Clinical multi-omics reveals the role of Tuomin Zhiti Decoction Intervention in Allergic Rhinitis from the perspective of biological network ============================================================================================================================================ * Weibo Zhao * Boyang Wang * Lingyao Kong * Qi Wang * Shao Li ## Abstract **Background** Increasing evidence showed that seasonal allergic rhinitis (SAR), as an allergy disease, could be alleviated with traditional Chinese medicine (TCM) formula, one example being Tuomin Zhiti Decoction (TZD). However, as a complex composition of TCM herbs, the mechanism of TZD in the treatment of SAR remain unclear. **Purpose** Uncover the mechanism of TZD for treating SAR based on computational analysis and clinical multi-omics experiments. **Study design** Integrate computational analysis including network target analysis and machine learning algorithms with clinical multi-omics experiments and public omics data. **Methods** By analyzing TZD’s composition through a network-based method, we identified the biological effects of each compound, constructing a comprehensive biological network to elucidate TZD’s molecular and pathway mechanisms against AR. Single-arm clinical trials on the gut microbiome and serum transcriptomics corroborated our computational insights. Further validation through public omics data highlighted key TZD compounds, paving the way for future research. **Results** TZD was discovered to exert a regulatory effect on various modules associated with AR, as demonstrated by the constructed biological network. Insights from gut microbiome and serum transcriptomics in clinical trials, where immune-related microbiomes represented by Prevotella, as well as pathways and biological processes including antigen processing and presentation, activation and regulating immune cell surface receptor, were markedly enriched (*P* value < 0.05), indicated that TZD played a pivotal role in modulating immune processes and the immune cells against AR. With the verification of multi-omics, it was determined that TZD potentially influences immune responses and downstream immune cells like CD4+ T cells, through both direct mechanisms involving antigen and indirect mechanisms mediated by gut microbiomes in the treatment of AR. **Conclusion** Through combination of computational prediction and analysis together with clinical multi-omics, a network target based framework provided a new insight for uncovering the mechanism of TZD for treating AR. Key words * seasonal allergic rhinitis * Tuomin Zhiti Decoction * traditional Chinese medicine * network target * multi-omics ## Introduction Allergic Rhinitis (AR) is an inflammatory disease of the nasal mucosa, mediated by allergen-specific Immunoglobulin E (IgE). Its clinical manifestations include sneezing, nasal itching, runny nose, and nasal congestion1. Severe cases can lead to olfactory dysfunction and various complications. Epidemiological studies have revealed a global incidence rate of approximately 40% for allergic rhinitis, with a particularly high prevalence rate of 19.1% in North China among Chinese adults2. These findings indicate that allergic rhinitis is not only a widespread issue but also a growing medical concern. While it may not be life-threatening, the recurrent symptoms and associated complications significantly impact patients’ quality of life and mental well-being. Given its high incidence and detrimental effects on patients, allergic rhinitis is increasingly recognized as a global health problem1,2. The common symptom-controlling treatments include nasal corticosteroids, antihistamine, and allergen immunotherapy3. These treatments, while recommended by the official guidelines, may not be an optimal solution for AR patients in the long term1.In recent years, more and more patients are looking for complementary and alternative medicines to treat AR, such as Chinese herbal medicine4. A growing number of studies have produced promising results on the curative effect of traditional Chinese medicine (TCM) treatment on AR, all with relatively few side effects5–8, such as TZD. Tuomin Zhiti Decoction (TZD) is a formula made up of eleven different herbs, including Magnoliae Flos (Xinyi), Xanthii Fructus (Cangerzi), Mume Fructus (Wumei), Cicadae Periostracum (Chantui), Saposhnikoviae Radix (Fangfeng), Ganoderma (Lingzhi), Astragali Radix (Huangqi), Scutellariae Radix (Huangqin), Lilii Bulbus (Baihe), Glycyrrhizae Radix et Rhizoma (Gancao) and Nidus Vespae (Fengfang). One animal studie have demonstrated that TZD could decrease the AR-induced damage to the nasal mucosa, and clearly ameliorate nasal symptoms through reducing OVA-specific immunoglobulin E (IgE) and histamine release9. However, TZD’s effect has not been clarified yet. As a combination of network science, artificial intelligence and multi-omics, network target was proposed by Li10–12 to uncover the complex role of TCM in the treatment of diseases. From the perspective of systemic biology and medicine13,14, network target has been continuously developing and succeeded in many aspects of TCM researches, including revealing mechanism of TCM15, discovering biomarkers16 and determining efficacy compounds17,18. Therefore, this study evaluates the clinical efficacy and safety of TZD in patients with AR, aims to uncover the mechanism of TZD against AR and provides high-quality evidence to support its clinical application on the basis of network target analysis and clinical multi-omics. ## Methods and Materials ### Prediction of biological effect profiles of TZD In order to estimate the molecular level function of TZD and its compositions, DrugCIPHER algorithm19 was performed to make prediction on genome-wide biological effect profiles for each compound in TZD. In addition, the compounds recorded in each herb of TZD was obtained from HERB database20. Then, a statistical strategy proposed in previous study21 was used to calculate the holistic targets of every herb in TZD, as well as TZD itself. Holistic targets with adjust *P* value less than 0.05 (Benjamini-Hochberg adjustment) were remained for further analysis. ### Enrichment analysis of AR and TZD Molecules related to AR were collected in two ways, including curated ones from CTD database22 and ones predicted by a text-mining algorithm Gendoo 23. Furthermore, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analysis was performed based on the biomolecules related to AR as well as holistic targets of herbs in TZD and biological effect profiles of compounds in TZD. KEGG pathways and GO terms with adjust *P* value less than 0.05 were kept for further use. In addition, KEGG pathways were classified into different categories based on the class and sub class pre-defined. ### Modular network construction of TZD against AR In order to depict the modular regulatory role of TZD against AR, modular biological network was constructed based on the molecular level and pathway level prediction results. Significantly enriched pathways and biological processes were classed into five modules depending on their roles in AR, including Allergic Reaction and Inflammatory Response, T Cell Immunity and Differentiation, Immune Cell Mobility and Response, Phagocytosis and Antigen Presentation, as well as Cell Signaling and Communication. Interactions between herbs and targets, pathways or biological processes, were based on the predicted holistic targets and the enrichment result, respectively. ### Clinical study and Trials Design This study conducted a 12-week, single-arm clinical trial involving 80 patients with allergic rhinitis (AR) who received TuominZhiti (TZD) intervention. The study adhered to the principles of the Declaration of Helsinki, obtained approval from the Ethics Committee of Shanghai Dongfang Hospital (No.), and was registered in the Chinese Clinical Trial Registry (ChiCTR) under registration number ChiCTR-IOR-1. Prior to enrollment, all participants provided informed consent. Clinicians recorded all clinical data, while efficacy and safety were evaluated through relevant clinical phenotypes measured and analyzed by the Clinical Laboratory of BUCM at baseline and after 12 weeks of intervention. Blood samples from each participant were collected following standard procedures and stored at -80℃ within two hours for subsequent multi-omics detection. ### Detection of intestinal flora structure and abundance by 16SrRNA gene sequencing Bacterial DNA extraction from fecal samples: fecal samples were suspended in sterile 0.1moL/L sodium phosphate buffer (PBS), vortexed for 30 min, and centrifuged at 200g for 3 times, 5min each time, to remove coarse particles and collect the supernatant; centrifuged at 9000g for 3min, collected the precipitate, washed, resuspended, and stored at -70°C for subsequent DNA extraction. Bronchial lavage fluid was centrifuged, precipitated, resuspended, and stored for DNA extraction. Total DNA was extracted by Bead beating method. All DNA samples were stored at -20°C for subsequent PCR analysis. ## Results ### Biological effect profiles of TZD To uncover the mechanism of TZD in the treatment of AR and build the macro-micro correlation models to construct relationship between phenotypes and genotypes, based on our previous algorithm DrugCIPHER19, the biological effect profiles of each compound belonging to herbs in TZD that recorded in HERB database20 were predicted and processed. With the validation of PubMed, the biological effect profiles of representative compounds in TZD were found to be highly covered by literature reports (Figure 1A). Furthermore, we predicted the holistic targets of herbs in TZD according to a statistical model proposed in our previous study21, as well as the holistic targets of TZD. It could be found that different herbs in TZD shared some common compounds as well as common targets (Figure 1B, C). These chemical composition and computational prediction results to some extent demonstrate the potential synergistic effects among various herbs in the intervention of AR by TZD, which aligns with the TCM theory “Jun-Chen-Zuo-Shi” (JCZS). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/13/2024.03.10.24303911/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2024/03/13/2024.03.10.24303911/F1) Figure 1. (A) Bar plot showing the literature cover rate of predicted biological effect profiles of some representative compounds in herbs of TZD. (B) and (C) Upset graphs showing shared compounds or predicted targets of different herbs of TZD, respectively. (D) Dot plot showing enriched pathways of collected molecules related to AR and their classification. (E) Network of top ranked pathways enriched by collected molecules related to AR, corresponding molecules in these pathways and their relationships. ### Network target analysis of TZD against AR In order to depict the potential intervention effects of TZD in treating AR, we collected AR-related molecules from CTD database22 as well as a disease-gene predicting method Gendoo23, and conducted analyses based on these molecules. Pathways related to these molecules were primarily concentrated in Organismal Systems module dominated by the immune system and Environmental Information Processing module represented by Signaling Transduction. In addition, some pathways related to cellular processes were also significantly enriched (Figure 1D, E). Certain molecules in these pathways were reported to play important role in AR. For example, IL-4, IFN-γ, and TNF-α were found to increase mucosal permeability and blocking IL-4 helped maintain the integrity of the mucosal barrier and prevented the reduction of tight junctions24. Cytokines such as IL-1 and TNF-αare commonly present in the body’s inflamed areas and these mediators play a key role in inflammation by recruiting additional inflammatory cells, triggering the release of more inflammatory agents, and activating afferent nerves, thereby amplifying the inflammatory response25. Type 2 cytokines (IL-4, IL-5, and IL-13) along with innate cytokines (IL-25, IL-33, and TSLP) were also recognized as significant biomarkers, indicating their crucial role in various allergic biological processes and allergic diseases26. With the combination of pathology of AR and potential effect of TZD, it was found that the holistic targets of TZD fell in five classification including Metabolism, Genetic Information Processing, Environmental Information Processing, Cellular Processing and Organismal Systems (Figure 2A). Among them, the regulation of TZD on sub classifications Signal Transduction and Immune System might play the most critical role in the treatment of AR, as the holistic targets of TZD located mostly on pathways related to these sub classifications. Focusing on the pathway level, we discovered that TZD exerts its therapeutic effect on AR through a modular regulation mechanism, including Allergic Reaction and Inflammatory Response, T Cell Immunity and Differentiation, Immune Cell Mobility and Response, Phagocytosis and Antigen Presentation, as well as Cell Signaling and Communication (Figure 2B). In these modules, pathways like Th1 and Th2 cell differentiation were reported to play dominant roles in AR27,28, which also served as important intervention targets in the treatment of AR. A shift Th1/Th2 balance towards IL-4 (Th2) has been linked to the onset of allergic and atopic diseases, like AR29. In addition, expression of Fc epsilon RI is recognized not only in the context of atopic dermatitis but is broadly associated with various allergic diseases, indicating its key role in the immune response related to allergies30. These pathways were also of vital importance in the targets network (Figure 2C, D). Besides, biological processes associated with IFN-γ were reported closely to allergic diseases represented by AR31–33, as well as those related to cell adhesion34–36 and T cell37–39, which aligned with their significant positions within the multi-layered biological networks (Figure 2E, F). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/13/2024.03.10.24303911/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2024/03/13/2024.03.10.24303911/F2) Figure 2. (A) Bar plot of the distributions of holistic targets of TZD in the treatment of AR categorized in different classifications and sub classifications. (B) 3D dot plot of pathways or biological processes representing modular mechanism of TZD against AR and their assigned modules. (C) Molecule-pathway relationship network showing associations between certain holistic targets of TZD and pathways in modular mechanism of TZD against AR. (D) Pathway-pathway network showing the associations between pathways in modular mechanism of TZD against AR. (E) Molecule-biological processes relationship network showing associations between certain holistic targets of TZD and biological processes in modular mechanism of TZD against AR. (F) Biological processes-biological processes network showing the associations between biological processes in modular mechanism of TZD against AR. (G) Dot plot showing the enrichment result of pathways or biological processes in modular mechanism of TZD against AR for herbs regarded as Jun, Chen or Zuoshi in TZD. (H) Multi-layer network composed of herbs, molecules, modules and pathways or biological processes showing the network regulatory effect of TZD in the treatment of AR. ### Potential roles of different herbs in TZD As it is widely acknowledged that the composition of Chinese formula follows certain compatibility principles, it is crucial to predict the role of each herb in TZD in the intervention of AR. The enrichment levels of different Chinese herbs in TZD on pathways and biological processes related to the modular mechanism of TZD in intervening AR (Figure 2G). Among these pathways or biological processes and their related pharmacological effects, there were certain reports associating them with herbs in TZD. For example, Jun herb Wu Mei, could significantly decrease the expression level of IL-1740 and formula composed of it were reported to have influence on Th1 cell related cytokines like IFN-γ41 and Th2 cell related cytokines like IL-442. In addition, Ling Zhi had been shown to boost the Th1 immune response, resulting in elevated levels of IFN-γ and IL-243,44 and reduce levels of IL-1β, IL-6, and TNF-α45. And Fang Feng could regulate the release of NO, TNF-α, IL-1β, and IL-6, as well as the density of immune cells like macrophages46. Finally, we constructed a multi-layer biological network representing the effect of TZD against AR (Figure 2H). This network was built on the basis of enrichment results for every herb of TZD in the modular pathways or biological processes. And Jun herbs were predicted to take effect mainly on Allergic Reaction and Inflammatory Response module together with T Cell Immunity and Differentiation module, while Chen herbs potentially affects all the five modules and might exerted a relatively comprehensive regulatory effect. And herbs regarded as Zuoshi, intended to regulate Phagocytosis and Antigen Presentation module and Immune Cell Mobility and Response module according to prediction and analysis. On the molecular level, JCZS herbs played synergistic regulatory roles on these AR-related molecules which acted important jobs in these modules. ### Clinical Trials demonstrating the effectiveness of TZD against AR A 12-week single-arm clinical trial was conducted to evaluate the efficacy of TZD intervention in patients with AR (Figure 3A). In this study, TZD significantly relieved both nasal and eye symptoms of SAR, including running noses, nasal itching, nasal congestion, ocular redness/itching etc. The results confirmed the significant clinical benefits of TZD in alleviating symptoms, including a notable reduction in TNSS, TESS, and IgE levels, as well as a substantial improvement in RQLQ (quality of life) for AR patients (*P* value <0.05; Figure 3B, C). Long-term follow-up revealed that during the second year and in 2022 (one year after intervention), the total score for AR was 5.88, which showed statistical significance compared to pre-intervention scores (*P* value < 0.05). In addition, TZD treatment was found to be safe given that most patients reported no adverse reactions. View this table: [Table1.](http://medrxiv.org/content/early/2024/03/13/2024.03.10.24303911/T1) Table1. Clinical trials of TZD against AR ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/13/2024.03.10.24303911/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2024/03/13/2024.03.10.24303911/F3) Figure 3. (A) Workflow of overall study process with the combination of computational prediction and clinical trials including multi-omics and follow-up. (B) Clinical statistical results of TNSS + TESS before and after TZD intervention and during follow-up. (C) Clinical statistical results of RQLQ before and after TZD intervention and during follow-up. To gain insights into the mechanism of action underlying TZD treatment for AR, we further investigated changes in gut microbiota and serum transcriptomics before and after TZD intervention. In total, gut microbiome metagenomics were conducted on 94 samples and serum transcriptomics were conducted on 10 samples. ### Regulatory effect of TZD intervention on gut microbiome The analysis of gut microbiome metagenomics revealed changes in species relative abundance across various taxonomic levels like phylum and genus. For instance, at the phylum level, the proportion of Firmicutes decreased, while Proteobacteria increased (Figure 4A). And at the genus level, an increase in the proportion of Megamonas and Bacteroides was observed, whereas the proportion of Prevotella_9 and Faecalibacterium decreased after treatment (Figure 4B, C). These shifts indicate significant microbial composition changes due to the treatment. More specifically, detailed statistical analyses were conducted on species with higher abundance in the categories of family, genus, and species (Figure 4D). Significantly changes species like Prevotella and Faecalibacterium were reported to be associated with AR or allergic inflammation47–49, as well as key modules in network target of TZD against AR. ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/13/2024.03.10.24303911/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2024/03/13/2024.03.10.24303911/F4) Figure 4. (A) Bar plot showing the proportion of gut microbiome classified in different phyla before and after the intervention of TZD. (B) Bar plot showing the proportion of gut microbiome classified in different genera before and after the intervention of TZD. (C) Classification tree plot of gut microbiome classified in different genera. (D) Significantly changed gut microbiome classified in different familiae, genera and species from left to right, respectively. (E) Random forest AUC score for gut microbiome classified in different familiae, genera and species from left to right, respectively. (F) Box plot of Shannon index showing Alpha diversity before and after the intervention of TZD. (G) Box plot of unweighted unifrac distance showing Beta diversity before and after the intervention of TZD. (H) Bar plot showing LDA score of significantly changed gut microbiome classified in different genera calculated by LEfSe algorithm. In addition, random forests model trained on the gut microbiomes also showed that species in the categories of family, genus, and species could be appropriate biomarkers for evaluating the efficacy of TZD in the treatment of AR (Figure 4E). Significant differences in the Shannon index of Alpha diversity before and after treatment (*P* value < 0.05) indicate notable changes in microbial abundance and evenness after TZD intervention (Figure 4F). Besides, the Beta diversity, using unweighted UniFrac distances (Figure 4G), revealed significant disparities between the groups before and after TZD intervention (*P* value < 0.001), with a greater variation observed within samples after intervention. Based on LEfSe (LDA Effect Size) analysis, it was found that LDA scores of species like Prevotella were higher in samples before TZD intervention and those of species including Proteobacteria and gamma Proteobacteria were higher in samples after TZD intervention (Figure 4H), which to some extent was consistent with previous statistical results. ### Transcriptomics revealing molecular level changes in serum We collected serum samples from 5 clinical patients before and after the intervention of TZD and conducted transcriptome sequencing. To focus on the transcriptional level changes in serum molecules before and after the intervention of TZD, R package DESeq2 was performed to obtain differential expressed genes (DEGs). Totally, 26 DEGs were filtered (adjust *P* value < 0.05, log2Foldchange ≥ 1 or ≤ -1) with paired statistical test and kept for further analysis (Figure 5A). Further, based on these DEGs, pathway like antigen process and presentation was significantly enriched (Figure 5B), so as some immune response and natural killer (NK) cell related biological processes. With previous findings, these enriched pathways and biological processes to some extent, suggested that the role TZD played in immune process like antigen process and presentation and those related to immune cells could be an important mechanism in the treatment of AR. According to previous studies, T cells acted as an important role in allergic inflammation in AR50–52. Besides, NK cells were also of vital importance in the pathology of AR53–55, like resolving eosinophilia and inflammatory crosstalk to eosinophil. Then, Gene Set Variation Analysis (GSVA)56 was performed to estimate pathway level changes (Figure 5C) and it was found that apart from antigen process and presentation, many T cell related biological processes were significantly enrich, including T cell cytokine production, activation, proliferation and chemotaxis. ![Figure 5.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/13/2024.03.10.24303911/F5.medium.gif) [Figure 5.](http://medrxiv.org/content/early/2024/03/13/2024.03.10.24303911/F5) Figure 5. (A) Volcano plot showing DEGs obtained through statistical calculations by DESeq2, with log2 Foldchange as x axis and -log10 adjust p value as y axis. (B) Bar plot of KEGG and GO enrichment results of DEGs. (C) Heatmap showing GSVA score of different gene sets in samples before and after the intervention of TZD. (D) PLS-DA model demonstrating the separability of transcriptomics in a low-dimensional principal component space before and after the intervention of TZD. (E) Bar plot of the directional VIP score of representative genes calculated from PLS-DA model. (F) GSEA result showing the significantly enriched pathways related to antigen process and T cell (*P* value < 0.05). (G) GSEA result showing the significantly enriched biological process related to previous predicted network target of TZD against AR (*P* value < 0.05). Further, from another perspective, a typical machine learning model, Partial Least Squares Discriminant Analysis (PLS-DA) to make deeper analysis. Based on the contributions of various features to the first two principal components, it was easy to see that samples before and after intervention are divided into two categories (Figure 5D). This meant that determining the contributions of various features to the two main principal components was significant, and the significance lay in the ability to identify features with the greatest differences between groups. On the basis of these findings, Variable Importance in Projection (VIP) score, which measured the contributes of each input features were calculated. And with the positive and negative values of feature loadings calculated by the PLS-DA model, we have assigned directionality to the VIP values, in which positive values with larger absolute value meant larger contributions to after intervention group and vice versa (Figure 5E). Then a ranked gene list was obtained and thus Gene Set Enrichment Analysis (GSEA) was conducted to evaluate pathway level changes from the perspective of machine learning. Last but not least, it was found that pathways like antigen process and presentation, as well as T cell receptor signaling pathway (Figure 5F) got significantly enriched (*P* value <0.05) by the gene list ranked by directional VIP score, while biological processes related to T-helper cells and biomolecules like IL-4 and IFN-γ were also enriched (Figure 5G). ### Directly and indirectly potential effects of TZD on regulating T cell in AR With the combination of 16S rRNA gut microbiome metagenomics, serum transcriptomics and network target analysis, we have found that TZD potentially affected biological processes and pathways related to T cells in multiple ways. Among them, directly potential regulation through pathways represented by antigen process and presentation, together with indirect potential regulation through gut microbiome like Prevotella were focused in this research owing to their significant role (Figure 6A) on the basis of the combination of network target analysis and calculation of omics data. In details, on the one hand, TZD played a regulatory role in gut microbiome, influencing on certain microbiomes represented by Prevotella, which had been reported to have closely association with immune response57,58. Specifically, they were involved in a variety of T cell related biological processes, like T cell differentiation59, regulatory T cells process60,61 and other T cell regulation62,63. On the other hand, in MHCI pathway, TZD potentially affected TAP1/2, CALR, HSP70 and HSP90, as well as IFN γ which played an immune-proteasome role. Then through MHCI, TZD potentially affected T cell receptor signaling of CD8+ T cells and regulate the activation of NK cells. Besides, TZD had a potentially regulatory effect on CIITA/CREB/MHCII and HLA-DM, regulated CD4+ T cells and thus influenced downstream pathways and biological processes, like cytokine production and activation of other immune cells. Taking the relationship between gut microbiome and antigen process into consideration, there have already been some researches revealing their potential association. Research has shown that Prevotella primarily triggers Toll-like receptor 2, leading antigen-presenting cells to produce cytokines that favor Th17 polarization, such as interleukin-23 (IL-23) and IL-164. Besides, enriched Faecalibacterium was observed in human leukocyte antigen (HLA)-B27+ individuals65 and a significant link between the Acinetobacter to Proteobacteria ratio and both the production of IL-13 and the likelihood of IL-13 production following allergen exposure was found66. ![Figure 6.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/13/2024.03.10.24303911/F6.medium.gif) [Figure 6.](http://medrxiv.org/content/early/2024/03/13/2024.03.10.24303911/F6) Figure 6. (A) Potential effects of TZD in antigen process and presentation on biomolecules and pathways, showing direct regulation on T cells through regulating antigen related process and indirect regulation on T cells through regulating gut microbiome. (B) Bar plot showing proportions of immune cells in different patients before and after the intervention of TZD. (C) Inferred proportion of CD4+ T cells before and after the intervention of TZD in AR patients. (D) Dot plot showing the increasing trend of CD4+ T cells and decreasing trend of CD8+ T cells in patients before and after the intervention of TZD. (E) Public validation based bar plot showing fold change and statistical significance of the regulatory effect of predicted key compounds on the expression of biomolecules related to the network target of TZD against AR. (F) Public validation based bar plot showing fold change and statistical significance of the regulatory effect of recorder herbs on the expression of biomolecules related to the network target of TZD against AR. Further focusing on the downstream immune processes affected by TZD intervention, we utilized a deconvolution algorithm, CIBERSORT67, to infer changes in the proportions of immune cells from the serum transcriptomes before and after the intervention of TZD (Figure 6B). It was found that the proportion of CD4+ T cells showed a significantly increase after the intervention (Figure 6C), which to some extent confirmed our findings from the perspective of downstream outcomes. Besides, the proportion of CD8+ T cells and the proportion of CD4+ T cells had a negative relationship in AR patients before and after the intervention of TZD (Figure 6D). In addition, validation of regulation of TZD on antigen process related pathways and biomolecules were conducted based on several public datasets. Firstly, based on ITCM database68, which recorded expression of MCF-7 cell lines after the intervention of compounds from usual herbs, transcriptomics data of certain compounds of herbs in TZD were collected. After estimating the potential effects on antigen process and T-helper cell related biological process, 21 compounds belonging to herbs of TZD were kept for further statistical analysis. And it was found that 9 of them, like Ursolic acid and Oleanolic acid, had significant regulation on the predicted network target of TZD against AR (Figure 6E) under certain cell line conditions. Besides, on the basis of previous study69, the expression omics data of THP-1 cell line after the intervention of certain Jun herbs including Fangfeng and Chantui, as well as Chen herbs like Huangqin, Huangqi and Wuweizi were also collected. And on the predicted network target, they had different regulatory effects (Figure 6F). These public data based validations to some extent confirmed the potential intervention effects of TZD on these predicted targets. It also indicated the presence of synergistic effects involving multiple components and targets, and provided certain evidence for the selection of key compounds of TZD in the treatment of AR as well as the elucidation of JCZS. ## Discussion As an allergic inflammation of the nasal mucosa characterized by symptoms such as nasal blockage, running nose, repeated sneezing, AR could seriously affect the quality of life of patients. TCM intervention has become an important way to effectively prevent and treat AR2. TZD, as a multi-component TCM formula, is an effective method for clinical intervention of AR, and its molecular mechanism needs to be further studied. Through integrating computational prediction, clinical multi-omics and experimental validation, this research uncovered the regulatory role of TZD against AR as well as identifying synergistic effects of herbs and compounds within TZD. In this study, based on network target analysis, we constructed a multi-layer modular network. In this network, where immune-related modules played a dominant role, the potential intervention effect of TZD on AR was characterized. Guided by computational predictions, a single-arm clinical trial was initiated to validate the efficacy and safety of TZD in intervening AR. And the trial confirmed TZD’s effectiveness in alleviating symptoms, reducing IgE levels, and its preventive role against AR recurrence. Alongside the clinical trial, a comprehensive multi-omics clinical study was conducted, encompassing gut microbiome metagenomics and serum transcriptomics. In gut microbiome analysis, it was found that microbiomes like Firmicutes, Prevotella and Faecalibacterium decreased after the intervention of TZD, while the relative abundance increased in Firmicutes, Megamonas and Bacteroides. And in serum transcriptomics, pathways and biological processes related to immune response represented by antigen process and immune cell processes were significantly enriched. Based on the combination of computational analysis and experimental validation, the regulatory effect on immune response of TZD was found on direct effect on antigen process which regulate downstream immune process, as well as indirect regulation through gut microbiomes which has been reported to play regulate role on immune response in allergic diseases and other diseases70–74. Finally, regulatory effects on predicted important biomolecules in antigen process of vital compounds of TZD and herbs of TZD were verified on the basis of public omics data, which validated the potential influence of TZD on antigen process. There are some limitations in our study. First, compounds of TZD were collected from the compositions of herbs recorded in database, which might have certain differences. Fortunately, algorithms applied in network target analysis could make up for the bias to some extent and still characterized the potential effects of TZD with representative compounds in a high precision. Second, our experimental validation has room for further improvement in both depth and breadth. This also represents a point worthy of focus in future research. In conclusion, this work focused on deciphering the complex regulatory mechanisms of TCM against diseases. With the combination of network analysis, clinic trials and multi-omics technologies, we revealed the potential mechanism of TZD in the treatment of AR. ## Data Availability All data produced in the present study are available upon reasonable request to the authors. All data produced in the present work are contained in the manuscript. ## Conflicts of Interest The authors declare that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome. ## Author Contributions W.Z., B.W., Q.W., and S.L. designed the experiments. W.Z. and J.W. recruited patients and provided samples. B.W. and W.Z. performed multi-omics data analysis. The manuscript was drafted by B.W., S.L., W.Z. and critically revised by all the authors and discussed the results and commented on the manuscript by all the authors. ## Acknowledgments This study was supported by the Anhui Province Traditional Chinese Medicine Science and Technology Research Project (202303a07020001) and Special project of the interpretation plan of the principles of traditional Chinese medicine of the State Administration of Traditional Chinese Medicine. ## Abbreviations TESS : Total Eye Symptom Score; TNSS : Total Nasal Symptom Score; TZD : Tuomin-Zhiti-Decoction; AR : Allergic Rhinitis; SAR : Seasonal Allergic Rhinitis; RQLQ : Rhino conjunctivitis Quality of Life Questionnaire; TCM : Traditional Chinese Medicine. * Received March 10, 2024. * Revision received March 10, 2024. * Accepted March 13, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. Bousquet, J. et al. Next-generation Allergic Rhinitis and Its Impact on Asthma (ARIA) guidelines for allergic rhinitis based on Grading of Recommendations Assessment, Development and Evaluation (GRADE) and real-world evidence. Journal of Allergy and Clinical Immunology 145, 70–80.e73, doi:10.1016/j.jaci.2019.06.049 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2019.06.049&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 2. Cheng, L. et al. Chinese society of allergy guidelines for diagnosis and treatment of allergic rhinitis. *Allergy*, Asthma and Immunology Research 10, 300–353, doi:10.4168/aair.2018.10.4.300 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4168/aair.2018.10.4.300&link_type=DOI) 3. Tang, R., Lei, S., Zhu, L., Lv, Y. & Li, H. Prevention of omalizumab for seasonal allergic rhinoconjunctivitis: a retrospective cohort study. Frontiers in Immunology 13, doi:10.3389/fimmu.2022.913424 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2022.913424&link_type=DOI) 4. Zhang, X., Lan, F., Zhang, Y. & Zhang, L. Chinese herbal medicine to treat allergic rhinitis: Evidence from a meta-analysis. *Allergy*, Asthma and Immunology Research 10, 34–42, doi:10.4168/aair.2018.10.1.34 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4168/aair.2018.10.1.34&link_type=DOI) 5. Chan, R. Y. P. & Chien, W. T. The effects of two Chinese herbal medicinal formulae vs. placebo controls for treatment of allergic rhinitis: A randomised controlled trial. Trials 15, doi:10.1186/1745-6215-15-261 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1745-6215-15-261&link_type=DOI) 6. Zhao, J. et al. Efficacy of Bimin decoction for patients with perennial allergic rhinitis: an open-label non-inferiority randomized controlled trial. Trials 20, 802, doi:10.1186/s13063-019-3763-z (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13063-019-3763-z&link_type=DOI) 7. Yang, S. H., Yu, C. L., Chen, Y. L., Chiao, S. L. & Chen, M. L. Traditional Chinese medicine, Xin-yi-san, reduces nasal symptoms of patients with perennial allergic rhinitis by its diverse immunomodulatory effects. International Immunopharmacology 10, 951–958, doi:10.1016/j.intimp.2010.05.008 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.intimp.2010.05.008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20546945&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 8. Luo, Q. et al. Potential effectiveness of Chinese herbal medicine Yu ping feng san for adult allergic rhinitis: A systematic review and meta-analysis of randomized controlled trials. BMC Complementary and Alternative Medicine 17, doi:10.1186/s12906-017-1988-5 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12906-017-1988-5&link_type=DOI) 9. Cheng, J., Zhang, M., Zheng, Y., Wang, J. & Wang, Q. Integrative analysis of network pharmacology and proteomics to identify key targets of Tuomin-Zhiti-Decoction for allergic rhinitis. Journal of Ethnopharmacology 296, doi:10.1016/j.jep.2022.115448 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jep.2022.115448&link_type=DOI) 10. Li, S. & Zhang, B. Traditional Chinese medicine network pharmacology: Theory, methodology and application. Chinese Journal of Natural Medicines 11, 110–120, doi:10.1016/S1875-5364(13)60037-0 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S18755364(13)60037-0&link_type=DOI) 11. Li, S. Network pharmacology evaluation method guidance-draft. World Journal of Traditional Chinese Medicine 7, 146–154 (2021). 12. Li, S. Mapping ancient remedies: applying a network approach to traditional Chinese medicine. Science 350, S72–S74 (2015). 13. Zhang, P. et al. Network pharmacology: towards the artificial intelligence-based precision traditional Chinese medicine. Briefings in bioinformatics 25, doi:10.1093/bib/bbad518 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bib/bbad518&link_type=DOI) 14. 张彦琼 & 李梢. 网络药理学与中医药现代研究的若干进展. 中国药理学与毒理学杂志 29 (2015). 15. Zhou, W. et al. Network pharmacology to unveil the mechanism of Moluodan in the treatment of chronic atrophic gastritis. Phytomedicine 95, doi:10.1016/j.phymed.2021.153837 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.phymed.2021.153837&link_type=DOI) 16. Wang, B. et al. Uncovering the mechanisms of Yi Qi Tong Qiao Pill in the treatment of allergic rhinitis based on Network target analysis. Chinese Medicine (United Kingdom*)* 18, doi:10.1186/s13020-023-00781-1 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13020-023-00781-1&link_type=DOI) 17. Wang, B. et al. Exploring the effect of Weifuchun capsule on the toll-like receptor pathway mediated HES6 and immune regulation against chronic atrophic gastritis. Journal of Ethnopharmacology 303, doi:10.1016/j.jep.2022.115930 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jep.2022.115930&link_type=DOI) 18. Zhang, S. et al. DrugAI: a multi-view deep learning model for predicting drug-target activating/inhibiting mechanisms. Briefings in bioinformatics 24, doi:10.1093/bib/bbac526 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bib/bbac526&link_type=DOI) 19. Zhao, S. & Li, S. Network-based relating pharmacological and genomic spaces for drug target identification. PLoS ONE 5, doi:10.1371/journal.pone.0011764 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0011764&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20668676&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 20. Fang, S. et al. HERB: A high-throughput experiment- And reference-guided database of traditional Chinese medicine. Nucleic Acids Research 49, D1197–D1206, doi:10.1093/nar/gkaa1063 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkaa1063&link_type=DOI) 21. Liang, X., Li, H. & Li, S. A novel network pharmacology approach to analyse traditional herbal formulae: the Liu-Wei-Di-Huang pill as a case study. Molecular bioSystems 10, 1014–1022, doi:10.1039/c3mb70507b (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1039/c3mb70507b&link_type=DOI) 22. Davis, A. P. et al. Comparative Toxicogenomics Database (CTD): update 2023. Nucleic Acids Research 51, D1257–D1262, doi:10.1093/nar/gkac833 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkac833&link_type=DOI) 23. Nakazato, T., Bono, H., Matsuda, H. & Takagi, T. Gendoo: Functional profiling of gene and disease features using MeSH vocabulary. Nucleic Acids Research 37, W166–W169, doi:10.1093/nar/gkp483 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkp483&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19498079&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000267889100030&link_type=ISI) 24. Steelant, B. et al. Histamine and T helper cytokine-driven epithelial barrier dysfunction in allergic rhinitis. The Journal of Allergy and Clinical Immunology 141, doi:10.1016/j.jaci.2017.08.039 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2017.08.039&link_type=DOI) 25. Mohamad, S. A. et al. A novel nasal co-loaded loratadine and sulpiride nanoemulsion with improved downregulation of TNF-α, TGF-β and IL-1 in rabbit models of ovalbumin-induced allergic rhinitis. Drug Delivery 28, 229–239, doi:10.1080/10717544.2021.1872741 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/10717544.2021.1872741&link_type=DOI) 26. Breiteneder, H. et al. Biomarkers for diagnosis and prediction of therapy responses in allergic diseases and asthma. Allergy: European Journal of Allergy and Clinical Immunology 75, 3039–3068, doi:10.1111/all.14582 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/all.14582&link_type=DOI) 27. Dong, J., Xu, O., Wang, J., Shan, C. & Ren, X. Luteolin ameliorates inflammation and Th1/Th2 imbalance via regulating the TLR4/NF-κB pathway in allergic rhinitis rats. Immunopharmacology and Immunotoxicology 43, 319–327, doi:10.1080/08923973.2021.1905659 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/08923973.2021.1905659&link_type=DOI) 28. Ke, X., Chen, Z., Wang, X., Kang, H. & Hong, S. Quercetin improves the imbalance of Th1/Th2 cells and Treg/Th17 cells to attenuate allergic rhinitis. Autoimmunity 56, doi:10.1080/08916934.2023.2189133 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/08916934.2023.2189133&link_type=DOI) 29. Piao, C. H. et al. PM2.5 exposure regulates Th1/Th2/Th17 cytokine production through NF-κB signaling in combined allergic rhinitis and asthma syndrome. International Immunopharmacology 119, doi:10.1016/j.intimp.2023.110254 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.intimp.2023.110254&link_type=DOI) 30. Semper, A. E. et al. Surface expression of FcεRI on Langerhans’ cells of clinically uninvolved skin is associated with disease activity in atopic dermatitis, allergic asthma, and rhinitis. Journal of Allergy and Clinical Immunology 112, 411–419, doi:10.1067/mai.2003.1626 (2003). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1067/mai.2003.1626&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12897750&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000184650600028&link_type=ISI) 31. Kong, Y. et al. SymMap database and TMNP algorithm reveal Huanggui Tongqiao granules for Allergic rhinitis through IFN-mediated neuroimmuno-modulation. Pharmacological Research 185, doi:10.1016/j.phrs.2022.106483 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.phrs.2022.106483&link_type=DOI) 32. Prigione, I. et al. Interferon-gamma and IL-10 may protect from allergic polysensitization in children: Preliminary evidence. Allergy: European Journal of Allergy and Clinical Immunology 65, 740–742, doi:10.1111/j.1398-9995.2009.02285.x (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1398-9995.2009.02285.x&link_type=DOI) 33. Hsu, U.-H. & Chiang, B.-L. γδ T Cells and Allergic Diseases. Clinical Reviews In Allergy & Immunology 65, 172–182, doi:10.1007/s12016-023-08966-0 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s12016-023-08966-0&link_type=DOI) 34. Xie, S. et al. Activated leukocyte cell adhesion molecule as a biomarker for disease severity and efficacy of sublingual immunotherapy in allergic rhinitis. International Immunopharmacology 88, doi:10.1016/j.intimp.2020.106975 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.intimp.2020.106975&link_type=DOI) 35. Liu, C. M., Shun, C. T. & Cheng, Y. K. Soluble adhesion molecules and cytokines in perennial allergic rhinitis. *Annals of Allergy*, Asthma and Immunology 81, 176–180, doi:10.1016/S1081-1206(10)62806-2 (1998). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1081-1206(10)62806-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9723565&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000075532400015&link_type=ISI) 36. Liu, H., Ai, J., Wang, T. & Tan, G. Adhesion Promotes Allergic Rhinitis CD4+IL4+ T Cell Differentiation via ICAM1 and E-Selectin. American Journal of Rhinology and Allergy 36, 521–528, doi:10.1177/19458924221086061 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/19458924221086061&link_type=DOI) 37. Sustiel, A. & Rocklin, R. T cell responses in allergic rhinitis, asthma and atopic dermatitis. Clinical and Experimental Allergy 19, 11–18, doi:10.1111/j.1365-2222.1989.tb02337.x (1989). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1365-2222.1989.tb02337.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2649212&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 38. Yu, X., Wang, M. & Cao, Z. Reduced CD4+T Cell CXCR3 Expression in Patients With Allergic Rhinitis. Frontiers in Immunology 11, doi:10.3389/fimmu.2020.581180 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2020.581180&link_type=DOI) 39. Bellinghausen, I., Khatri, R. & Saloga, J. Current Strategies to Modulate Regulatory T Cell Activity in Allergic Inflammation. Frontiers in Immunology 13, doi:10.3389/fimmu.2022.912529 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2022.912529&link_type=DOI) 40. Liu, Z. et al. An integrated strategy for anti-inflammatory quality markers screening of traditional Chinese herbal medicine Mume Fructus based on phytochemical analysis and anti-colitis activity. Phytomedicine 99, doi:10.1016/j.phymed.2022.154002 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.phymed.2022.154002&link_type=DOI) 41. Lu, C. et al. Herbal components of a novel formula PSORI-CM02 interdependently suppress allograft rejection and induce CD8+CD122+PD-1+ regulatory T cells. Frontiers in Pharmacology 9, doi:10.3389/fphar.2018.00088 (2018). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fphar.2018.00088&link_type=DOI) 42. Wu, D. H. et al. PSORI-CM02 alleviates IMQ-induced mouse dermatitis via differentially regulating pro- and anti-inflammatory cytokines targeting of Th2 specific transcript factor GATA3. Biomedicine and Pharmacotherapy 110, 265–274, doi:10.1016/j.biopha.2018.11.092 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopha.2018.11.092&link_type=DOI) 43. Habijanic, J., Berovic, M., Boh, B., Plankl, M. & Wraber, B. Submerged cultivation of Ganoderma lucidum and the effects of its polysaccharides on the production of human cytokines TNF-α, IL-12, IFN-γ, IL-2, IL-4, IL-10 and IL-17. New Biotechnology 32, 85–95, doi:10.1016/j.nbt.2014.07.007 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.nbt.2014.07.007&link_type=DOI) 44. Ren, L., Zhang, J. & Zhang, T. Immunomodulatory activities of polysaccharides from Ganoderma on immune effector cells. Food Chemistry 340, doi:10.1016/j.foodchem.2020.127933 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.foodchem.2020.127933&link_type=DOI) 45. Chen, X. P. et al. Free radical scavenging of *Ganoderma lucidum* polysaccharides and its effect on antioxidant enzymes and immunity activities in cervical carcinoma rats. CARBOHYDRATE POLYMERS 77, 389–393, doi:10.1016/j.carbpol.2009.01.009 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.carbpol.2009.01.009&link_type=DOI) 46. Fan, H. et al. Structure characterization and immunomodulatory activity of a polysaccharide from Saposhnikoviae Radix. International Journal of Biological Macromolecules 233, doi:10.1016/j.ijbiomac.2023.123502 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ijbiomac.2023.123502&link_type=DOI) 47. Chen, Z. et al. Fecal and serum metabolomic signatures and gut microbiota characteristics of allergic rhinitis mice model. Frontiers in Cellular and Infection Microbiology 13, doi:10.3389/fcimb.2023.1150043 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fcimb.2023.1150043&link_type=DOI) 48. Arrieta, M. C. et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Science Translational Medicine 7, doi:10.1126/scitranslmed.aab2271 (2015). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE0OiI3LzMwNy8zMDdyYTE1MiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAzLzEzLzIwMjQuMDMuMTAuMjQzMDM5MTEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 49. Wu, Y. et al. Tetrahydrocurcumin alleviates allergic airway inflammation in asthmatic mice by modulating the gut microbiota. Food & function 12, 6830–6840, doi:10.1039/d1fo00194a (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1039/d1fo00194a&link_type=DOI) 50. Seumois, G. et al. Single-cell transcriptomic analysis of allergen-specific T cells in allergy and asthma. Science Immunology 5, doi:10.1126/SCIIMMUNOL.ABA6087 (2020). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1126/SCIIMMUNOL.ABA6087&link_type=DOI) 51. Suhrkamp, I., Scheffold, A. & Heine, G. T-cell subsets in allergy and tolerance induction. European Journal of Immunology 53, doi:10.1002/eji.202249983 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/eji.202249983&link_type=DOI) 52. Grydziuszko, E., Phelps, A., Bruton, K., Jordana, M. & Koenig, J. F. E. Heterogeneity, subsets, and plasticity of T follicular helper cells in allergy. Journal of Allergy and Clinical Immunology 150, 990–998, doi:10.1016/j.jaci.2022.08.023 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2022.08.023&link_type=DOI) 53. Sin, B., Misirligil, Z., Aybay, C., Gürbüz, L. & Imir, T. Effect of allergen specific immunotherapy (IT) on natural killer cell activity (NK), IgE, IFN-γ levels and clinical response in patients with allergic rhinitis and asthma. Journal of Investigational Allergology and Clinical Immunology 6, 341–347 (1996). 54. El-Shazly, A. E. & Lefebvre, P. P. Modulation of NK cell autocrine-induced eosinophil chemotaxis by interleukin-15 and vitamin D 3: A possible NK-eosinophil crosstalk via IL-8 in the pathophysiology of allergic rhinitis. Mediators of Inflammation 2011, doi:10.1155/2011/373589 (2011). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1155/2011/373589&link_type=DOI) 55. Pawlak, E. A. et al. Diesel exposure suppresses natural killer cell function and resolution of eosinophil inflammation: A randomized controlled trial of exposure in allergic rhinitics. Particle and Fibre Toxicology 13, doi:10.1186/s12989-016-0135-7 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12989-016-0135-7&link_type=DOI) 56. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC bioinformatics 14, 7, doi:10.1186/1471-2105-14-7 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2105-14-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23323831&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 57. Shi, N., Li, N., Duan, X. & Niu, H. Interaction between the gut microbiome and mucosal immune system. Military Medical Research 4, 14, doi:10.1186/s40779-017-0122-9 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s40779-017-0122-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28465831&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 58. De Filippis, F. et al. Specific gut microbiome signatures and the associated pro-inflamatory functions are linked to pediatric allergy and acquisition of immune tolerance. Nature Communications 12, doi:10.1038/s41467-021-26266-z (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-021-26266-z&link_type=DOI) 59. Fujimura, K. E. et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nature Medicine 22, 1187–1191, doi:10.1038/nm.4176 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.4176&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27618652&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 60. Akagawa, S. & Kaneko, K. Gut microbiota and allergic diseases in children. Allergology International : Official Journal of the Japanese Society of Allergology 71, 301–309, doi:10.1016/j.alit.2022.02.004 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.alit.2022.02.004&link_type=DOI) 61. Bunyavanich, S. & Berin, M. C. Food allergy and the microbiome: Current understandings and future directions. The Journal of Allergy and Clinical Immunology 144, 1468–1477, doi:10.1016/j.jaci.2019.10.019 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jaci.2019.10.019&link_type=DOI) 62. Hegazy, A. N. et al. Circulating and Tissue-Resident CD4+ T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation. Gastroenterology 153, 1320–1337.e1316, doi:10.1053/j.gastro.2017.07.047 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1053/j.gastro.2017.07.047&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 63. Simonyté Sjödin, K., et al. Temporal and long-term gut microbiota variation in allergic disease: A prospective study from infancy to school age. Allergy 74, 176–185, doi:10.1111/all.13485 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/all.13485&link_type=DOI) 64. Larsen, J. M. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology 151, 363–374, doi:10.1111/imm.12760 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/imm.12760&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28542929&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 65. Essex, M. et al. Shared and Distinct Gut Microbiota in Spondyloarthritis, Acute Anterior Uveitis, and Crohn’s Disease. Arthritis and Rheumatology 76, 48–58, doi:10.1002/art.42658 (2024). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/art.42658&link_type=DOI) 66. Turturice, B. A. et al. Perinatal bacterial exposure contributes to IL-13 aeroallergen response. American Journal of Respiratory Cell and Molecular Biology 57, 419–427, doi:10.1165/rcmb.2017-0027 (2017). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1165/rcmb.2017-0027&link_type=DOI) 67. Newman, A. M. et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nature Biotechnology 37, 773–782, doi:10.1038/s41587-019-0114-2 (2019). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41587-019-0114-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31061481&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) 68. Tian, S. et al. Exploring pharmacological active ingredients of traditional Chinese medicine by pharmacotranscriptomic map in ITCM. Briefings in bioinformatics 24, doi:10.1093/bib/bbad027 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bib/bbad027&link_type=DOI) 69. Qiao, L. et al. Evaluation of the immunomodulatory effects of anti-COVID-19 TCM formulae by multiple virus-related pathways. Signal Transduction and Targeted Therapy 6, doi:10.1038/s41392-021-00475-w (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41392-021-00475-w&link_type=DOI) 70. Round, J. L. & Mazmanian, S. K. The gut microbiota shapes intestinal immune responses during health and disease. Nature Reviews Immunology 9, 313–323, doi:10.1038/nri2515 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nri2515&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19343057&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000265960100012&link_type=ISI) 71. Gevers, D. et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host and Microbe 15, 382–392, doi:10.1016/j.chom.2014.02.005 (2014). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.chom.2014.02.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24629344&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000333098600016&link_type=ISI) 72. Atarashi, K. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341, doi:10.1126/science.1198469 (2011). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzMzEvNjAxNS8zMzciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8xMy8yMDI0LjAzLjEwLjI0MzAzOTExLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 73. Clemente, J. C., Ursell, L. K., Parfrey, L. W. & Knight, R. The impact of the gut microbiota on human health: An integrative view. Cell 148, 1258–1270, doi:10.1016/j.cell.2012.01.035 (2012). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2012.01.035&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22424233&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000301889500020&link_type=ISI) 74. Tang, W. H. W. et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. New England Journal of Medicine 368, 1575–1584, doi:10.1056/NEJMoa1109400 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1109400&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23614584&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F13%2F2024.03.10.24303911.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000317956300006&link_type=ISI)