Mechanisms of indigo naturalis on treating ulcerative colitis explored by GEO gene chips combined with network pharmacology and molecular docking

Sizhen Gu, Yan Xue, Yang Gao, Shuyang Shen, Yuli Zhang, Kanjun Chen, Shigui Xue, Ji Pan, Yini Tang, Hui Zhu, Huan Wu, Danbo Dou, Sizhen Gu, Yan Xue, Yang Gao, Shuyang Shen, Yuli Zhang, Kanjun Chen, Shigui Xue, Ji Pan, Yini Tang, Hui Zhu, Huan Wu, Danbo Dou

Abstract

Oral administration of indigo naturalis (IN) can induce remission in ulcerative colitis (UC); however, the underlying mechanism remains unknown. The main active components and targets of IN were obtained by searching three traditional Chinese medicine network databases such as TCMSP and five Targets fishing databases such as PharmMapper. UC disease targets were obtained from three disease databases such as DrugBank,combined with four GEO gene chips. IN-UC targets were identified by matching the two. A protein-protein interaction network was constructed, and the core targets were screened according to the topological structure. GO and KEGG enrichment analysis and bioGPS localization were performed,and an Herbs-Components-Targets network, a Compound Targets-Organs location network, and a Core Targets-Signal Pathways network were established. Molecular docking technology was used to verify the main compounds-targets. Ten core active components and 184 compound targets of IN-UC, of which 43 were core targets, were enriched and analyzed by bioGPS, GO, and KEGG. The therapeutic effect of IN on UC may involve activation of systemic immunity, which is involved in the regulation of nuclear transcription, protein phosphorylation, cytokine activity, reactive oxygen metabolism, epithelial cell proliferation, and cell apoptosis through Th17 cell differentiation, the Jak-STAT and IL-17 signaling pathways, toll-like and NOD-like receptors, and other cellular and innate immune signaling pathways. The molecular mechanism underlying the effect of IN on inducing UC remission was predicted using a network pharmacology method, thereby providing a theoretical basis for further study of the effective components and mechanism of IN in the treatment of UC.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Framework based on an integration strategy of network pharmacology.
Figure 2
Figure 2
Differential genes volcano map jointly analyzed by four GEO chips. mRNA of intestinal mucosal biopsies from normal group and UC group.
Figure 3
Figure 3
Venn diagram of the targets in UC and IN.
Figure 4
Figure 4
Herb-ingredients-targets (H-I-T) network. Red node represents IN, green nodes represent core active compounds of IN, purple nodes represent targets of IN.
Figure 5
Figure 5
The process of topological screening for the PPI network. The PPI network diagram of 43 core targets was obtained by screening 182 IN-UC composite targets through DC,BC,CC.
Figure 6
Figure 6
The PPI network of 182 nodes. The node size is proportional to the target degree in the network. The Blue nodes are the core targets of IN-UC.
Figure 7
Figure 7
Gene expression data were based on gene expression microarray analysis results in BioGPS. Targets-organs location network (H–O): Nodes represent targets and organ locations. Node size is relative to degree.
Figure 8
Figure 8
The GO enrichment analysis of core nodes. Including cellular components, molecular functions, biological processes and GO secondary classification.
Figure 9
Figure 9
Targets-pathways network (T-P network). The green Rhombus nodes represent the targets of IN-UC. The red triangles represent the related pathways. Node size is relative to degree.
Figure 10
Figure 10
The protein–ligand of the docking simulation. The three core compounds (indigo, indirubin, tryptamethrin) of IN are docked with three targets.

References

    1. Naganuma M. Treatment with indigo naturalis for inflammatory bowel disease and other immune diseases. Immunol. Med. 2019;42:16–21. doi: 10.1080/25785826.2019.1599158.
    1. Koning M, Ailabouni R, Gearry RB, Frampton CM, Barclay ML. Use and predictors of oral complementary and alternative medicine by patients with inflammatory bowel disease: a population-based, case-control study. Inflamm. Bowel Dis. 2013;19:767–778. doi: 10.1097/MIB.0b013e31827f27c8.
    1. Weizman W, et al. Characterisation of complementary and alternative medicine use and its impact on medication adherence in inflammatory bowel disease. Aliment. Pharmacol. Therap. 2012;35:342–349. doi: 10.1111/j.1365-2036.2011.04956.x.
    1. Kim S, et al. Adjunctive herbal medicine therapy for inflammatory bowel disease: a systematic review and meta-analysis. Eur. J. Integ. Med. 2017;12:12–22. doi: 10.1016/j.eujim.2017.03.009.
    1. Opheim R, Hoivik ML, Solberg IC, Moum B. Complementary and alternative medicine in patients with inflammatory bowel disease: the results of a population-based inception cohort study (IBSEN) J. Crohn's Colitis. 2012;6:345–353. doi: 10.1016/j.crohns.2011.09.007.
    1. Dossett ML, Davis RB, Lembo AJ, Yeh GY. Complementary and alternative medicine use by US adults with gastrointestinal conditions: results from the 2012 National Health Interview Survey. Am. J. Gastroenterol. 2014;109:1705–1711. doi: 10.1038/ajg.2014.108.
    1. Fukunaga K, et al. Placebo controlled evaluation of Xilei San, a herbal preparation in patients with intractable ulcerative proctitis. J. Gastroenterol Hepatol. 2012;27:1808–1815. doi: 10.1111/j.1440-1746.2012.07215.x.
    1. Zhang F, Li Y, Xu F, Chu Y, Zhao W. Comparison of Xilei-san, a Chinese herbal medicine, and dexamethasone in mild/moderate ulcerative proctitis: a double-blind randomized clinical trial. J Altern Complemet Med New York (N.Y.) 2013;19:838–842. doi: 10.1089/acm.2012.0296.
    1. Lin L, et al. Efficacy and safety of Indigo naturalis extract in oil (Lindioil) in treating nail psoriasis: a randomized, observer-blind, vehicle-controlled trial. Phytomed. Int. J. Phytother. Phytopharmacol. 2014;21:1015–1020. doi: 10.1016/j.phymed.2014.02.013.
    1. Gang L. Analysis of therapeutic effect of Kuijietong series prescription on 30 cases of chronic non-specific ulcerative colitis. Liaoning J. Tradit. Chin. Med. 2006;33:823–824.
    1. Sizhen G, et al. Effect of Kuijietong on phosphorylation of I κ B α/NF-κ B p65 signal pathway in rats with ulcerative colitis induced by sodium dextran sulfate. J. Guangzhou Univ. Tradit. Chin. Med. 2018;35:297–301.
    1. Sizhen G, et al. Intervention effect of Kuijietong on ulcerative colitis induced by sodium dextran sulfate. Chin. J. Integ. Tradit. Chin. West. Med. Dig. 2017;25:742–746.
    1. Zhanqi T, et al. A multicenter, randomized, double-blind, controlled study of compound Sophora flavescens colonic capsule in the treatment of ulcerative colitis of damp-heat type. Chin. J. Integ. Tradit. Chin. West. Med. 2011;31:172–176.
    1. Gong Y, et al. Efficacy and safety of Fufangkushen colon-coated capsule in the treatment of ulcerative colitis compared with mesalazine: a double-blinded and randomized study. J. Ethnopharmacol. 2012;141:592–598. doi: 10.1016/j.jep.2011.08.057.
    1. Sugimoto S, Naganuma M, Kanai T. Indole compounds may be promising medicines for ulcerative colitis. J. Gastroenterol. 2016;51:853–861. doi: 10.1007/s00535-016-1220-2.
    1. Ru J, et al. TCMSP: a database of systems pharmacology for drug discovery from herbal medicines. J. Cheminform. 2014;6:13. doi: 10.1186/1758-2946-6-13.
    1. Liu Z, et al. BATMAN-TCM: a bioinformatics analysis tool for molecular mechANism of traditional Chinese medicine. Sci. Rep. 2016;6:21146. doi: 10.1038/srep21146.
    1. Huang L, et al. TCMID 2.0: a comprehensive resource for TCM. Nucleic Acids Res. 2018;46:D1117–D1120. doi: 10.1093/nar/gkx1028.
    1. Xu X, et al. A novel chemometric method for the prediction of human oral bioavailability. Int. J. Mol. Sci. 2012;13:6964–6982. doi: 10.3390/ijms13066964.
    1. Mexia N, et al. A biomimetic, one-step transformation of simple indolic compounds to malassezia-related alkaloids with high AhR potency and efficacy. Chem. Res. Toxicol. 2019;32:2238–2249. doi: 10.1021/acs.chemrestox.9b00270.
    1. Micallef MJ, et al. The natural plant product tryptanthrin ameliorates dextran sodium sulfate-induced colitis in mice. Int. Immunopharmacol. 2002;2:565–578. doi: 10.1016/s1567-5769(01)00206-5.
    1. Wang Z, et al. Tryptanthrin protects mice against dextran sulfate sodium-induced colitis through inhibition of TNF-alpha/NF-kappaB and IL-6/STAT3 pathways. Molecules. 2018 doi: 10.3390/molecules23051062.
    1. Keiser MJ, et al. Relating protein pharmacology by ligand chemistry. Nat. Biotechnol. 2007;25:197–206. doi: 10.1038/nbt1284.
    1. Szklarczyk D, et al. STITCH 5: augmenting protein-chemical interaction networks with tissue and affinity data. Nucleic Acids Res. 2016;44:D380–384. doi: 10.1093/nar/gkv1277.
    1. Gfeller D, Michielin O, Zoete V. Shaping the interaction landscape of bioactive molecules. Bioinform. (Oxf. Engl.) 2013;29:3073–3079. doi: 10.1093/bioinformatics/btt540.
    1. Wang X, et al. PharmMapper 2017 update: a web server for potential drug target identification with a comprehensive target pharmacophore database. Nucleic Acids Res. 2017;45:W356–W360. doi: 10.1093/nar/gkx374.
    1. Wishart DS, et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46:D1074–D1082. doi: 10.1093/nar/gkx1037.
    1. Li YH, et al. Therapeutic target database update 2018: enriched resource for facilitating bench-to-clinic research of targeted therapeutics. Nucleic Acids Res. 2018;46:D1121–D1127. doi: 10.1093/nar/gkx1076.
    1. Piñero J, et al. DisGeNET: a comprehensive platform integrating information on human disease-associated genes and variants. Nucleic Acids Res. 2017;45:D833–D839. doi: 10.1093/nar/gkw943.
    1. UniProt: the universal protein knowledgebase. Nucleic Acids Res.45, D158–D169. 10.1093/nar/gkw1099 (2017).
    1. Szklarczyk D, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45:D362–D368. doi: 10.1093/nar/gkw937.
    1. Shannon P, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13:2498–2504. doi: 10.1101/gr.1239303.
    1. Missiuro PV, et al. Information flow analysis of interactome networks. PLoS Comput. Biol. 2009;5:e1000350. doi: 10.1371/journal.pcbi.1000350.
    1. Raman K, Damaraju N, Joshi GK. The organisational structure of protein networks: revisiting the centrality-lethality hypothesis. Syst Synth. Biol. 2014;8:73–81. doi: 10.1007/s11693-013-9123-5.
    1. Tang Y, Li M, Wang J, Pan Y, Wu FX. CytoNCA: a cytoscape plugin for centrality analysis and evaluation of protein interaction networks. Bio Syst. 2015;127:67–72. doi: 10.1016/j.biosystems.2014.11.005.
    1. Song W, Ni S, Fu Y, Wang Y. Uncovering the mechanism of Maxing Ganshi Decoction on asthma from a systematic perspective: a network pharmacology study. Sci. Rep. 2018;8:17362. doi: 10.1038/s41598-018-35791-9.
    1. Wan Y, et al. Utilising network pharmacology to explore the underlying mechanism of Wumei Pill in treating pancreatic neoplasms. BMC Complement. Altern. Med. 2019;19:158. doi: 10.1186/s12906-019-2580-y.
    1. Zhang W, et al. Systems pharmacology dissection of the integrated treatment for cardiovascular and gastrointestinal disorders by traditional chinese medicine. Sci. Rep. 2016;6:32400. doi: 10.1038/srep32400.
    1. Prevoršek Z, Gorjanc G, Paigen B, Horvat S. Congenic and bioinformatics analyses resolved a major-effect Fob3b QTL on mouse Chr 15 into two closely linked loci. Mamm. Genome Off. J. Int. Mamm. Genome Soc. 2010;21:172–185. doi: 10.1007/s00335-010-9252-z.
    1. Suh SY, An WG. Pulsatillae radixsystems pharmacological approach of on treating Crohn's Disease. Evid. Based Complement. Altern. Med.: eCAM. 2017;2017:4198035. doi: 10.1155/2017/4198035.
    1. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics: J. Integ. Biol. 2012;16:284–287. doi: 10.1089/omi.2011.0118.
    1. Xiong Y, et al. Network pharmacology-based research on the active component and mechanism of the antihepatoma effect of Rubia cordifolia L. J. Cell. Biochem. 2019;120:12461–12472. doi: 10.1002/jcb.28513.
    1. Li J, et al. Exploring the mechanism of Danshen against myelofibrosis by network pharmacology and molecular docking. Evid. Based Complement. Altern. Med. eCAM. 2018;2018:8363295. doi: 10.1155/2018/8363295.
    1. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010;31:455–461. doi: 10.1002/jcc.21334.
    1. Kawai S, et al. Indigo Naturalis ameliorates murine dextran sodium sulfate-induced colitis via aryl hydrocarbon receptor activation. J. Gastroenterol. 2017;52:904–919. doi: 10.1007/s00535-016-1292-z.
    1. Lin YK, et al. Protective effect of indigo naturalis extract against oxidative stress in cultured human keratinocytes. J. Ethnopharmacol. 2012;139:893–896. doi: 10.1016/j.jep.2011.12.037.
    1. Lihua C, et al. Extraction and antioxidant activity of indirubin from indigo indigo. J. Jishou Univ. (Nat. Sci. Ed.) 2013;25:72–76.
    1. Peiying W, et al. Textual Research on Materia Medica of Folium Isatidis, Radix Isatidis and Qingdai. J. Shanghai Acad. Tradit. Chin. Med. Shanghai Univ. Tradit. Chin. Med. 1996;25:50–52.
    1. Zhou D, et al. Evidence-based practice guideline of Chinese herbal medicine for psoriasis vulgaris (Bai Bi) Eur. J. Integ. Med. 2014;6:135–146. doi: 10.1016/j.eujim.2014.01.010.
    1. Han R. Highlight on the studies of anticancer drugs derived from plants in China. Stem Cells (Dayton, OH) 1994;12:53–63. doi: 10.1002/stem.5530120110.
    1. Suzuki H, et al. Therapeutic efficacy of the Qing Dai in patients with intractable ulcerative colitis. World J. Gastroenterol. 2013;19:2718–2722. doi: 10.3748/wjg.v19.i17.2718.
    1. Sugimoto S, et al. Clinical efficacy and safety of oral Qing-Dai in patients with ulcerative colitis: a single-center open-label prospective study. Digestion. 2016;93:193–201. doi: 10.1159/000444217.
    1. Naganuma M, et al. Efficacy of indigo naturalis in a multicenter randomized controlled trial of patients with ulcerative colitis. Gastroenterology. 2018;154:935–947. doi: 10.1053/j.gastro.2017.11.024.
    1. Li S, Zhang B. Traditional Chinese medicine network pharmacology: theory, methodology and application. Chin. J. Nat. Med. 2014;11:110–120. doi: 10.3724/sp.j.1009.2013.00110.
    1. Li H, et al. A network pharmacology approach to determine active compounds and action mechanisms of ge-gen-qin-lian decoction for treatment of type 2 diabetes. Evid. Based Complement. Altern. Med. 2014;2014:495840. doi: 10.1155/2014/495840.
    1. Zhang B, Wang X, Li S. An integrative platform of TCM network pharmacology and its application on a herbal formula Qing-Luo-Yin. Evid. Based Complement. Altern. Med. 2013;2013:456747. doi: 10.1155/2013/456747.
    1. Gao W, et al. Indirubin, a constituent of the Chinese herbal medicine Qing-Dai, attenuates dextran sulfate sodium-induced murine colitis. Yonago Acta Med. 2018;96:636–645. doi: 10.1139/bcb-2018-0041.
    1. Gaot W, et al. The combination of indirubin and isatin attenuates dextran sodium sulfate induced ulcerative colitis in mice. Biochem. Cell Biol. 2018;96:636–645. doi: 10.1139/bcb-2018-0041.
    1. Study on the anti-inflammatory effects of indirubin and tryptamine on RAW264.7 inflammatory cell model induced by LPS. World J. Integ. Tradit. Chin. West. Med. 10,1069–1072(1996).
    1. Lee IA, Kim EJ, Kim DH. Inhibitory effect of β-sitosterol on TNBS-induced colitis in mice. Planta Med. 2012;78:896–898. doi: 10.1055/s-0031-1298486.
    1. Ding K, et al. β-Sitosterol improves experimental colitis in mice with a target against pathogenic bacteria. J. Cell. Biochem. 2019;120:5687–5694. doi: 10.1002/jcb.27853.
    1. Kim KA, Lee IA, Gu W, Hyam SR, Kim DH. β-Sitosterol attenuates high-fat diet-induced intestinal inflammation in mice by inhibiting the binding of lipopolysaccharide to toll-like receptor 4 in the NF-κB pathway. Mol. Nutr. Food Res. 2014;58:963–972. doi: 10.1002/mnfr.201300433.
    1. Wang Y, et al. Effects of indigo naturalis on colonic mucosal injuries and inflammation in rats with dextran sodium sulphate-induced ulcerative colitis. Exp. Ther. Med. 2017;14:1327–1336. doi: 10.3892/etm.2017.4701.
    1. Liang YN, et al. Indigo naturalis ameliorates dextran sulfate sodium-induced colitis in mice by modulating the intestinal microbiota community. Molecules (Basel, Switz.) 2019 doi: 10.3390/molecules24224086.
    1. Xiao HT, et al. Qing-dai powder promotes recovery of colitis by inhibiting inflammatory responses of colonic macrophages in dextran sulfate sodium-treated mice. Chin. Med. 2015;10:29. doi: 10.1186/s13020-015-0061-x.
    1. Shi J, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526:660–665. doi: 10.1038/nature15514.
    1. Jiang H, et al. Identification of a selective and direct NLRP3 inhibitor to treat inflammatory disorders. J. Exp. Med. 2017;214:3219–3238. doi: 10.1084/jem.20171419.
    1. Zou Y, et al. Baicalin attenuates TNBS-induced colitis in rats by modulating the Th17/Treg paradigm. Arch. Pharmacal Res. 2015;38:1873–1887. doi: 10.1007/s12272-014-0486-2.
    1. Zhang L, et al. Heme oxygenase-1 ameliorates dextran sulfate sodium-induced acute murine colitis by regulating Th17/Treg cell balance. J. Biol. Chem. 2014;289:26847–26858. doi: 10.1074/jbc.M114.590554.
    1. Kimura A, Naka T, Nohara K, Fujii-Kuriyama Y, Kishimoto T. Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells. Proc. Natl. Acad. Sci. U.S.A. 2008;105:9721–9726. doi: 10.1073/pnas.0804231105.
    1. Li S, et al. Aryl hydrocarbon receptor signaling cell intrinsically inhibits intestinal group 2 innate lymphoid cell function. Immunity. 2018;49:915–928.e915. doi: 10.1016/j.immuni.2018.09.015.
    1. Bano JK, et al. ILC2s are the predominant source of intestinal ILC-derived IL-10. J. Exp. Med. 2020 doi: 10.1084/jem.20191520.
    1. Wang S, et al. Regulatory innate lymphoid cells control innate intestinal inflammation. Cell. 2017;171:201–216.e218. doi: 10.1016/j.cell.2017.07.027.

Source: PubMed

3
订阅