Figure - available from: Evidence-based Complementary and Alternative Medicine
This content is subject to copyright. Terms and conditions apply.
OPLS-DA of samples based on serum metabolomics. (a) Overview of the OPLS-DA model of CG and MG samples based on the serum metabolomics. (b) Permutation test of the OPLS-DA model of CG and MG samples based on the serum metabolomics. (c) Observation diagnostics of the OPLS-DA model of CG and MG samples. (d) Score plot of the OPLS-DA model based on the first and second components. (e) Root mean square error of cross validation (RMSECV) of the OPLS-DA model of CG and FC samples. (f) Overview of the OPLS-DA model of FC and MG samples based on the serum metabolomics. (g) Permutation test of the OPLS-DA model of FC and MG samples based on the serum metabolomics. (h) Observation diagnostics of the OPLS-DA model of FC and MG samples. (i) Score plot of the OPLS-DA model of FC and MG samples based on the first and second components. (j) Root mean square error of cross validation (RMSECV) of the OPLS-DA model of MG and FC samples.

OPLS-DA of samples based on serum metabolomics. (a) Overview of the OPLS-DA model of CG and MG samples based on the serum metabolomics. (b) Permutation test of the OPLS-DA model of CG and MG samples based on the serum metabolomics. (c) Observation diagnostics of the OPLS-DA model of CG and MG samples. (d) Score plot of the OPLS-DA model based on the first and second components. (e) Root mean square error of cross validation (RMSECV) of the OPLS-DA model of CG and FC samples. (f) Overview of the OPLS-DA model of FC and MG samples based on the serum metabolomics. (g) Permutation test of the OPLS-DA model of FC and MG samples based on the serum metabolomics. (h) Observation diagnostics of the OPLS-DA model of FC and MG samples. (i) Score plot of the OPLS-DA model of FC and MG samples based on the first and second components. (j) Root mean square error of cross validation (RMSECV) of the OPLS-DA model of MG and FC samples.

Source publication
Article
Full-text available
This study was intended to identify the shifts in the metabolomics profile of the hepatic tissue damaged by alcohol consumption and verify the potential restorative action of flos carthami (the flowers of Carthamus tinctorius, FC) in the protection of alcohol-induced injury by attenuating the level of identified metabolites. Rats were treated with...

Citations

... In addition, a rigorous toxicity assessment of concentrated water solution of HH using a dose of 1000 mg/kg suggested that HH did not show any toxic effects on the reproduction and early development of maternal animals and their offspring [69]. Furthermore, HH at 476 mg/kg, 1430 mg/kg, or 4290 mg/kg doses was able to improve histopathological changes in the liver and reduce ALT and AST levels to exert hepatoprotective effects by modulating hepatic metabolic profiles, reducing inflammatory responses, and inhibiting oxidative stress in an alcohol-induced acute liver injury model in rats [70]. In light of numerous conflicting evidences available, it is difficult to state whether HH has significant hepatotoxicity. ...
Article
Full-text available
Carthamus tinctorius L. (Honghua, HH) is an herbal medicine and functional food widely used to treat chronic liver diseases, including liver fibrosis. By using network pharmacology and molecular docking experiments, the present study aims to determine the bioactive components, potential targets, and molecular mechanisms of HH for treating liver fibrosis. The components of HH were screened from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform and literature, and the SwissTargetPrediction database was used to predict the treatment targets of HH. Genecards and DisGeNET databases contained targets for liver fibrosis, and the STRING database provided networks of protein–protein interactions. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed using the Database of Annotation, Visualization and Integrated Discovery. The protein–protein interactive network and drug–component–major target–pathway interactive network were visualized and analyzed by Cytoscape software. Finally, Autodock Vina and Discovery Studio software were used for molecular docking Validation. A total of 23 candidate bioactive compounds with 187 treatment targets of HH were acquired from the databases and literature. A total of 121 overlapping targets between HH and liver fibrosis were found to provide the molecular basis for HH on liver fibrosis. Quercetin, beta carotene, and lignan were identified as key components with targeting to ESR1, PIK3CA, and MTOR. HH is engaged in the intervention of various signaling cascades associated with liver fibrosis, such as PI3K/AKT/mTOR pathway, MAPK pathway, and PPAR pathway. In conclusion, HH treats liver fibrosis through multi-component, multi-target, and multi-pathway mechanisms.
Article
Full-text available
Purpose Compound Anoectochilus roxburghii (Wall.) Lindl oral liquid (CAROL) is often as a hepatoprotective agent. The present study aimed to elucidate the protective mechanism of CAROL against alcoholic liver injury in rats by untargeted metabolomics combined with multivariate statistical analysis. Methods An alcoholic liver disease model was established in sprague-dawley (SD) rats by gavage of alcohol, and CAROL treatment was administered. The hepatoprotective effect of CAROL was evaluated by examining liver tissues changes and detecting biochemical index activities and cytokines in serum and liver homogenates. The metabolites in serum samples were examined using ultrahigh-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UHPLC–QTOF/MS) and multivariate statistical analysis to screen for differentially expressed metabolites and Kyoto Encyclopedia of Genes and Genomes (KEGG) to assess potential metabolic pathways. Results CAROL has the potential to downregulate inflammation levels and alleviate oxidative stress. The differential metabolites are mainly engaged in riboflavin metabolism, arginine and proline metabolism, phenylalanine, tyrosine and tryptophan biosynthesis metabolism, phenylalanine metabolism, pyrimidine metabolism, and vitamin B6 metabolism to achieve hepatoprotective effects. Conclusion CAROL may exhibit beneficial hepatoprotective effects by reducing inflammation, mitigating oxidative stress, and modulating metabolites and their metabolic pathways.This study has important implications for advancing the clinical application of CAROL.