Back to Journals » Journal of Inflammation Research » Volume 18
Characterization of Mucosal Immune-Related lncRNAs and mRNAs in a Mouse Model of Allergic Conjunctivitis
Authors Zhang H, Zhang H, Leng Q, Zheng YJ
Received 10 December 2024
Accepted for publication 25 April 2025
Published 8 May 2025 Volume 2025:18 Pages 6061—6076
DOI https://doi.org/10.2147/JIR.S511048
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 3
Editor who approved publication: Professor Ning Quan
Hong Zhang,1,2 Hongyu Zhang,3 Qing Leng,3 Ya Juan Zheng1
1Department of Ophthalmology, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, People’s Republic of China; 2Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China; 3Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, People’s Republic of China
Correspondence: Ya Juan Zheng, Email [email protected]
Background: Allergic conjunctivitis (AC) is a common inflammatory condition characterized by immune dysregulation in response to environmental allergens. Despite extensive research into general allergic mechanisms, the specific immunological features of the ocular mucosal microenvironment remain poorly understood. Investigating immune-related mRNAs and LncRNAs may provide insights into the mechanisms underlying AC and potential novel targets for therapeutic intervention.
Methods: An AC model was established using female BALB/c mice sensitized with ragweed pollen. Conjunctival tissues from AC and control groups were pooled for RNA extraction, followed by Illumina sequencing. Differential gene expression was identified using DESeq2, and functional enrichment was analyzed using GO, KEGG, and GSEA. RT-qPCR validated results, while the Human Protein Atlas was used to assess protein expression.
Results: A murine model of AC was successfully established, confirmed by progressively increasing clinical scores and significantly elevated scratching frequency. Transcriptomic analysis revealed significant differences in mRNAs and lncRNAs expression between AC and control groups. GO analysis indicated that both upregulated and downregulated genes were enriched in biological processes related to response to stimulus, immune system processes, signaling, and metabolic processes. KEGG analysis showed that upregulated genes were enriched in pathways such as steroid hormone biosynthesis, histidine metabolism, glycolysis/gluconeogenesis, and IL-17 signaling, while downregulated genes were involved in cytokine-cytokine receptor interaction and hematopoietic cell lineage. GSEA identified significant enrichment in inflammatory pathways, including MAPK, STAT1, and STAT2. Mucosal immunity-related genes such as Bpifa1, Lcn2, and Reg3g were upregulated in AC. Co-expression analysis also revealed several upregulated lncRNAs, including Stoml3-202 and Etohd2-205.
Conclusion: This study is the first to systematically analyze immune-related mRNAs and LncRNAs in AC, identifying mucosal immunity molecules like Bpifa1 and Reg3g. These findings underscore the unique involvement of mucosal immunity in AC and provide potential new targets for immune modulation in ocular allergy treatment.
Keywords: allergic conjunctivitis, lncRNA, transcriptomic analysis, immune-related genes
Introduction
Allergic conjunctivitis (AC) is a common yet challenging inflammatory condition to treat effectively. Triggered by allergens such as pollen, animal dander, and environmental irritants, it presents with symptoms including red and itchy eyes, burning sensations, stinging, swelling, and tearing.1 Despite its high prevalence, affecting 10–20% of the global population, the underlying mechanisms are not fully understood.2,3 The widely accepted mechanism is that the immunopathogenesis of AC begins with the conjunctiva’s initial exposure to environmental allergens.4 These allergens disrupt the conjunctival epithelial barrier, allowing allergens to be captured by antigen-presenting cells (APCs) in the conjunctival stroma.5 This results in the production of IL-4-secreting Th2 cells, which induce B cells to differentiate into plasma cells that secrete allergen-specific immunoglobulin E (IgE). IgE binds to FcεRI receptors on mast cells. Upon re-exposure to allergens, activated conjunctival mast cells degranulate, releasing mediators like histamine, lipid mediators, cytokines and chemokines, leading to the clinical manifestations of AC.6
The ocular surface is highly exposed to environmental threats and combats pathogenic threats. However, excessive inflammation can lead to ocular dysfunction, and even impair vision, potentially resulting in blindness.7,8 To protect the eye, the ocular mucosa maintains immune tolerance through various mechanisms, exhibiting a lower immune response to external stimuli and thereby reducing the occurrence of excessive inflammatory reactions. This is distinct from the immune privilege observed within intraocular structures.9,10 Peripheral immune tolerance involves various immunosuppressive factors and immune cells that create an anti-inflammatory microenvironment.11 Currently, conjunctival goblet cells are known to secrete the immunosuppressive factor TGF-β.12 Further research is needed to identify other secretions and cell types involved in ocular immune tolerance.
However, low immune response increases susceptibility to infections. To combat microbial infections, corneal and conjunctival epithelial cells respond to pathogens via functional Toll-like and NOD-like receptor signaling systems, secreting antimicrobial components and immunomodulatory factors like lysozyme, lactoferrin, and secretory antibody.12–15 These mechanisms protect the eye, minimizing tissue damage and scar formation caused by inflammation. Due to these unique low immune response mechanisms, the pathogenesis of AC in the conjunctiva may differ from other tissues. For example, studies have shown that mast cells may not play a significant role in the development of ragweed-induced AC in mouse models.16 Current research on AC often relies on classical immune mechanisms, lacking studies that focus on the unique immunological aspects of AC. This gap may impact our understanding of the allergic mechanisms occurring in the conjunctiva.
Non-coding RNA species, including long non-coding RNAs (LncRNAs), are integral in regulating immune responses, particularly in allergic diseases such as asthma, atopic dermatitis, and food allergies.17 Recent studies have emphasized the involvement of LncRNAs in type-2 immune responses, which play a pivotal role in mediating allergic inflammation. Transcriptome sequencing (RNA-seq) is a powerful tool for profiling gene expression and discovering novel RNA molecules, including LncRNAs, that may be involved in the pathogenesis of these diseases.18 However, obtaining conjunctival tissue samples from patients during allergic reactions remains challenging, resulting in a limited number of transcriptomic studies on AC.
In our study, we simulated clinically common seasonal AC by sensitizing mice with ragweed pollen to establish an AC model. We performed gene sequencing on the conjunctival tissues of these mice to identify immune-related mRNAs and LncRNAs. This research aims to elucidate the molecular mechanisms underlying AC and potentially provide new perspectives and therapeutic targets for future studies and treatment development.
Materials and Methods
Establishment of AC Model
All experimental procedures were approved by the Animal Ethics Committee of Jilin University (Approval No. 197) and strictly complied with China’s national standard GB/T 35892–2018 “Guidelines for Ethical Review of Laboratory Animal Welfare”. The study was conducted at the Laboratory Animal Center of Basic Medical Sciences, Jilin University, which holds the institutional animal care license (SYXK (Ji) 2023–0010) for SPF-grade rodents under barrier conditions. AC model was established as described previously.19 Female BALB/c mice (8–10 weeks old) were purchased from Yisi Laboratory Animal Technology Co., Ltd. (Changchun, China). All animals were housed under standard laboratory conditions, including a 12-hour light/dark cycle, temperature of 22 ± 2°C, and humidity of 55 ± 5%, with food and water available ad libitum. Female BALB/c mice were chosen for their strong Th2 immune response, relevant to allergic reactions. On day 0, mice were sensitized via footpad injection of 100 µg short ragweed (SRW) pollen (Greer Lab, Lenoir, NC, USA) mixed with 2 mg Imject Alum (InvivoGen, San Diego, CA, USA), while control mice received Imject Alum alone. Starting on day 10, 10 µL of 50 mg/mL SRW pollen solution was administered to each eye daily for five consecutive days; control mice received 10 µL phosphate buffered solution (PBS), which was used as the diluent for SRW.
Clinical symptoms, including chemosis, conjunctival redness, eyelid edema, and tearing, were scored 20 minutes after each SRW challenge on a scale of 0 (absent) to 3 (severe). In addition, scratching behavior was recorded as a measure of ocular itch. The frequency of scratching was assessed during the challenge phase (Days 10 to 14), within 20 minutes after each SRW administration. Scratching behavior was manually observed using a stopwatch in a double-blinded manner. A single scratching event was defined as a continuous contact of the eye area with either the forepaw or hind paw lasting ≥1 second, with at least a 2-second interval between events to avoid counting unconscious movements.
RNA Isolation and Sequencing
Conjunctival tissues from both eyes of two mice were pooled to generate a single RNA sample, yielding a total of 5 samples from 10 control mice and 5 samples from 10 AC model mice. This pooling was necessary due to the limited tissue volume from individual conjunctival samples, which restricts RNA yield. Total RNA was extracted from these pooled samples using TRIzol reagent (Invitrogen, Carlsbad, CA, USA), following the manufacturer’s instructions. RNA integrity was evaluated via standard denaturing agarose gel electrophoresis. RNA concentration for each sample was measured using a using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Wilmington, DE, USA). Agilent 2100 bioanalyzer (Agilent Technologies, SantaClara, CA, USA) was used to test the RNA integrity. Each RNA sample was then divided into two portions: one for transcriptome sequencing and the other for reverse transcription quantitative polymerase chain reaction (RT-qPCR).
RNA library preparation and RNA library sequencing were performed by Gene Denovo Biotechnology Co., Ltd. (Guangzhou, China). Prior to the construction of the RNA libraries, the rRNA Ribo-off rRNA Depletion Kit (Vazyme, N406, Nanjing, China) was used to remove the rRNA. Then, rRNA-depleted RNA was used to construct the RNA library with a VAHTS Universal V6 RNA-seq Library Prep Kit for Illumina (Vazyme, NR604, Nanjing, China). In the process, the extracted mRNA is enriched using mRNA Capture Beads. After purification with beads, the mRNA is fragmented using high temperatures. The fragmented mRNA is then used as a template to synthesize the first strand of cDNA in a reverse transcription enzyme mixture system. While synthesizing the second strand of cDNA, end repair and A-tailing are completed. Next, adapters are ligated, and VAHTS DNA Selection Beads are used for purification to select target fragments. PCR library amplification is then performed. The library quality was evaluated with a DNA 1000 assay Kit (Agilent Technologies, SantaClara, CA, USA). Finally, detection is carried out using the Illumina Novaseq X Plus by Gene Denovo Biotechnology Co. Ltd. (Guangzhou, China). All raw transcriptome data are accessible through Genome Sequence Archive (GSA) database.
To analyze lncRNAs and mRNAs, we first processed the sequenced reads using fastp (version 0.18.0). This step involved removing low-quality reads to ensure the retention of high-quality data. Subsequently, Clean reads were then compared to the ribosome database using Bowtie2 (version 2.2.8), retaining unmapped reads for later transcriptome analysis. Additionally, paired-end clean reads were matched to the mouse genome version release 112 from the Ensembl database with HISAT2 software20 (version 2.1.0), and each measured lncRNA was annotated according to the corresponding genome annotation file. Transcriptome data are accessible through Genome Sequence Archive (GSA) database (accession numbers CRA020805).
RNA Sequencing Data Analysis
Differentially expressed genes between the control groups and the AC model groups were identified using differential expression analysis for sequence count data 2 (DEseq2) software. Genes were screened based on fold change and significance testing results. The criteria for selection included a fold change ≥ 1.5 and a p-value < 0.05. Partial bioinformatic analysis was performed using Omicsmart, a real-time interactive online platform for data analysis (http://www.omicsmart.com).
Functional Enrichment Analysis
To investigate the mechanisms underlying the differentially expressed lncRNAs and mRNAs in AC, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. These analyses aimed to predict potential signal transduction pathways, biological functions, and biochemical metabolic pathways associated with the differentially expressed RNA. The GO analysis encompasses three ontologies: biological process (BP), cellular component (CC), and molecular function (MF). This framework allows for the annotation of differentially expressed RNAs and facilitates the assessment of their functional significance. A hypergeometric test with a corrected p-value of < 0.05 was considered to indicate significantly enriched GO terms and KEGG pathways. Gene Ontology (http://www.geneontology.org/), the KEGG database (http://www.genome.jp/kegg).
Gene Set Enrichment Analysis (GSEA) is a method used to analyze gene expression data through a gene set-based approach. To investigate the differences in biological processes between the control groups and the AC model groups, Gene Set Enrichment Analysis was performed utilizing the GSEA database (http://www.gsea-msigdb.org).
The PPI network of differentially expressed mRNAs was constructed using the STRING database (https://string-db.org). Interactions with a validation score greater than 0.4 were considered significant. The resulting networks were visualized using Cytoscape software (version 3.7.2).
Construction of lncRNA-mRNA Network
To investigate the latent functions of differentially expressed lncRNAs and their interactions with mRNAs, we constructed a co-expression network of lncRNA–mRNA transcripts. We calculated the Pearson correlation coefficient (PCC) to evaluate the correlation between these lncRNAs and mRNAs. The resulting co-expression network, which highlights these significant correlation pairs, was visualized using Cytoscape software (version 3.7.2).
Protein Expression in the Human Protein Atlas
Protein expression data for genes were obtained from the Human Protein Atlas (HPA, https://www.proteinatlas.org). The Institutional Review Board of China-Japan Union Hospital of Jilin University (IRB Exemption No. 2025-KYLL-003) confirmed this study’s exemption from ethical review, as it involved only secondary analysis of anonymized data per China’s ethical guidelines and institutional policies. Immunohistochemical (IHC) data were used to determine the localization of these proteins in tissues such as the lung, small intestine, cervix, and eye under normal physiological conditions. IHC protocols in the HPA involve antibody-based staining of tissue microarrays, ensuring high reproducibility. Additionally, single-cell RNA sequencing (scRNA-seq) data from the HPA were analyzed to assess gene expression in specific cell types, focusing on mucosal surfaces, particularly the lung and eye. This integrated approach highlighted the role of these molecules in mucosal immunity.
RT-qPCR
The SYBR green RT-qPCR assay was used to confirm the differentially expressed lncRNAs and mRNAs identified by transcriptome sequencing. Total RNA isolated from the tissue samples described above was used for reverse transcription and PCR. RT-qPCR was performed using the SYBR green assay kit (TaKaRa Biotechnology, Dalian, China) and a QuantStudio 3 Real-Time PCR System (Thermo Fisher, Waltham, MA, United States). The specific primers used for RT-qPCR are listed in Supplementary Table S1. The RT-qPCR program was as follows: denaturation at 95◦C for 30s, followed by 40 cycles at 95◦C for 5s, and 60◦C for 34s. Each sample was run in triplicate. GAPDH was the internal reference of lncRNA and mRNA. The relative quantitative analysis of the data was performed by 2−ΔΔCt method.
Statistical Analysis
Data were expressed as the mean ± standard deviation. The fold change value and P-value were used to evaluate the differentially expressed LncRNAs and mRNAs. The significance of the difference of the expression levels of LncRNAs and mRNAs between the Control and AC groups was estimated using the Student’s t-test. The false discovery rate (FDR) was calculated to correct the P-value. P<0.05 was regarded as statistically significant.
Results
Establishment and Evaluation of AC in a Murine Model
To establish and evaluate a murine model of AC, we used the method illustrated in Figure 1A. Mice were sensitized on day 0 with 100 μg/100 μL of SRW and alum via footpad injection. Starting on day 10, mice received daily eye drops of 0.5 mg/10 μL SRW and PBS for five days. As shown in Figure 1B, compared to the normal group, the AC model group exhibited significant eyelid edema (white arrows) and mucopurulent discharge (red arrows) from day 10 to day 14. Clinical symptoms were quantified using the scoring criteria in Figure 1C, which includes chemosis, conjunctival redness, lid edema, and tearing/discharge. Figure 1D shows a progressive increase in clinical scores from day 10 to day 14 in the AC model group. Additionally, the scratching frequency, a behavioral indicator of ocular irritation, was significantly higher in the AC model group compared to controls (Figure 1E). The above results confirmed the successful establishment of the AC model.
Comparative Analysis of mRNAs and lncRNAs Characteristics in AC
The distribution of exon numbers between lncRNAs and mRNAs shows a significant difference. lncRNAs predominantly have fewer exons compared to mRNAs, while mRNAs display a broader distribution across multiple exon numbers (Figure 2A). The length of lncRNA transcripts is generally shorter compared to mRNAs (Figure 2B). Various lncRNA types are classified into several categories, including sense, antisense, intronic, bidirectional, intergenic, and others LncRNA. The majority of lncRNAs fall into the sense LncRNA category (Figure 2C). Heatmaps exhibit distinct differential expression profiles of mRNAs and lncRNAs between control and AC samples (Figure 2D). The volcano plots, generated by screening with log2 fold change (FC)>1 and p-value<0.05, highlight significantly upregulated and downregulated genes in AC samples (Figure 2E).
Functional Enrichment Analysis of Differentially Expressed Genes
To investigate the biological functions of the differentially expressed genes between health and AC, GO enrichment analysis was performed. As shown in Figure 3A, upregulated genes in terms of biological process are involved in response to stimulus, metabolic processes, signaling, developmental processes, and immune system processes. For molecular function, binding is prominently represented, with enrichment also seen in catalytic activity and molecular transducer activity. In terms of cellular component, cellular anatomical entities and protein-containing complexes are enriched. As shown in Figure 3B, downregulated genes in terms of biological process are primarily associated with response to stimulus, metabolic process, signaling, positive regulation of biological process and immune system process. For molecular function, binding is the most significantly enriched category. In terms of cellular component, cellular anatomical entities and protein-containing complexes are also enriched.
KEGG pathway analysis was employed to understand the pathways in which the different expressed genes are involved. As shown in Figure 3C, the upregulated genes show significant enrichment in pathways such as steroid hormone biosynthesis (ko00140), histidine metabolism (ko00340), glycolysis/gluconeogenesis (ko00010), and IL-17 signaling pathway (ko04657), which is associated with inflammation. For the downregulated genes, KEGG pathway analysis indicates significant involvement in pathways associated with cytokine-cytokine receptor interaction (ko04060), hematopoietic cell lineage (ko04640), and inflammatory bowel disease (ko05321), as shown in Figure 3D.
Functional and Pathway Enrichment Analysis of Differentially Expressed lncRNAs in AC
To elucidate the biological functions of differentially expressed lncRNAs in AC, GO enrichment analysis and KEGG pathway analysis were performed. GO enrichment analysis of upregulated lncRNAs revealed significant enrichment in various biological processes, molecular functions, and cellular components. Upregulated lncRNAs are notably involved in response to stimulus, multicellular organismal process, signaling and metabolic process. Binding is prominently represented among molecular functions, and cellular anatomical entity are significantly enriched within cellular components (Figure 4A). Downregulated lncRNAs are primarily associated with response to stimulus, metabolic process, multicellular organismal process and signaling. Molecular functions such as binding and catalytic activity are notably enriched, as well as cellular components including cellular anatomical entity and protein-containing complex (Figure 4B).
KEGG pathway analysis for upregulated lncRNAs highlights significant enrichment in pathways related to pentose and glucuronate interconversions (ko00040), steroid hormone biosynthesis (ko00140), beta-Alanine metabolism (ko00410), and lysosome (ko04142) (Figure 4C). For downregulated lncRNAs, KEGG pathway analysis indicates significant involvement in pathways associated with cytokine-cytokine receptor interaction (ko04060), inflammatory bowel disease (ko05321), hematopoietic cell lineage (ko04640), and the IL-17 signaling pathway (ko04657) (Figure 4D).
Inflammation-Related Signaling Pathways and Key Immune Genes in AC
AC is characterized by significant involvement of the immune system in its pathophysiology. To further analyze the inflammation-related signaling pathways involved in AC, we utilized REACTOME for GSEA, which is effective in identifying pathways based on biological knowledge. As shown in Figure 5A, the GSEA results revealed significant enrichment in several pathways, including immune response in mucosa, xenobiotics metabolism, cell-cell communication, arachidonic acid metabolism, and multiple phosphorylation pathways such as mitogen-activated protein kinase (MAPK), signal transducer and activator of transcription 1 (STAT1), and signal transducer and activator of transcription 2 (STAT2). These changes indicate that AC is intricately linked to inflammatory and immune responses.
In Figure 5B, we aimed to identify key immune-related genes, which were obtained from the IMMPORT database. A Venn diagram showed the intersection of differentially expressed mRNAs and immune-related genes (IRGs), revealing 17 upregulated and 21 downregulated IRGs. Subsequently, Figure 5C highlights the top 20 significantly differentially expressed genes. GO analysis (Figure 5D) of the upregulated IRGs indicated involvement in processes like the innate immune system, Toll-like receptor signaling, and antimicrobial peptide activity. Co-expression analysis (Figure 5E) identified core genes, including Bpifa1, Lcn2, Reg3g, Bpifb3, Bpifb6, Bpifb4 and Pltp.
Mucosal Immunity-Related Genes in AC
Interestingly, the core genes identified are not typical allergy-associated molecules but are primarily involved in mucosal immunity, with direct antimicrobial functions. In Figure 6A, immunohistochemical analysis using the Human Protein Atlas database revealed that Bpifa1, Lcn2, and Reg3g, molecules with direct antimicrobial activity, are expressed under physiological conditions in the mucosa of the lung, small intestine, and cervix, but are not expressed in smooth muscle and the cerebral cortex. Since the conjunctiva is also a mucosal surface, we used single-cell analysis from the Human Protein Atlas to compare the expression of these genes in the lung and eye. As shown in Figure 6B, single-cell analysis indicates that in the lung, these molecules are primarily secreted by club cells and ciliated cells, both types of airway epithelial cells. However, under physiological conditions, Bpifa1, Lcn2, and Reg3g are not expressed by any cell types in the eye.
To explore the relationship between these antimicrobial molecules and LncRNAs, we conducted a co-expression analysis, as shown in Figure 6C. To assess the expression of these molecules in the conjunctiva in response to AC, we performed PCR analysis on conjunctival tissue from AC mice. The result showed that antimicrobial molecules such as Bpifa1, Bpifb3, Bpifb4, Bpifb6, and Reg3g are upregulated in the AC group compared to the control group (Figure 6D). PCR validation revealed that Stoml3-202, Etohd2-205, D3Ertd751e-209, Myrf-206, and Eps15l1-206 were significantly upregulated in AC (Figure 6E). This suggests that these LncRNAs may contribute to the ocular immune response in AC by regulating the expression of antimicrobial molecules.
Discussion
AC is a common clinical manifestation of allergy-related immune dysregulation.21 While allergic mechanisms have been extensively studied, few investigations have addressed the unique immunological features of the ocular mucosal microenvironment, highlighting the need to better understand its specific molecular processes. In this study, we established a murine model of seasonal AC using ragweed pollen, which closely mimics the allergic responses observed in human AC.22 This model allowed us to perform comprehensive transcriptomic analyses to explore the roles of mRNAs and LncRNAs in the pathogenesis of AC.23 Our findings clarify the immune and inflammatory pathways in AC and identify key genes and LncRNAs associated with mucosal immunity that may regulate conjunctival immune responses.
The transcriptomic analysis revealed distinct differences in the expression of both mRNAs and LncRNAs between the control and AC groups. GO analysis indicated that both upregulated and downregulated genes in AC are involved in immune system processes. KEGG analysis showed that pathways such as steroid hormone biosynthesis, histidine metabolism, and the immune-related IL-17 signaling pathway were upregulated in AC. Among these, the Steroid hormone biosynthesis pathway may play a protective role in mucosal immunity during allergic reactions. Allergic responses can lead to the activation of local immune cells, which, in turn, increase cortisol synthesis.24 Cortisol modulates immune responses through negative feedback, potentially alleviating excessive inflammation in the conjunctiva.25 This could act as a protective mechanism against ocular tissue damage.26 Another key pathway identified is histidine metabolism. Histidine is a precursor of histamine, a critical bioactive molecule mediating allergic responses.27,28 Thus, increased histidine metabolism following AC induction may intensify allergic symptoms by enhancing histamine production.
In addition, we found it intriguing that the IL-17 signaling pathway was upregulated following AC. IL-17 is typically elevated in bacterial infections, as it plays a critical role in activating neutrophils, enhancing barrier functions, and promoting antimicrobial immune responses.29–31 However, recent studies have indicated that IL-17 levels also tend to rise in certain allergic conditions, such as asthma and atopic dermatitis, suggesting its potential involvement in allergic responses.32–34 This upregulation of IL-17 may indicate an attempt by the immune system to bolster local barrier defenses in response to compromised mucosal integrity in allergic patients, potentially reducing susceptibility to secondary infections.
In addition to the changes in mRNA expression, we also identified a large number of differentially expressed LncRNAs. LncRNAs play a crucial role in regulating gene expression at both transcriptional and post-transcriptional levels. The results of the GO analysis indicated that these differentially expressed LncRNAs are involved in immune system-related biological processes, suggesting that they may play an important role in immune regulation in AC.
To further explore the potential functions of these LncRNAs, we conducted KEGG pathway analysis, which revealed that the differentially expressed LncRNAs are associated with steroid hormone biosynthesis and the IL-17 signaling pathway. Interestingly, these pathways overlap with those identified in the mRNA KEGG analysis, suggesting that both LncRNAs and mRNAs may play crucial roles in the development of allergic conjunctivitis (AC). This consistency implies that LncRNAs and mRNAs may be involved in the same or closely related physiological processes, where LncRNAs potentially regulate gene expression associated with mRNA, influencing the activity of key biological pathways and processes.
Given the pivotal roles of these pathways, it is likely that LncRNAs play a significant role in modulating immune responses and pathological processes in AC, possibly in collaboration with mRNAs. This insight into the molecular mechanisms underlying AC offers new potential therapeutic targets, particularly for LncRNAs that are closely related to immune and inflammatory pathways. Considering that current treatments for AC, such as corticosteroids and antihistamines, may have associated side effects, further exploration of LncRNA’s role in immune regulation could lead to more precise and targeted therapies. By targeting LncRNAs, it may be possible to develop therapeutic strategies that reduce inflammation and improve immune modulation, offering a potentially safer and more effective approach to managing AC.
GSEA is a method used to identify significant enrichment of gene sets involved in specific biological functions. In the context of AC, we applied GSEA to explore immune-related pathways altered during the allergic response. Following AC induction, genes were significantly enriched in several immune-related pathways, including MAPK, STAT1, and STAT2 pathways.35,36 The enrichment of these pathways suggests increased activation of inflammatory signaling and allergic responses. The MAPK pathway is known to play a crucial role in mediating inflammatory responses, while STAT1 and STAT2 are key players in cytokine signaling, potentially indicating enhanced immune activation and inflammation following allergen exposure in AC.37–39 These findings imply that allergic inflammation in AC involves coordinated activation of multiple pro-inflammatory signaling networks.
Further analysis of immune-related differentially expressed genes in AC revealed an interesting pattern: several key upregulated genes, including Bpifa1, Lcn2, and Reg3g, are primarily recognized for their roles in mucosal immunity and antimicrobial defense, rather than for the classic pro-inflammatory functions typically associated with allergic responses. Bpifa1 is known for its broad expression in respiratory epithelium, where it contributes to innate immune defense against pathogens and helps maintain epithelial barrier integrity.40,41 Lcn2, an iron-binding protein, limits bacterial growth and modulates immune responses, providing protective effects in various mucosal tissues.42,43 Similarly, Reg3g, a C-type lectin, has notable antimicrobial activity, particularly in the gut, where it prevents bacterial invasion and supports mucosal homeostasis.44,45 These findings suggest that mucosal immunity-related genes, traditionally associated with infection defense, may play a novel role in modulating the ocular immune response during AC.
Interestingly, prior research has not reported the expression of these mucosal immunity-related antimicrobial molecules in the context of AC. Our study demonstrates their upregulation in the conjunctiva during AC, suggesting that they may play a key role in modulating immune responses at the ocular surface. Given the constant exposure of the conjunctival epithelium to environmental pathogens and allergens, the upregulation of these immune defense molecules could represent a unique mechanism by which the conjunctiva responds to allergic inflammation. This response may help prevent microbial invasion while simultaneously regulating excessive inflammatory responses, thus protecting the ocular tissue from damage.
However, it remains unclear whether these molecules are directly involved in regulating the inflammatory response in AC, in addition to their potential role in antimicrobial protection of the mucosal barrier. The exact mechanisms underlying the upregulation of these molecules during AC induction are still unknown. Therefore, further research is needed to elucidate the precise functions of these molecules in AC, as well as the mechanisms by which they modulate the severity of allergic inflammation in the conjunctiva.
Conclusions
In conclusion, this study identifies key mRNAs and LncRNAs that are differentially expressed in the conjunctiva of a murine model of AC. These molecules are associated with immune modulation and mucosal defense mechanisms, and may play important roles in regulating conjunctival immune responses. Notably, we found that several mucosal immunity-related molecules, such as Bpifa1 and Reg3g, were upregulated in AC, suggesting that they may contribute to the adaptive immune response at the ocular surface. Additionally, the involvement of LncRNAs in regulating immune-related genes highlights their potential role in immune responses in AC. However, several limitations exist in this study. First, validation in human conjunctival samples is necessary to assess the clinical relevance of these findings. Second, additional experimental studies are required to further elucidate the exact mechanisms by which these mucosal immunity-related genes and LncRNAs contribute to the pathogenesis of AC. Future research will be crucial to better understand their roles in ocular immune responses and their potential implications for the treatment of AC.
Data Sharing Statement
The original data presented in this study are publicly available and can be found at: https://ngdc.cncb.ac.cn/gsa/browse/CRA020805.
Acknowledgments
We thank Kidio Biotechnology Co., Ltd. (Guangzhou, China) for high-throughput whole-transcriptome sequencing and further bioinformatics analysis.
Funding
There is no funding to report.
Disclosure
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
References
1. Leonardi A, Quintieri L, Presa IJ, et al. Allergic conjunctivitis management: update on ophthalmic solutions. Curr Allergy Asthma Rep. 2024;24(7):347–360. doi:10.1007/s11882-024-01150-0
2. Patel DS, Arunakirinathan M, Stuart A, et al. Allergic eye disease. BMJ. 2017;359:j4706.
3. Miyazaki D, Fukagawa K, Okamoto S, et al. Epidemiological aspects of allergic conjunctivitis. Allergol Int. 2020;69(4):487–495. doi:10.1016/j.alit.2020.06.004
4. Elieh Ali Komi D, Rambasek T, Bielory L. Clinical implications of mast cell involvement in allergic conjunctivitis. Allergy. 2018;73(3):528–539. doi:10.1111/all.13334
5. Manzouri B, Flynn T, Ohbayashi M, et al. The dendritic cell in allergic conjunctivitis. Ocul Surf. 2008;6(2):70–78. doi:10.1016/s1542-0124(12)70270-7
6. Ono SJ, Abelson MB. Allergic conjunctivitis: update on pathophysiology and prospects for future treatment. J Allergy Clin Immunol. 2005;115(1):118–122. doi:10.1016/j.jaci.2004.10.042
7. Li J, Bao X, Guo S, et al. Cell death pathways in dry eye disease: insights into ocular surface inflammation. Ocul Surf. 2024;34:535–544. doi:10.1016/j.jtos.2024.11.004
8. Baudouin C, Kolko M, Melik-Parsadaniantz S, et al. Inflammation in Glaucoma: from the back to the front of the eye, and beyond. Prog Retin Eye Res. 2021;83:100916. doi:10.1016/j.preteyeres.2020.100916
9. Dua HS, Donoso LA, Laibson PR. Conjunctival instillation of retinal antigens induces tolerance does it invoke mucosal tolerance mediated via conjunctiva associated lymphoid tissues (CALT)? Ocul Immunol Inflamm. 1994;2(1):29–36. doi:10.3109/09273949409057799
10. Niederkorn JY. The eye sees eye to eye with the immune system: the 2019 proctor lecture. Invest Ophthalmol Vis Sci. 2019;60(13):4489–4495. doi:10.1167/iovs.19-28632
11. Foulsham W, Marmalidou A, Amouzegar A, et al. Review: the function of regulatory T cells at the ocular surface. Ocul Surf. 2017;15(4):652–659. doi:10.1016/j.jtos.2017.05.013
12. Galletti JG, Guzman M, Giordano MN. Mucosal immune tolerance at the ocular surface in health and disease. Immunology. 2017;150(4):397–407. doi:10.1111/imm.12716
13. Dalpke A, Zimmermann S, Heeg K. CpG DNA in the prevention and treatment of infections. BioDrugs. 2002;16(6):419–431. doi:10.2165/00063030-200216060-00003
14. Mariathasan S, Monack DM. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007;7(1):31–40. doi:10.1038/nri1997
15. Dong X, Limjunyawong N, Sypek EI, et al. Keratinocyte-derived defensins activate neutrophil-specific receptors Mrgpra2a/b to prevent skin dysbiosis and bacterial infection. Immunity. 2022;55(9):1645–1662e1647. doi:10.1016/j.immuni.2022.06.021
16. Ueta M, Kinoshita S. Ocular surface inflammation is regulated by innate immunity. Prog Retin Eye Res. 2012;31(6):551–575. doi:10.1016/j.preteyeres.2012.05.003
17. Guidi R, Wedeles CJ, Wilson MS. ncRNAs in type-2 immunity. Noncoding RNA. 2020;6(1):10. doi:10.3390/ncrna6010010
18. Yi Q, Feng J, Lan W, et al. CircRNA and lncRNA-encoded peptide in diseases, an update review. mol Cancer. 2024;23(1):214. doi:10.1186/s12943-024-02131-7
19. Li D, Ou Q, Shen Q, et al. Subconjunctival injection of human umbilical cord mesenchymal stem cells alleviates experimental allergic conjunctivitis via regulating T cell response. Stem Cell Res Ther. 2023;14(1):281. doi:10.1186/s13287-023-03484-4
20. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12(4):357–360. doi:10.1038/nmeth.3317
21. Upaphong P, Thonusin C, Wanichthanaolan O, et al. Consequences of exposure to particulate matter on the ocular surface: mechanistic insights from cellular mechanisms to epidemiological findings. Environ Pollut. 2024;345:123488. doi:10.1016/j.envpol.2024.123488
22. Alexander M, Patel P, Allegro S, et al. Supplementation of fexofenadine therapy with nedocromil sodium 2% ophthalmic solution to treat ocular symptoms of seasonal allergic conjunctivitis. Clin Exp Ophthalmol. 2003;31(3):206–212. doi:10.1046/j.1442-9071.2003.00646.x
23. Chen X, Deng R, Chi W, et al. IL-27 signaling deficiency develops Th17-enhanced Th2-dominant inflammation in murine allergic conjunctivitis model. Allergy. 2019;74(5):910–921. doi:10.1111/all.13691
24. Takei K, Denda S, Kumamoto J, et al. Low environmental humidity induces synthesis and release of cortisol in an epidermal organotypic culture system. Exp Dermatol. 2013;22(10):662–664. doi:10.1111/exd.12224
25. Kostadinova F, Schwaderer J, Sebeo V, et al. Why does the gut synthesize glucocorticoids? Ann Med. 2014;46(7):490–497. doi:10.3109/07853890.2014.932920
26. Susarla R, Liu L, Walker EA, et al. Cortisol biosynthesis in the human ocular surface innate immune response. PLoS One. 2014;9(4):e94913. doi:10.1371/journal.pone.0094913
27. Hu J, Gao J, Wang C, et al. FLI1 regulates histamine decarboxylase expression to control inflammation signaling and leukemia progression. J Inflamm Res. 2023;16:2007–2020.
28. Yang TB, Kim BS. Pruritus in allergy and immunology. J Allergy Clin Immunol. 2019;144(2):353–360. doi:10.1016/j.jaci.2019.06.016
29. Blaschitz C, Raffatellu M. Th17 cytokines and the gut mucosal barrier. J Clin Immunol. 2010;30(2):196–203.
30. Abusleme L, Moutsopoulos NM. IL-17: overview and role in oral immunity and microbiome. Oral Dis. 2017;23(7):854–865.
31. Wiskott L, von der Malsburg C. Recognizing faces by dynamic link matching. Neuroimage. 1996;4(3 Pt 2):S14–18. doi:10.1006/nimg.1996.0043
32. Metz M, Maurer M. Innate immunity and allergy in the skin. Curr Opin Immunol. 2009;21(6):687–693. doi:10.1016/j.coi.2009.09.009
33. Berin MC, Lozano-Ojalvo D, Agashe C, et al. Acute FPIES reactions are associated with an IL-17 inflammatory signature. J Allergy Clin Immunol. 2021;148(3):895–901e896. doi:10.1016/j.jaci.2021.04.012
34. Pablo-Torres C, Izquierdo E, Tan TJ, et al. Deciphering the role of platelets in severe allergy by an integrative omics approach. Allergy. 2023;78(5):1319–1332. doi:10.1111/all.15621
35. Huang IH, Chung WH, Wu PC, et al. JAK-STAT signaling pathway in the pathogenesis of atopic dermatitis: an updated review. Front Immunol. 2022;13:1068260.
36. Pfitzner E, Kliem S, Baus D, et al. The role of STATs in inflammation and inflammatory diseases. Curr Pharm Des. 2004;10(23):2839–2850. doi:10.2174/1381612043383638
37. Galganska H, Jarmuszkiewicz W, Galganski L. Carbon dioxide and MAPK signalling: towards therapy for inflammation. Cell Commun Signal. 2023;21(1):280. doi:10.1186/s12964-023-01306-x
38. Clark DN, Begg LR, Filiano AJ. Unique aspects of IFN-gamma/STAT1 signaling in neurons. Immunol Rev. 2022;311(1):187–204. doi:10.1111/imr.13092
39. Lee CJ, An HJ, Cho ES, et al. Stat2 stability regulation: an intersection between immunity and carcinogenesis. Exp Mol Med. 2020;52(9):1526–1536. doi:10.1038/s12276-020-00506-6
40. Akram KM, Moyo NA, Leeming GH, et al. An innate defense peptide BPIFA1/SPLUNC1 restricts influenza A virus infection. Mucosal Immunol. 2018;11(1):71–81. doi:10.1038/mi.2017.45
41. Tsou YA, Tung MC, Alexander KA, et al. The role of BPIFA1 in upper airway microbial infections and correlated diseases. Biomed Res Int. 2018;2018:2021890. doi:10.1155/2018/2021890
42. Steigedal M, Marstad A, Haug M, et al. Lipocalin 2 imparts selective pressure on bacterial growth in the bladder and is elevated in women with urinary tract infection. J Immunol. 2014;193(12):6081–6089. doi:10.4049/jimmunol.1401528
43. Asaf S, Maqsood F, Jalil J, et al. Lipocalin 2-not only a biomarker: a study of current literature and systematic findings of ongoing clinical trials. Immunol Res. 2023;71(3):287–313. doi:10.1007/s12026-022-09352-2
44. Shin JH, Bozadjieva-Kramer N, Shao Y, et al. The gut peptide Reg3g links the small intestine microbiome to the regulation of energy balance, glucose levels, and gut function. Cell Metab. 2022;34(11):1765–1778e1766.
45. Greenhill C. REG3G - a potential link between the intestinal microbiome and host physiology. Nat Rev Endocrinol. 2023;19(1):5.
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.