsa05202:Transcriptional misregulation in cancer121.78.3 × 10−24.7 × 10−1
hsa04610:Complement and coagulation cascades111.68.5 × 10−41.1 × 10−2
hsa04650:Natural killer cell mediated cytotoxicity111.61.5 × 10−21.1 × 10−1
hsa05161:Hepatitis B111.66.5 × 10−23.9 × 10−1
hsa04657:IL-17 signaling pathway101.46.3 × 10−36.4 × 10−2
hsa05145:Toxoplasmosis101.41.9 × 10−21.3 × 10−1
hsa05310:Asthma91.39.3 × 10−63.1 × 10−4
hsa05330:Allograft rejection91.34.6 × 10−51.1 × 10−3
hsa05332:Graft-versus-host disease91.39.9 × 10−52.0 × 10−3
hsa04940:Type I diabetes mellitus91.31.2 × 10−42.2 × 10−3
hsa04672:Intestinal immune network for IgA production91.33.0 × 10−44.5 × 10−3
hsa05320:Autoimmune thyroid disease91.35.3 × 10−47.0 × 10−3
hsa04662:B cell receptor signaling pathway91.38.7 × 10−38.2 × 10−2
hsa04612:Antigen processing and presentation81.12.1 × 10−21.4 × 10−1
hsa05146:Amoebiasis81.17.2 × 10−24.2 × 10−1
hsa04625:C-type lectin receptor signaling pathway81.17.8 × 10−24.5 × 10−1
hsa05416:Viral myocarditis71.02.0 × 10−21.3 × 10−1
hsa05133:Pertussis71.05.4 × 10−23.3 × 10−1
hsa05219:Bladder cancer60.91.5 × 10−21.1 × 10−1
hsa04664:Fc epsilon RI signaling pathway60.99.5 × 10−25.2 × 10−1
Asp-Ctrl UPhsa05200:Pathways in cancer246.36.4 × 10−31.3 × 10−1
hsa04151:PI3K-Akt signaling pathway205.21.3 × 10−34.5 × 10−2
hsa04060:Cytokine–cytokine receptor interaction195.03.9 × 10−41.9 × 10−2
hsa04062:Chemokine signaling pathway164.28.4 × 10−51.0 × 10−2
hsa05202:Transcriptional misregulation in cancer153.93.0 × 10−41.8 × 10−2
hsa04630:JAK-STAT signaling pathway143.71.9 × 10−41.6 × 10−2
hsa05206:MicroRNAs in cancer143.74.6 × 10−24.1 × 10−1
hsa05205:Proteoglycans in cancer133.45.1 × 10−31.3 × 10−1
hsa04610:Complement and coagulation cascades123.17.4 × 10−61.8 × 10−3
hsa05166:Human T-cell leukemia virus 1 infection123.12.3 × 10−23.0 × 10−1
hsa04020:Calcium signaling pathway123.13.7 × 10−23.5 × 10−1
hsa05167:Kaposi sarcoma-associated herpesvirus infection112.92.3 × 10−23.0 × 10−1
hsa05132:Salmonella infection112.99.3 × 10−26.2 × 10−1
hsa04144:Endocytosis112.99.4 × 10−26.2 × 10−1
hsa04380:Osteoclast differentiation102.64.6 × 10−31.2 × 10−1
hsa04015:Rap1 signaling pathway102.67.9 × 10−26.0 × 10−1
hsa04625:C-type lectin receptor signaling pathway92.34.3 × 10−31.2 × 10−1
hsa04066:HIF−1 signaling pathway92.35.7 × 10−31.3 × 10−1
hsa04061:Viral protein interaction with cytokine and cytokine receptor82.11.2 × 10−22.3 × 10−1
hsa05142:Chagas disease82.11.3 × 10−22.4 × 10−1
hsa04064:NF-kappa B signaling pathway82.11.5 × 10−22.4 × 10−1
hsa04668:TNF signaling pathway82.12.2 × 10−23.0 × 10−1
hsa04650:Natural killer cell mediated cytotoxicity82.13.8 × 10−23.5 × 10−1
hsa05135:Yersinia infection82.15.5 × 10−24.8 × 10−1
hsa04072:Phospholipase D signaling pathway82.17.6 × 10−26.0 × 10−1
hsa04218:Cellular senescence82.19.4 × 10−26.2 × 10−1
hsa05219:Bladder cancer71.84.8 × 10−42.0 × 10−2
hsa04657:IL-17 signaling pathway71.82.9 × 10−23.5 × 10−1
hsa05150:Staphylococcus aureus infection71.83.2 × 10−23.5 × 10−1
hsa04666:Fc gamma R-mediated phagocytosis71.83.4 × 10−23.5 × 10−1
hsa04640:Hematopoietic cell lineage71.83.7 × 10−23.5 × 10−1
hsa04115:p53 signaling pathway61.63.5 × 10−23.5 × 10−1
hsa04012:ErbB signaling pathway61.66.0 × 10−25.1 × 10−1
hsa05210:Colorectal cancer61.66.3 × 10−25.1 × 10−1
hsa04216:Ferroptosis51.31.8 × 10−22.8 × 10−1
hsa05211:Renal cell carcinoma51.39.3 × 10−26.2 × 10−1
hsa05230:Central carbon metabolism in cancer51.39.6 × 10−26.2 × 10−1
hsa05120:Epithelial cell signaling in Helicobacter pylori infection51.39.6 × 10−26.2 × 10−1
Table 2. Upregulated genes for “cytokine–cytokine receptor interaction.” Upregulated DEGs were represented with 〇. CCL13, CCL18, CCL26, TNFRSF18, INHBB, IL1RL1, and IL2RA in ECRS-Ctrl were more upregulated compared to nonECRS-Ctrl, and CCL2, CCL8, CCL20, CCR5, CXCL1, CXCL6, CXCR2, FAS, TNFRSF1B, TNFSF13B, CSF3, IL2RG, IL20RB, and IL23A in nonECRS-Ctrl were more upregulated compared to ECRS-Ctrl. The upregulated genes in Asp-Ctrl were similar to the DEGs of ECRS-Ctrl and nonECRS-Ctrl, but TNFRSF10D, IL6, IL11, IL31RA were only upregulated in Asp-Ctrl compared with the DEGs of ECRS-Ctrl and nonECRS-Ctrl.
Table 2. Upregulated genes for “cytokine–cytokine receptor interaction.” Upregulated DEGs were represented with 〇. CCL13, CCL18, CCL26, TNFRSF18, INHBB, IL1RL1, and IL2RA in ECRS-Ctrl were more upregulated compared to nonECRS-Ctrl, and CCL2, CCL8, CCL20, CCR5, CXCL1, CXCL6, CXCR2, FAS, TNFRSF1B, TNFSF13B, CSF3, IL2RG, IL20RB, and IL23A in nonECRS-Ctrl were more upregulated compared to ECRS-Ctrl. The upregulated genes in Asp-Ctrl were similar to the DEGs of ECRS-Ctrl and nonECRS-Ctrl, but TNFRSF10D, IL6, IL11, IL31RA were only upregulated in Asp-Ctrl compared with the DEGs of ECRS-Ctrl and nonECRS-Ctrl.
GenesECRS-CtrlNonECRS-CtrlAsp-Ctrl
Upregulated DEGsCCL2
CCL8
CCL11
CCL13
CCL15
CCL18
CCL20
CCL26
CCR1
CCR5
CXCL1
CXCL6
CXCL8
CXCR2
CD4
FAS
LIF
TNFRSF1B
TNFRSF10D
TNFRSF12A
TNFRSF18
TNFRSF21
TNFSF13B
ACKR3
CSF1R
CSF2RB
CSF3
INHBB
IL1RL1
IL2RA
IL2RB
IL2RG
IL4R
IL5RA
IL6
IL6R
IL11
IL16
IL18R1
IL20RB
IL23A
IL31RA
OSMR
TGFB3
Table 3. KEGG pathways using the downregulated DEGs into the DAVID. Genes associated with “metabolic pathways”, “salivary secretion”, “mucin type O-glycan biosynthesis”, and “glycine, serine and threonine metabolism” were commonly downregulated. The gene count for “metabolic pathways” was the highest among all the CRSwNP groups vs control, and the gene counts were widely distributed as ECRS-Ctrl of 55, nonECRS-Ctrl of 36, and Asp-Ctrl of 65.
Table 3. KEGG pathways using the downregulated DEGs into the DAVID. Genes associated with “metabolic pathways”, “salivary secretion”, “mucin type O-glycan biosynthesis”, and “glycine, serine and threonine metabolism” were commonly downregulated. The gene count for “metabolic pathways” was the highest among all the CRSwNP groups vs control, and the gene counts were widely distributed as ECRS-Ctrl of 55, nonECRS-Ctrl of 36, and Asp-Ctrl of 65.
ComparisonTermCount%p-ValueBenjamini
ECRS-Ctrl downhsa01100:Metabolic pathways5511.5 5.6 × 10−32.4 × 10−1
hsa04970:Salivary secretion153.1 7.6 × 10−81.9 × 10−5
hsa04024:cAMP signaling pathway122.5 2.5 × 10−28.1 × 10−1
hsa04972:Pancreatic secretion112.3 2.5 × 10−43.2 × 10−2
hsa04550:Signaling pathways regulating pluripotency of stem cells112.3 3.4 × 10−31.8 × 10−1
hsa04020:Calcium signaling pathway112.3 8.5 × 10−21.0
hsa05215:Prostate cancer91.9 3.2 × 10−31.8 × 10−1
hsa00260:Glycine, serine and threonine metabolism61.3 3.2 × 10−31.8 × 10−1
hsa05412:Arrhythmogenic right ventricular cardiomyopathy61.3 4.6 × 10−21.0
hsa04911:Insulin secretion61.3 6.7 × 10−21.0
hsa04350:TGF-beta signaling pathway61.3 9.0 × 10−21.0
hsa00512:Mucin type O-glycan biosynthesis51.0 1.3 × 10−24.6 × 10−1
hsa00514:Other types of O-glycan biosynthesis51.0 3.1 × 10−28.8 × 10−1
hsa05031:Amphetamine addiction51.0 9.8 × 10−21.0
hsa00100:Steroid biosynthesis30.6 9.0 × 10−21.0
nonECRS-Ctrl downhsa01100:Metabolic pathways3611.6 7.1 × 10−33.9 × 10−1
hsa04970:Salivary secretion134.2 1.4 × 10−83.1 × 10−6
hsa04972:Pancreatic secretion103.2 2.4 × 10−52.6 × 10−3
hsa04024:cAMP signaling pathway103.2 6.6 × 10−33.9 × 10−1
hsa04911:Insulin secretion61.9 1.0 × 10−24.4 × 10−1
hsa05231:Choline metabolism in cancer51.6 6.3 × 10−21.0
hsa00512:Mucin type O-glycan biosynthesis41.3 1.7 × 10−25.9 × 10−1
hsa05143:African trypanosomiasis41.3 1.9 × 10−25.9 × 10−1
hsa00260:Glycine, serine and threonine metabolism41.3 2.3 × 10−26.3 × 10−1
hsa00280:Valine, leucine and isoleucine degradation41.3 3.7 × 10−29.0 × 10−1
hsa05031:Amphetamine addiction41.3 8.8 × 10−21.0
Asp-Ctrl downhsa01100:Metabolic pathways6514.0 2.4 × 10−65.9 × 10−4
hsa04970:Salivary secretion122.6 1.3 × 10−51.7 × 10−3
hsa01200:Carbon metabolism81.7 2.1 × 10−27.5 × 10−1
hsa00280:Valine, leucine and isoleucine degradation71.5 9.7 × 10−48.0 × 10−2
hsa04972:Pancreatic secretion71.5 3.7 × 10−29.1 × 10−1
hsa00514:Other types of O-glycan biosynthesis61.3 5.3 × 10−33.1 × 10−1
hsa00520:Amino sugar and nucleotide sugar metabolism61.3 6.3 × 10−33.1 × 10−1
hsa04976:Bile secretion61.3 6.4 × 10−21.0
hsa00564:Glycerophospholipid metabolism61.3 8.8 × 10−21.0
hsa01250:Biosynthesis of nucleotide sugars51.1 1.2 × 10−24.8 × 10−1
hsa00620:Pyruvate metabolism51.1 2.6 × 10−28.0 × 10−1
hsa00650:Butanoate metabolism40.9 2.9 × 10−28.0 × 10−1
hsa00640:Propanoate metabolism40.9 4.1 × 10−29.3 × 10−1
hsa00512:Mucin type O-glycan biosynthesis40.9 5.5 × 10−21.0
hsa00260:Glycine, serine and threonine metabolism40.9 7.2 × 10−21.0
hsa02010:ABC transporters40.9 9.4 × 10−21.0
hsa00100:Steroid biosynthesis30.6 8.3 × 10−21.0
hsa00900:Terpenoid backbone biosynthesis30.6 9.8 × 10−21.0
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ishino, T.; Takeno, S.; Takemoto, K.; Yamato, K.; Oda, T.; Nishida, M.; Horibe, Y.; Chikuie, N.; Kono, T.; Taruya, T.; et al. Distinct Gene Set Enrichment Profiles in Eosinophilic and Non-Eosinophilic Chronic Rhinosinusitis with Nasal Polyps by Bulk RNA Barcoding and Sequencing. Int. J. Mol. Sci. 2022, 23, 5653. https://doi.org/10.3390/ijms23105653

AMA Style

Ishino T, Takeno S, Takemoto K, Yamato K, Oda T, Nishida M, Horibe Y, Chikuie N, Kono T, Taruya T, et al. Distinct Gene Set Enrichment Profiles in Eosinophilic and Non-Eosinophilic Chronic Rhinosinusitis with Nasal Polyps by Bulk RNA Barcoding and Sequencing. International Journal of Molecular Sciences. 2022; 23(10):5653. https://doi.org/10.3390/ijms23105653

Chicago/Turabian Style

Ishino, Takashi, Sachio Takeno, Kota Takemoto, Kensuke Yamato, Takashi Oda, Manabu Nishida, Yuichiro Horibe, Nobuyuki Chikuie, Takashi Kono, Takayuki Taruya, and et al. 2022. "Distinct Gene Set Enrichment Profiles in Eosinophilic and Non-Eosinophilic Chronic Rhinosinusitis with Nasal Polyps by Bulk RNA Barcoding and Sequencing" International Journal of Molecular Sciences 23, no. 10: 5653. https://doi.org/10.3390/ijms23105653

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