Identifying Active Compounds and Targets of <i>Fritillariae thunbergii</i> against Influenza-Associated Inflammation by Network Pharmacology Analysis and Molecular Docking

Complications due to influenza are often associated with inflammation with excessive release of cytokines. The bulbs of <i>Fritillariae thunbergii</i> (FT) have been traditionally used to control airway inflammatory diseases, such as bronchitis and pneumonia. To elucidate active compound...

Full description

Bibliographic Details
Main Authors: Minjee Kim, Ki Hoon Park, Young Bong Kim
Format: Article
Language:English
Published: MDPI AG 2020-08-01
Series:Molecules
Subjects:
Online Access:https://www.mdpi.com/1420-3049/25/17/3853
Description
Summary:Complications due to influenza are often associated with inflammation with excessive release of cytokines. The bulbs of <i>Fritillariae thunbergii</i> (FT) have been traditionally used to control airway inflammatory diseases, such as bronchitis and pneumonia. To elucidate active compounds, the targets, and underlying mechanisms of FT for the treatment of influenza-induced inflammation, systems biology was employed. Active compounds of FT were identified through the TCMSP database according to oral bioavailability (OB) and drug-likeness (DL) criteria. Other pharmacokinetic parameters, Caco-2 permeability (Caco-2), and drug half-life (HL) were also identified. Biological targets of FT were retrieved from DrugBank and STITCH databases, and target genes associated with influenza, lung, and spleen inflammation were collected from DisGeNET and NCBI databases. Compound-disease-target (C-D-T) networks were constructed and merged using Cytoscape. Target genes retrieved from the C-D-T network were further analyzed with GO enrichment and KEGG pathway analysis. In our network, GO and KEGG results yielded two compounds (beta-sitosterol (BS) and pelargonidin (PG)), targets (PTGS1 (COX-1) and PTGS2 (COX-2)), and pathways (nitric oxide, TNF) were involved in the inhibitory effects of FT on influenza-associated inflammation. We retrieved the binding affinity of each ligand-target, and found that PG and COX-1 showed the strongest binding affinity among four binding results using a molecular docking method. We identified the potential compounds and targets of FT against influenza and suggest that FT is an immunomodulatory therapy for influenza-associated inflammation.
ISSN:1420-3049