Clinical and Translational Research Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Jun 16, 2024; 12(17): 3105-3122
Published online Jun 16, 2024. doi: 10.12998/wjcc.v12.i17.3105
Unraveling the mechanism of malancao in treating ulcerative colitis: A multi-omics approach
Xing-Long Huang, Lu-Na Wu, Qin Huang, Yue Zhou, Feng Xiong, Hui-Ping Dong, Kai-Li Wang, Jue Liu, Hospital of Traditional Chinese Medicine in Qijiang District, Chongqing 401420, China
Xing-Long Huang, Lu-Na Wu, Xing-Long Huang and Lu-Na Wu.
Lei Qing, Qijiang Health Center for Maternal and Child Care, Chongqing 401420, China
Tai-Min Zhou, College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou Province, China
ORCID number: Xing-Long Huang (0009-0009-0576-9162); Jue Liu (0009-0000-2252-5957).
Co-corresponding authors: Kai-Li Wang and Jue Liu.
Author contributions: Huang XL and Liu J were involved in the study design and implementation, provided material support for obtaining the grant, and supervised the study; Wang KL coordinated and directed the implementation of the experimental validation; Wu LN performed the network analysis and wrote the manuscript; Huang Q, Zhou Y, Qing L, Xiong F, Dong HP and Zhou TM completed some of the experiments and statistical analyses; all authors reviewed and approved the final manuscript.
Supported by The Chongqing Science and Health Joint Chinese Medicine Technology Innovation and Application Development Project, No. 2022MSXM209.
Conflict-of-interest statement: 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.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/licenses/by-nc/4.0/.
Corresponding author: Jue Liu, PhD, Lecturer, Hospital of Traditional Chinese Medicine in Qijiang District, No. 69 Huideng Road, Tonghui Street, Qijiang District, Chongqing 401420, China. 30112067@qq.com
Received: February 27, 2024
Revised: April 11, 2024
Accepted: April 23, 2024
Published online: June 16, 2024
Processing time: 98 Days and 6.7 Hours

Abstract
BACKGROUND

Malancao (MLC) is a traditional Chinese medicine with a long history of utilization in treating ulcerative colitis (UC). Nevertheless, the precise molecular mechanisms underlying its efficacy remain elusive. This study leveraged ultra-high-performance liquid chromatography coupled with exactive mass spectrometry (UHPLC-QE-MS), network pharmacology, molecular docking (MD), and gene microarray analysis to discern the bioactive constituents and the potential mechanism of action of MLC in UC management.

AIM

To determine the ingredients related to MLC for treatment of UC using multiple databases to obtain potential targets for fishing.

METHODS

This research employs UHPLC-QE-MS for the identification of bioactive compounds present in MLC plant samples. Furthermore, the study integrates the identified MLC compound-related targets with publicly available databases to elucidate common drug disease targets. Additionally, the R programming language is utilized to predict the central targets and molecular pathways that MLC may impact in the treatment of UC. Finally, MD are conducted using AutoDock Vina software to assess the affinity of bioactive components to the main targets and confirm their therapeutic potential.

RESULTS

Firstly, through a comprehensive analysis of UHPLC-QE-MS data and public database resources, we identified 146 drug-disease cross targets related to 11 bioactive components. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis highlighted that common disease drug targets are primarily involved in oxidative stress management, lipid metabolism, atherosclerosis, and other processes. They also affect AGE-RAGE and apoptosis signaling pathways. Secondly, by analyzing the differences in diseases, we identified key research targets. These core targets are related to 11 active substances, including active ingredients such as quercetin and luteolin. Finally, MD analysis revealed the stability of compound-protein binding, particularly between JUN-Luteolin, JUN-Quercetin, HSP90AA1-Wogonin, and HSP90AA1-Rhein. Therefore, this suggests that MLC may help alleviate intestinal inflammation in UC, restore abnormal lipid accumulation, and regulate the expression levels of core proteins in the intestine.

CONCLUSION

The utilization of MLC has demonstrated notable therapeutic efficacy in the management of UC by means of the compound target interaction pathway. The amalgamation of botanical resources, metabolomics, natural products, MD, and gene chip technology presents a propitious methodology for investigating therapeutic targets of herbal medicines and discerning novel bioactive constituents.

Key Words: Malancao, Ulcerative colitis, Mass spectrum, Network pharmacology, Molecular docking

Core Tip: Eleven primary active constituents (nicotinic acid, luteolin, kaempferol, genkwanin, bessisterol, aloe emodin, wogonin, stigmasterol, rhein, quercetin, oroxylin A) and five key genes (AKT1, JUN, HSP90AA1, CASP3, IL6) were identified in malancao (MLC). Furthermore, three principal signaling pathways were determined through enrichment analysis from the Kyoto Encyclopedia of Genes and Genomes, suggesting that MLC exhibits a multi-component, multi-target, and multi-pathway approach in the treatment of ulcerative colitis.



INTRODUCTION

Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by colonic ulceration. The cause of its pathogenesis remains unclear but is typically associated with factors such as genetics, immune dysregulation, external environmental stimuli, and intestinal flora[1,2]. Currently, UC has emerged as a significant threat to human health, primarily characterized by intestinal inflammation and elevated levels of inflammatory factors and increased macrophage activation[3,4]. The prevalence of UC is 0.3%, and it is steadily rising, imposing a serious burden on individuals, families, and society as a whole[5,6]. Present treatments for UC include drugs such as sulfonamide antibiotics, peroxisome proliferator-activated receptor γ-specific agonists, glucocorticoids, and immune stimulants, which regulate the levels of inflammatory factors through different mechanisms of action to exert efficacy[7,8]. However, due to significant concerns such as unsatisfactory patient outcomes, high costs, and severe side effects, there is a clinical imperative to explore alternative therapeutic strategies to improve the prognosis of individuals with UC.

Traditional Chinese medicine (TCM) is perceived as a potent therapeutic option, utilized for thousands of years to address a range of clinical conditions[9]. Malancao (MLC), a TCM, has been extensively used in Chinese medicine practice. Numerous long-term clinical investigations indicate that MLC dramatically lowers atypical colonic tissue proliferation and suppresses pro-inflammatory factor expression, thereby reducing symptoms and slowing the course of UC[10]. Pharmacologically, MLC exerts anti-inflammatory and immunomodulatory effects[11]. The fundamental rationale and prospective processes underpinning MLC's efficacy in UC, however, remain unknown.

Network pharmacology (NP) is an approach that studies the modulation of disease targets by drug chemistry[12,13]. Analyzes systematic bioinformation and constructs biological interaction networks using public data in conjunction with multi-omics, further identifying target proteins and signaling pathways related to drug efficacy and disease[14,15]. Therefore, the goal of this study is to employ an innovative strategy to identify the molecular mechanisms and potential targets of MLC through plant resources, metabolomics, NP, molecular docking (MD), and gene microarray technologies to gain insights into the therapeutic properties of MLC in the treatment of UC (Figure 1).

Figure 1
Figure 1 Technical roadmap of malancao in treating ulcerative colitis. GEO: Gene Expression Omnibus; GO: Gene Ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes; UC: Ulcerative colitis; UHPLC-QE-MS: Ultra-high-performance liquid chromatography coupled with exactive mass spectrometry; MLC: Malancao.
MATERIALS AND METHODS
Preparation of MLC

MLC, which is widely distributed in various districts and counties in southern China, belongs to Kalimeris indica (L) Sch-Bip with a height of 30 cm-80 cm. MLC has slender underground rhizomes that are creeping and flat, and white in color. The leaves are simple and alternate, ranging from obovate to elliptic or lanceolate, with flower heads borne on the apices of upper branches, as shown in Figure 2.

Figure 2
Figure 2 Morphological characteristics of malancao. Malancao is a variety of Kalimeris indica (L) Sch-Bip.

Based on the collected data regarding the altitude, latitude, and longitude from ten regions in Dazu, Banan, Bishan, Fuling, Qijiang, Shizhu, Tongnan, Wanzhou, Wulong, and Shapingba districts of Chongqing Municipality, a digital elevation model image was generated using ArcGIS 10.2 software to illustrate the distribution map of the collection area for MLC, depicted in Figure 3. Three sampling points were randomly selected, and 3-10 samples were collected to create specimens at each point. These specimens were verified by Prof. Wei Shenghua from the School of Pharmacy at Guizhou University of Traditional Chinese Medicine and labeled as MLC-2021-09, before being stored in the Department of Pharmacy, Qijiang Hospital of Traditional Chinese Medicine.

Figure 3
Figure 3 The distribution of ten main producing areas of malancao. Sample site: The sampling locations are dispersed across multiple districts and counties within Chongqing, China.
Instruments and reagents

Ultra-high liquid phase (Vanquish, Thermo Fisher Scientific), high-resolution mass spectrometry (Orbitrap Exploris 120, Thermo Fisher Scientific), centrifuge (Heraeus Fresco17, Thermo Fisher Scientific), balance (BSA124S-CW, Thermo Fisher Scientific), ultrasound (PS-60AL, Shenzhen Leidebang Electronics Co., Ltd.).

Methanol and acetonitrile (CNW Technologies), ammonium acetate (SIGMA-ALDRICH), ethanoic acid (Fisher Chemical), ultrapure water (ddH20, Watson's).

Sample treatment

A 20 mg specimen was accurately weighed and amalgamated with 1000 μL of a specified extraction solution, comprising a methanol: Water ratio of 3 : 1 (v/v), and inclusive of an isotope-labeled internal standard mixture. This mixture was subjected to grinding at a frequency of 35 Hz for a duration of 4 min, followed by ultrasonication lasting 5 min within an ice-water bath. This procedure was repeated triply, after which the sample was conserved at -40 °C for an hour. Subsequently, it was centrifuged at a speed of 12000 rpm (equating to a relative centrifugal force of 13800 xg and a radius of 8.6 cm) maintained for 15 min at a temperature of 4 °C. To conclude, the resultant supernatant was methodically filtered utilizing a 0.22 μm microporous membrane and then decanted into a sterile glass vial in preparation for analytical procedures.

Ultra-high-performance liquid chromatography coupled with exactive mass spectrometry analysis

The target compounds were chromatographically separated on a Waters ACQUITY UPLC HSS T3 liquid chromatography column using a Vanquish ultra-high-performance liquid chromatography (UHPLC). The liquid chromatographic phase A was an aqueous phase containing 5 mmol/L ammonium acetate and 5 mmol/L acetic acid, and phase B was acetonitrile, followed by performing gradient elution with a column temperature of 35 °C, sample injection chamber temperature of 4 °C, and sample injection volume of 2 μL.

The Orbitrap Exploris 120 mass spectrometer conducted primary and secondary mass spectrometry data acquisition, controlled by Xcalibur software.

NP

NP, a novel method combining computer science and pharmacology, facilitates comprehensive elucidation of the mechanisms through which drugs act on diseases[16,17]. Currently, combining NP with multi-omics approaches is emerging as a future trend for studying the pharmacological effects and mechanisms of TCMs. Therefore, the active ingredients, targets, and signaling pathways of MLC in treating UC were identified from the perspectives of NP and plant metabolomics. The binding energies between drug molecules and their targets were calculated using MD simulations to further validate the predicted results of NP.

Collection of active compounds

All MLC compounds were identified based on the preliminary mass spectrometry analysis and previous literature studies. The TCM Systematic Pharmacology (TCMSP) database (https://old.tcmsp-e.com/) and SwissADME database (http://www.swissadme.ch/) were applied to screen all the candidate compounds of MLC, with the oral bioavailability (OB) ≥ 30% and drug-likeness (DL) ≥ 0.18.

Active compound target prediction

The active compound target prediction function in the TCMSP database was utilized to search for the relevant targets of the active ingredients in MLC. The UniProt database (https://www.uniprot.org/) was employed to convert all target proteins into gene symbols corresponding to the "Homo sapiens" species, standardizing gene names and biological information to prevent over-annotation of similar protein targets.

Prediction of UC-related targets and therapeutic targets

Using "UC" as the search phrase, comprehensive data on genes relevant to UC in "Homo sapiens" were collected from the GeneCards (https://www.genecards.org/), DisGeNET (https://www.disgenet.org/), and Online Mendelian Inheritance in Man databases (https://www.omim.org/). Following that, the Venny 2.1.0 online tool (https://bioinfogp.cnb.csic.es/tools/venny) was used to connect the relevant targets of the active compounds with those linked with UC.

Protein-protein interaction analysis

The STRING search tool (https://cn.string-db.org/) was utilized to query human gene data in order to retrieve protein-protein interaction (PPI) data for subsequent analysis, with a focus on the species "Homo sapiens" and a confidence score threshold of 0.4 or higher.

Network topology analysis

The "UC disease Gene-MLC targets" interaction network was constructed based on the molecular interaction data sourced from the STRING database. All networks were visualized using the Cytoscape 3.8.2 tool, an efficient tool for analyzing and visualizing molecular interaction networks. The topological significance of the central node of the network was evaluated by calculating the three parameters of degree centrality (DC), betweenness centrality (BC), and closeness centrality (CC) for each node[18,19]. A node with higher values for these three parameters indicates greater importance within the network, aiding in screening the key targets of MLC action in UC. Ultimately, targets with values ≥2 times the degree median were identified as core targets and incorporated for PPI visualization using cytoscape software.

Enrichment analysis

To explore the biological processes (BP) of UC proteins and their roles in signaling pathway transduction, disease-drug common targets were annotated, visualized, and integrated for discovery using the DAVID database (https://davidbioinformatics.nih.gov/) based on potential target proteins analyzed by the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Furthermore, potential targets were classified according to their associated BP, molecular functions (MF), and cellular localization through KEGG analysis.

Microarray and disease differential analysis

To obtain differentially expressed genes in inflammatory and non-inflammatory tissues of UC participants, the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/) was utilized to access GSE107499 gene chips. Hierarchical cluster analysis was conducted using the heatmap package in R (version 4.2.0), establishing disease-target libraries for subsequent analysis with significant expression variations (|log2FC| > 1, P < 0.05).

MD

Utilizing AutoDock Vina's MD functions, the interactions between MLC's active components and UC-associated target proteins were investigated via a software platform. Structures of the small molecule compounds were obtained in Mol2 format from the PubChem database (https://pubchem.ncbi.nlm.nih.gov/), while the receptor proteins' crystal structures were procured from the Protein Data Bank database (https://www.rcsb.org/). The thorough docking procedure included pre-simulation preparation of receptor proteins and small molecules, docking site identification, and binding energy computation.

RESULTS
Screening and identification of active components

To investigate the active components of MLC, compounds present in MLC were identified using UHPLC coupled with exactive mass spectrometry (UHPLC-QE-MS) secondary mass spectrometry qualitative matching, supplemented by a literature search. Using ProteoWizard software, the raw data were transformed into mzXML format and subsequently analyzed with a tailored R package (kernel XCMS) for peak identification, extraction, alignment, and integration[20]. Thereafter, compounds were cross-referenced against the BiotreeDB secondary mass spectrometry database for substance annotation, using an algorithmic score threshold of 0.3. MLC candidate compounds were rigorously assessed, necessitating the OB of ≥ 30% and the DL of ≥ 0.18, leading to the identification of 11 active ingredients within MLC (Figure 4 and Figure 5).

Figure 4
Figure 4 Total ion chromatogram plot of positive and negative ion modes of malancao detected by ultra-high-performance liquid chromatography coupled with exactive mass spectrometry. A: Positive ion mode mass spectrometry; B: Negative ion mode mass spectrometry.
Figure 5
Figure 5 Bioactive Compouds of malancao, n (%). CID: Pubchem's compound identifier, a non-zero integer for a unique chemical structure; 2D: Two-dimensional flat structures of compounds.
Active ingredient action targets and disease-related targets

MLC primarily contains flavonoids, terpenoids, alkaloids, organic acids, and other chemical constituents. A collection of 133 compounds was gathered from previous literature studies[21-24]. The TCMSP was employed to search for relevant targets of the active ingredients in MLC, resulting in 204 potential targets after duplicate removal.

4844 UC-related targets were found using the search of disease targets in the GeneCards database, utilizing "UC" as the keyword. To further elucidate the mechanism of MLC's action in treating UC, the Venny 2.1.0 online platform was used to match the relevant targets of the active ingredients with those associated with UC, identifying 146 cross-targets. Subsequently, the Cytoscape software facilitated the construction of the drug-compound-target network of MLC against UC (Figure 6 and Figure 7).

Figure 6
Figure 6 Venn diagram of active ingredient targets versus disease targets. Blue area: Targets of the malancao 's action; Yellow area: Protein targets of ulcerative colitis. MLC: Malancao; UC: Ulcerative colitis.
Figure 7
Figure 7 Drug-compound-target network of malancao against ulcerative colitis. Yellow nodes: Drug; Blue nodes: Compounds; Red nodes: Protein targets.
PPI and network topology analysis

To delineate the mechanism of MLC's effect on UC in detail, the final 146 common targets of the compound and the disease were entered into the STRING to create a PPI network. Analysis of the STRING network indicated that, after removing invalid nodes, the network comprised 145 gene nodes and 2805 edge interactions, boasting an average node degree of 38.40. It was noted that the ADRA1B gene did not interact with any other gene. Within the network, network nodes represent protein targets, and edges represent target-protein target associations.

To gauge the significance of nodes within the entire network, network topology analysis was conducted, calculating the DC, BC, and CC parameters for each node, and identifying targets exceeding twice the median degree as core targets. This analysis revealed 28 targets with a degree ≥ 24, representing the network's core targets (Figure 8), which included AKT1, JUN, HSP90AA1, CASP3, IL6, EGFR, RELA, MYC, MAPK1, among other targets.

Figure 8
Figure 8  Protein-protein interaction network between drug-disease related targets and core targets.
Enrichment analysis

Based on GO classification analysis, the target proteins were categorized into primary metabolic process, cellular process, biological regulation, and cellular communication. Regarding BP, 32 proteins were involved in response to lipopolysaccharide, 32 in response to reactive oxygen species, and 32 in response to molecule of bacterial origin. In MF, 18 proteins were implicated in ubiquitin-like protein ligase binding, 10 in nuclear receptor activity, and 10 in ligand-activated transcription factor activity. In CC, 17 proteins participated in membrane raft signaling, 9 in serine/threonine protein kinase complex, and 7 in cyclin-dependent protein kinase holoenzyme complex processes. Consequently, MLC could be utilized for the treatment of UC through several pathways, such as regulating protein secretion, and activating polymerase activity. The final GO classification annotation analysis results are displayed in (Figure 9A).

Figure 9
Figure 9 Visualization of Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis. A: Visualization of Gene Ontology classification annotation analysis; B: Visualization of Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. A node between two lines symbolizes interaction, with larger nodes indicating stronger relationships.

To unravel the mechanism of MLC's therapeutic action, the assembled protein targets were inputted into the KEGG database to pinpoint relevant pathways. A total of 137 target genes were enriched based on KEGG analysis with P < 0.05, predominantly involving in lipid, atherosclerosis, diabetes, inflammation, cancer, and virus-related pathways. In the exploration of inflammation-related pathways, AGE-RAGE, TNF, NF-κB, IL-17, and PI3K-Akt were the most prevalent signaling pathways[25]. The top 20 entries encompassing 102 gene targets most significantly enriched by MLC for UC were chosen for KEGG visualization analysis, and the relationship between these enriched pathways and the related targets is shown in (Figure 9B). Within the related signaling pathways, the AGE-RAGE pathway emerged as the most prominent, with 29 related target genes enriched, including CASP3, JUN, AKT1, VEGFA, CCND1, MMP2, MAPK1, CDK4, IL6, MAPK8, STAT1, IL1B, CCL2, IL1A, MAPK14, and others. This suggests that the AGE-RAGE pathway is a potential pathway in the treatment of UC with MLC.

Disease differential analysis

Gene expression profiles pertinent to UC were procured from the GEO database using "UC", "Homo sapiens", and "array expression profiles" as search terms. The microarray data file (GSE107499) was ultimately retrieved for further analysis. The experimental platform file (GPL15207) utilized in the above study was the Affymetrix Human Gene Expression Array, comprising a total sample size of 119 cases, with 51 female patients and 68 male patients. The microarray data was scrutinized for 18835 gene targets, encompassing 849 genes with significant differences, 309 up-regulated genes, and 540 down-regulated genes. Furthermore, the microarray data outlined the comprehensive gene expression in inflamed versus non-inflamed colon tissues in individuals with UC. Differential expression analysis was conducted to ascertain whether PPI core targets exhibited disparate expressions between inflamed and non-inflamed samples, with P < 0.05 indicating statistically significant. Subsequently, box plots were used to show the specific expression of the target genes (Figure 10). The findings indicated that the top 20 core target genes in degree like JUN, HSP90AA1, EGFR, and VEGFA were lowly expressed in inflamed colon tissues, while IL6, MYC, CCND1, IL1B, ESR1, STAT1, MMP9, and HIF1A were markedly expressed in inflamed colon tissues. As the gene expression profiles were sourced from GEO, we were exempt from requiring ethical approval and conducting new experiments on patients or animals.

Figure 10
Figure 10  Disease differential analysis for ulcerative colitis in Gene Expression Omnibus database. A: Heatmap; B: Volcano plot; C-N: Show the differential expression of degree's top 20 core target genes in inflamed and non-inflamed colonic tissues, respectively.
MD validation

Based on the PPI interaction network, key targets were identified by MD to further validate the affinity of the corresponding chemical components of the drug to the protein. The small molecule ligand and protein receptor files necessary for the study were obtained from the Protein Data Bank database. All receptor structures were processed to remove organic matter and water molecules, and to add hydrogenated charge distribution. Ligand small molecule structures were optimized using Chem 3D software, while PyMOL software facilitated the optimization of both ligand compounds and receptors. MD was performed with AutoDock Vina software. The top five core targets were molecularly docked with their corresponding eight active ingredients based on the degree value. MD results indicated binding energies between the active ingredients and the target proteins ranging from -5.9 kcal/mol to -8.6 kcal/mol. Lower binding energies signify greater affinity between the active ingredients and UC's core targets (Table 1). Additionally, four target protein-active molecules with low binding energies were selected to plot a docking pattern, as shown in Figure 11. The results further demonstrated that the active components of MLC, including luteolin, quercetin, wogonin, and rheinic acid, acted on the core targets of protein kinase receptors and exhibited good affinity for the key targets, suggesting that the active components of MLC can treat or alleviate UC through these targets.

Figure 11
Figure 11  Molecular docking of some core targets with active ingredients. A: Docking models for JUN-Luteolin; B: Docking models for JUN-Quercetin; C: Docking models for HSP90AA1-Wogonin; D: Docking models for HSP90AA1-Rhein.
Table 1 Binding energy of eight active components and five target gene.
Chemical name
Chemical formula
Protein data bank ID
Target
Binding energy
(kcal/mol)
LuteolinC15H10O61H10AKT1-6.2
1A02JUN-8.6
1BYQHSP90AA1-7.4
1CP3CASP3-7.3
1ALUIL6-7.1
QuercetinC15H10O71H10AKT1-5.9
1A02JUN-7.9
1BYQHSP90AA1-7.4
1CP3CASP3-7.4
1ALUIL6-7.1
WogoninC16H12O51H10AKT1-6.1
1A02JUN-7.7
1BYQHSP90AA1-7.9
1CP3CASP3-7.4
1ALUIL6-6.0
KaempferolC15H10O61H10AKT1-6.0
1A02JUN-7.1
1BYQHSP90AA1-7.1
1CP3CASP3-7.6
RheinC15H8O61A02JUN-7.4
1BYQHSP90AA1-8.0
Aloe-emodinC15H10O51BYQHSP90AA1-6.4
1CP3CASP3-7.0
GenkwaninC16H12O51BYQHSP90AA1-7.0
Oroxylin AC16H12O51BYQHSP90AA1-7.0
1CP3CASP3-7.2
1ALUIL6-6.0
DISCUSSION

UC has emerged as a global disease in recent years, with a growing frequency in developed countries. Inflammation has been linked to the development of numerous cancers, according to epidemiological studies. Chronic inflammation of the intestinal mucosa, a major risk factor for colorectal cancer, is a side effect of UC development, with damage to intestinal epithelial cells being one of the primary causes of UC development[26-28]. Mesalazine (5-ASA) and glucocorticoids are the principal treatments for UC according to current diagnostic and therapeutic guidelines. Mesalazine is the conventional treatment for mild to severe UC that is not complex. Oral budesonide-MMX, a new formulation of budesonide in a multimatrix system, is recommended for mild to moderately active inflammatory UC that has not responded to mesalazine[29,30]. Kucharzik noted that systemic glucocorticoids are recommended for UC treatment and serve as the primary approach for patients with acute, severe UC when other methods fail to induce remission.

The existing pharmacological treatment of UC is accompanied by a series of problems and a high incidence of adverse effects[31,32]. Moreover, multiple morbidities tend to occur during treatment, and the long-term use of drugs such as sulfonamides and glucocorticosteroids can lead to a gradual decrease in medication adherence and a substantial economic burden for patients[33]. Therefore, alternative treatments with plant medicine are particularly important.

Techniques such as proteomics, plant metabolomics, and both in vivo and in vitro assays have been widely employed to explore disease diagnosis and drug therapy mechanisms, as evidenced by prior studies[34-36]. Recent findings indicate that Kuijieling decoction is closely linked with T cell activation, Th17 cell differentiation, and immune response. Furthermore, Kuijieling decoction has been observed to enhance vitamin A metabolism and RA/RARα signaling in UC rats and T lymphocytes[37]. Investigated loganic acid's impact on UC using in vivo and in vitro methods, revealing its potential to diminish oxidative and inflammatory reactions triggered by lipopolysaccharide and dextran sodium sulfate in UC[38]. This action is mediated by the regulation of TLR4/NF-κB and SIRT1/Nrf2 pathways. Similarly, Wang et al[39] discovered that Siwu decoction modulates UC's mechanisms by suppressing the STAT3 and NF-κB pathways, reducing inflammatory cytokine production, and enhancing epithelial repair in experimental colitis.

In the study, HPLC, NP, and bioinformatics were initially applied to successfully identify 11 active molecules and 3 important pathways associated with UC. The GO enrichment analysis indicated that MLC might exert its anti-UC effects by regulating biological functions. Pathway enrichment analysis revealed that many other diseases besides UC were also enriched, possibly due to the presence of the same molecular targets in different pathological processes of the disease. This is both a limitation of NP and a point of divergence for research directions. Therefore, enrichment pathways closely related to UC were selected for our study. Eventually, 28 core targets of the drug-disease target network were identified, and three pathways related to UC, were significantly enriched based on network construction and central network evaluation. Studies have indicated that RAGE signaling plays a role in the development of intestinal inflammation and permeability in both Crohn's disease and UC[40]. TNF-α, a crucial cytokine involved in the pathogenesis of inflammatory bowel disease, is traditionally viewed as a key mediator in amplifying mucosal inflammation, which is a central process of Crohn's disease pathogenesis[41-43]. Emerging evidence supports the significant role of the IL-23/IL-17 axis in the development of UC, demonstrating that inhibiting the IL-23/IL-17 axis and the downstream activation of pro-inflammatory factors is essential for suppressing the inflammatory response and alleviating UC[44-46]. Therefore, it is evident that MLC's treatment of UC primarily directly influences the AGE-RAGE pathway, TNF pathway, and IL-17 pathway, with the AGE-RAGE pathway being the most closely related to UC. Additionally, disease differential analysis revealed variations in the expression of each protein target between inflamed and non-inflamed samples, and identified protein targets with significant differences for subsequent studies. Lastly, MD verified that the binding energy of the core target was lower than -5.9 kcal/mol, suggesting that the compounds adhere tightly to the UC targets, promising better therapeutic outcomes. Using the aforementioned methods, we can more distinctly identify the targets and specific mechanisms of plant medicine in treating diseases and pinpoint key active molecules, which holds considerable significance for the development of plant medicine and disease treatment.

CONCLUSION

In this study, a novel combinatorial strategy based on plant resources, plant metabolomics, NP, MD techniques, and gene chip analysis techniques was first applied to pinpoint potential targets and mechanisms of MLC in treating UC. This will establish a new paradigm for uncovering the potential mechanisms underlying the pharmacological effects of medicinal plants. Furthermore, the study furnishes information and a theoretical basis for deeper exploration of the mechanism, bolstering supports clinical practice. Future systematic molecular biology experiments are necessary to verify the precise mechanisms.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade A

P-Reviewer: Venkatesan N, India S-Editor: Luo ML L-Editor: A P-Editor: Zhao YQ

References
1.  Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nat Rev Gastroenterol Hepatol. 2015;12:205-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 930]  [Cited by in F6Publishing: 1053]  [Article Influence: 117.0]  [Reference Citation Analysis (0)]
2.  Keum N, Giovannucci E. Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies. Nat Rev Gastroenterol Hepatol. 2019;16:713-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 777]  [Cited by in F6Publishing: 1217]  [Article Influence: 243.4]  [Reference Citation Analysis (1)]
3.  Tatiya-Aphiradee N, Chatuphonprasert W, Jarukamjorn K. Immune response and inflammatory pathway of ulcerative colitis. J Basic Clin Physiol Pharmacol. 2018;30:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 183]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
4.  Zhu L, Gu P, Shen H. Protective effects of berberine hydrochloride on DSS-induced ulcerative colitis in rats. Int Immunopharmacol. 2019;68:242-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 148]  [Article Influence: 29.6]  [Reference Citation Analysis (0)]
5.  Gajendran M, Loganathan P, Jimenez G, Catinella AP, Ng N, Umapathy C, Ziade N, Hashash JG. A comprehensive review and update on ulcerative colitis. Dis Mon. 2019;65:100851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 228]  [Article Influence: 45.6]  [Reference Citation Analysis (0)]
6.  Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, Panaccione R, Ghosh S, Wu JCY, Chan FKL, Sung JJY, Kaplan GG. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2017;390:2769-2778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2677]  [Cited by in F6Publishing: 3285]  [Article Influence: 469.3]  [Reference Citation Analysis (0)]
7.  Helal MG, Abd Elhameed AG. Graviola mitigates acetic acid-induced ulcerative colitis in rats: insight on apoptosis and Wnt/Hh signaling crosstalk. Environ Sci Pollut Res Int. 2021;28:29615-29628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
8.  Wada K, Nakajima A, Blumberg RS. PPARgamma and inflammatory bowel disease: a new therapeutic target for ulcerative colitis and Crohn's disease. Trends Mol Med. 2001;7:329-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
9.  Johnson SB, Park HS, Gross CP, Yu JB. Complementary Medicine, Refusal of Conventional Cancer Therapy, and Survival Among Patients With Curable Cancers. JAMA Oncol. 2018;4:1375-1381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 164]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
10.  Wang L, Liu Y, Shen G, He G, Qiu S, Li B. Mechanisms of Si-Wu Decoction in the treatment of ulcerative colitis revealed by network pharmacology and experimental verification. J Ethnopharmacol. 2023;317:116847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Zhong RF, Xu GB, Wang Z, Wang AM, Guan HY, Li J, He X, Liu JH, Zhou M, Li YJ, Wang YL, Liao SG. Identification of anti-inflammatory constituents from Kalimeris indica with UHPLC-ESI-Q-TOF-MS/MS and GC-MS. J Ethnopharmacol. 2015;165:39-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
12.  Wang X, Wang ZY, Zheng JH, Li S. TCM network pharmacology: A new trend towards combining computational, experimental and clinical approaches. Chin J Nat Med. 2021;19:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 141]  [Article Influence: 47.0]  [Reference Citation Analysis (0)]
13.  Zhou Z, Chen B, Chen S, Lin M, Chen Y, Jin S, Chen W, Zhang Y. Applications of Network Pharmacology in Traditional Chinese Medicine Research. Evid Based Complement Alternat Med. 2020;2020:1646905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 159]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
14.  Noor F, Tahir Ul Qamar M, Ashfaq UA, Albutti A, Alwashmi ASS, Aljasir MA. Network Pharmacology Approach for Medicinal Plants: Review and Assessment. Pharmaceuticals (Basel). 2022;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 94]  [Article Influence: 47.0]  [Reference Citation Analysis (0)]
15.  Zhang YQ, Mao X, Guo QY, Lin N, Li S. Network pharmacology-based approaches capture essence of chinese herbal medicines. Chinese Herbal Med. 2016;82:107-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
16.  Wang YL, Cui T, Li YZ, Liao ML, Zhang HB, Hou WB, Zhang TJ, Liu L, Huang H, Liu CX. Prediction of quality markers of traditional chinese medicines based on network pharmacology. Chinese Herbal Med. 2019;114:349-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
17.  Yuan H, Ma Q, Cui H, Liu G, Zhao X, Li W, Piao G. How Can Synergism of Traditional Medicines Benefit from Network Pharmacology? Molecules. 2017;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 263]  [Article Influence: 37.6]  [Reference Citation Analysis (0)]
18.  Guo Q, Zhong M, Xu H, Mao X, Zhang Y, Lin N. A systems biology perspective on the molecular mechanisms underlying the therapeutic effects of buyanghuanwu decoction on ischemic stroke. Rejuv Res. 2015;184:313-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
19.  Zhang Y, Bai M, Zhang B, Liu C, Guo Q, Sun Y, Wang D, Wang C, Jiang Y, Lin N, Li S. Author Correction: Uncovering pharmacological mechanisms of Wu-tou decoction acting on rheumatoid arthritis through systems approaches: drug-target prediction, network analysis and experimental validation. Sci Rep. 2018;8:15924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
20.  Smith CA, Want EJ, O'Maille G, Abagyan R, Siuzdak G. XCMS: processing mass spectrometry data for metabolite profiling using nonlinear peak alignment, matching, and identification. Anal Chem. 2006;78:779-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3171]  [Cited by in F6Publishing: 3286]  [Article Influence: 193.3]  [Reference Citation Analysis (0)]
21.  Huang W, Wen Z, Wang Q, Chen R, Li Z, Feng Y, Yang S. The chemical profile of active fraction of Kalimeris indic and its quantitative analysis. Biomed Chromatogr. 2020;347:161-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
22.  Liu J, Zhao YT, Lu WC, Huang PH, Song TY, Li PH. Bioactive Compounds in Malanto (Kalimeris indica) Leaves and Their Antioxidant Characteristics. Agriculture. 2023;131:61-79.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Wang GK, Zhang N, Wang Y, Liu JS, Wang G, Zhou ZY, Lu CC, Yang JS. The hepatoprotective activities of Kalimeris indica ethanol extract against liver injury in vivo. Food Sci Nutr. 2019;7:3797-3807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
24.  Xie L, Lin Q, Guo K, Tong C, Shi S, Shi F. HPLCDADQTOF-MS/MS based comprehensive metabolomic profiling of phenolic compounds in Kalimeris indica anti-inflammatory fractions. IND CROP PROD. 2019;14:263-281.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Gao B, Ahmad MF, Nagy LE, Tsukamoto H. Inflammatory pathways in alcoholic steatohepatitis. J Hepatol. 2019;70:249-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 211]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
26.  Kucharzik T, Koletzko S, Kannengiesser K, Dignass A. Ulcerative Colitis-Diagnostic and Therapeutic Algorithms. Dtsch Arztebl Int. 2020;117:564-574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 45]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
27.  Rajamäki K, Taira A, Katainen R, Välimäki N, Kuosmanen A, Plaketti RM, Seppälä TT, Ahtiainen M, Wirta EV, Vartiainen E, Sulo P, Ravantti J, Lehtipuro S, Granberg KJ, Nykter M, Tanskanen T, Ristimäki A, Koskensalo S, Renkonen-Sinisalo L, Lepistö A, Böhm J, Taipale J, Mecklin JP, Aavikko M, Palin K, Aaltonen LA. Genetic and Epigenetic Characteristics of Inflammatory Bowel Disease-Associated Colorectal Cancer. Gastroenterology. 2021;161:592-607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 82]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
28.  Samadder NJ, Valentine JF, Guthery S, Singh H, Bernstein CN, Leighton JA, Wan Y, Wong J, Boucher K, Pappas L, Rowe K, Burt RW, Curtin K, Smith KR. Family History Associates With Increased Risk of Colorectal Cancer in Patients With Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol. 2019;17:1807-1813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
29.  Sandborn WJ, Travis S, Moro L, Jones R, Gautille T, Bagin R, Huang M, Yeung P, Ballard ED 2nd. Once-daily budesonide MMX® extended-release tablets induce remission in patients with mild to moderate ulcerative colitis: results from the CORE I study. Gastroenterology. 2012;143:1218-1226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 166]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
30.  Travis SP, Danese S, Kupcinskas L, Alexeeva O, D'Haens G, Gibson PR, Moro L, Jones R, Ballard ED, Masure J, Rossini M, Sandborn WJ. Once-daily budesonide MMX in active, mild-to-moderate ulcerative colitis: results from the randomised CORE II study. Gut. 2014;63:433-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 170]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
31.  Fiala O, Ostasov P, Hosek P, Sorejs O, Liska V, Buchler T, Poprach A, Kucera R, Topolcan O, Sustr J, Sedivcova M, Finek J. The Predictive Role of Primary Tumour Sidedness in Metastatic Colorectal Cancer Treated With Targeted Agents. Anticancer Res. 2019;39:5645-5652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
32.  Ginghină O, Hudiță A, Zaharia C, Tsatsakis A, Mezhuev Y, Costache M, Gălățeanu B. Current Landscape in Organic Nanosized Materials Advances for Improved Management of Colorectal Cancer Patients. Materials (Basel). 2021;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
33.  Jaramillo AP, Castells J, Ibrahimli S, Siegel S. Recurrent Multidrug-Resistant Clostridium difficile Infection Secondary to Ulcerative Colitis a Case Report. Med Sci (Basel). 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
34.  Liu CS, Xia T, Luo ZY, Wu YY, Hu YN, Chen FL, Tang QF, Tan XM. Network pharmacology and pharmacokinetics integrated strategy to investigate the pharmacological mechanism of Xianglian pill on ulcerative colitis. Phytomedicine. 2021;82:153458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 43]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
35.  Lu Y, Chen Y, Li Y, Xu S, Lian D, Liang J, Jiang D, Chen S, Hou S. Monotropein inhibits colitis associated cancer through VDR/JAK1/STAT1 regulation of macrophage polarization. Int Immunopharmacol. 2023;124:110838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
36.  Mills RH, Dulai PS, Vázquez-Baeza Y, Sauceda C, Daniel N, Gerner RR, Batachari LE, Malfavon M, Zhu Q, Weldon K, Humphrey G, Carrillo-Terrazas M, Goldasich LD, Bryant M, Raffatellu M, Quinn RA, Gewirtz AT, Chassaing B, Chu H, Sandborn WJ, Dorrestein PC, Knight R, Gonzalez DJ. Multi-omics analyses of the ulcerative colitis gut microbiome link Bacteroides vulgatus proteases with disease severity. Nat Microbiol. 2022;7:262-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 103]  [Article Influence: 51.5]  [Reference Citation Analysis (0)]
37.  Xiao S, Yan Y, Shao M, Zhou X, Niu Z, Wu Y, Li Y, Cui Y, Long Y, Du Q. Kuijieling decoction regulates the Treg/Th17 cell balance in ulcerative colitis through the RA/RARα signaling pathway. J Ethnopharmacol. 2024;318:116909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
38.  Prakash AN, Prasad N, Puppala ER, Panda SR, Jain S, Ravichandiran V, Singh M, Naidu VGM. Loganic acid protects against ulcerative colitis by inhibiting TLR4/NF-κB mediated inflammation and activating the SIRT1/Nrf2 anti-oxidant responses in-vitro and in-vivo. Int Immunopharmacol. 2023;122:110585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
39.  Wang MF, Li H, Cui J, Chen YH, Cui Y. Effects of Kalimeris indica (L.) Sch Bip on colitis-associated colorectal cancer. Front Pharmacol. 2022;13:1119091.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
40.  Snelson M, Lucut E, Coughlan MT. The Role of AGE-RAGE Signalling as a Modulator of Gut Permeability in Diabetes. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 23]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
41.  Pugliese D, Felice C, Papa A, Gasbarrini A, Rapaccini GL, Guidi L, Armuzzi A. Anti TNF-α therapy for ulcerative colitis: current status and prospects for the future. Expert Rev Clin Immunol. 2017;13:223-233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 70]  [Article Influence: 8.8]  [Reference Citation Analysis (1)]
42.  Su C, Salzberg BA, Lewis JD, Deren JJ, Kornbluth A, Katzka DA, Stein RB, Adler DR, Lichtenstein GR. Efficacy of anti-tumor necrosis factor therapy in patients with ulcerative colitis. Am J Gastroenterol. 2002;97:2577-2584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 134]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
43.  Tian L, Huang YX, Tian M, Gao W, Chang Q. Downregulation of electroacupuncture at ST36 on TNF-alpha in rats with ulcerative colitis. World J Gastroenterol. 2003;9:1028-1033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 56]  [Cited by in F6Publishing: 55]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
44.  Eastaff-Leung N, Mabarrack N, Barbour A, Cummins A, Barry S. Foxp3+ regulatory T cells, Th17 effector cells, and cytokine environment in inflammatory bowel disease. J Clin Immunol. 2010;30:80-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 304]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
45.  Wang Z, Chen J, Chen Z, Xie L, Wang W. Clinical effects of ursodeoxycholic acid on patients with ulcerative colitis may improve via the regulation of IL-23-IL-17 axis and the changes of the proportion of intestinal microflora. Saudi J Gastroenterol. 2021;27:149-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
46.  Zhang Z, Zheng M, Bindas J, Schwarzenberger P, Kolls JK. Critical role of IL-17 receptor signaling in acute TNBS-induced colitis. Inflamm Bowel Dis. 2006;12:382-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 359]  [Cited by in F6Publishing: 355]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]