Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2023

Open Access 01.12.2023 | Research

Network pharmacology -based study on the mechanism of traditional Chinese medicine in the treatment of glioblastoma multiforme

verfasst von: Chen Liang, Binbin Zhang, Ruichun Li, Shiwen Guo, Xiaoxuan Fan

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2023

Abstract

Background

Glioblastoma multiforme (GBM) is one of the most common primary malignant brain tumors. Yi Qi Qu Yu Jie Du Fang (YYQQJDF) is a traditional Chinese medicine (TCM) prescription for GBM. The present study aimed to use a network pharmacology method to analyze the underlying mechanism of YQQYJDF in treating GBM.

Methods

GBM sample data, active ingredients and potential targets of YQQYJDF were obtained from databases. R language was used to screen differentially expressed genes (DEGs) between GBM tissues and normal tissues, and to perform enrichment analysis and weighted gene coexpression network analysis (WGCNA). The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database was used to perform a protein‒protein interaction (PPI) analysis. A Venn diagram was used to obtain the core target genes of YQQYJDF for GBM treatment. Molecular docking was used to verify the binding between the active ingredient molecules and the proteins corresponding to the core target genes. Cell proliferation assays and invasion assays were used to verify the effect of active ingredients on the proliferation and invasion of glioma cells.

Results

A total of 73 potential targets of YQQYJDF in the treatment of GBM were obtained. Enrichment analyses showed that the biological processes and molecular functions involved in these target genes were related to the activation of the G protein-coupled receptor (GPCR) signaling pathway and the regulation of hypoxia. The neuroactive ligand‒receptor pathway, the cellular senescence pathway, the calcium signaling pathway, the cell cycle pathway and the p53 signaling pathway might play important roles. Combining the results of WGCNA and PPI analysis, five core target genes and their corresponding four core active ingredients were screened. Molecular docking indicated that the core active ingredient molecules and the proteins corresponding to the core target genes had strong binding affinities. Cell proliferation and invasion assays showed that the core active ingredients of YQQYJDF significantly inhibited the proliferation and invasion of glioma cells (P < 0.01).

Conclusions

The present study predicted the possible active ingredients and targets of YQQYJDF in treating GBM, and analyzed its possible mechanism. These results may provide a basis and ideas for further research.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12906-023-04174-7.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
GBM
Glioblastoma multiforme
TCM
Traditional Chinese medicine
YQQJDF
Yi Qi Qu Yu Jie Du Fang
TCGA
The cancer genome atlas
DEG
Differentially expressed genes
WGCNA
Weighted gene coexpression network analysis
STRING
Search Tool for the Retrieval of Interacting Genes/Proteins
OS
Overall survival
TCMSP
Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform
OB
Oral bioavailability
DL
Drug likeness
GO
Gene ontology
KEGG
Kyoto encyclopedia of genes and genomes
PPI
Protein-protein interaction
DMSO
Dimethyl sulfoxide
CCK-8
Cell counting kit-8
FBS
Fetal bovine serum
MMP1
Matrix metalloproteinase-1/Interstitial collagenase
MMP2
Matrix metalloproteinase-2
MMP9
Matrix metalloproteinase-9
PLAU
Urokinase-type plasminogen activator
SELE
E-selectin
5-HT
5-Hydroxytryptamine
GPCR
G protein-coupled receptor
HIF-1
Hypoxia-inducible factor 1
VEGF
Vascular endothelial growth factor
VEGFR
Vascular endothelial growth factor receptor

Background

Glioblastoma multiforme (GBM) is one of the most common human primary malignant brain tumors [1]. At present, the standard treatment for GBM is surgery combined with radiotherapy and chemotherapy, but the prognosis of GBM patients receiving standard treatment is still poor [1]. Therefore, it is very important to find more effective treatments to improve the prognosis of GBM patients, which is also a challenge for neurosurgeons.
Traditional Chinese medicine (TCM) has been used to treat brain tumors for thousands of years. The TCM classic “Zhongzang Jing”, which was written during the Han Dynasty in ancient China (around 200 AD), records the treatment of brain tumors with TCM. Many ingredients of TCM have been proven to have therapeutic effects on GBM [2, 3]. Yi Qi Qu Yu Jie Du Fang (YYQQJDF) is a TCM prescription for GBM based on classical TCM theory and data mining analysis of glioma cases of TCM [4, 5], which can improve the prognosis of patients with GBM in clinical application. This TCM prescription is composed of eight TCMs, including “Huangqi”, “Chuangxiong”, “Banxia”, “Baihuashecao”, “Gancao”, “Shancigu”, “Shichangpu” and “Taizishen”. However, due to the large number of active ingredients and targets of this TCM prescription, the mechanisms of its treatment effect in GBM are still unclear.
Network pharmacology is a method of understanding drug actions and interactions with multiple targets [6]. It uses bioinformatic methods to systematically catalog the molecular interactions of a drug. It is also an effective method of studying the mechanism of TCM prescriptions in treating diseases [7, 8]. The present study aimed to use a network pharmacology method to analyze the underlying mechanism of YQQYJDF in treating GBM.

Methods

GBM sample data collection

Transcriptome sequencing data and corresponding clinical information of The Cancer Genome Atlas (TCGA)-GBM cohort were obtained from the TCGA database [9] using the “TCGAbiolinks” package in R software (The R Foundation for Statistical Computing, Vienna, Austria). These data include 174 samples, of which 160 GBM samples have fully available survival data and five samples are normal tissue.

Screening of Differentially Expressed Genes (DEGs)

Using the “limma” and “DESeq2” packages in R software, the differentially expressed mRNAs were identified between five normal and 169 tumor tissues in the TCGA-GBM cohort based on adjusted standards of P < 0.01 and | log2 (fold change) |> 2. The “ggplot” package was used to plot the volcano plot.

Weighted Gene Coexpression Network Analysis (WGCNA)

The DEG data and clinical data of the TCGA-GBM cohort were used to construct the weighted gene coexpression network using the “WGCNA” package in R software to identify DEGs related to the overall survival (OS) of patients with GBM. The soft threshold power was seven.

Selection of active ingredients of YQQYJDF

The formulas of YQQYJDF were “Huangqi” (Hedysarum Multijugum Maxim.), “Chuangxiong” (Chuanxiong Rhizoma), “Banxia” (Arum Ternatum Thunb.), “Baihuashecao” (Hedyotis Diffusae Herba), “Gancao” (Liquorice), “Shancigu” (Pseudobulbus Cremastrae Seu Pleiones), “Shichangpu” (Acoritataninowii Rhizoma) and “Taizishen” (Pseudostellariae Radix). The related ingredients were retrieved from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) [10] and Traditional Chinese Medicine database [11]. Ingredients with oral bioavailability (OB) ≥ 30% and drug likeness (DL) ≥ 0.18 were selected as the active ingredients.

Prediction of the targets of YQQYJDF active ingredients

The targets of the active ingredients of YQQYJDF were retrieved from the TCMSP database. The gene symbols and Entrez ID of targets were obtained by the “org.Hs.eg.db” package in R software.

Prediction of potential targets of YQQYJDF in the treatment of GBM

The target genes of YQQYJDF active ingredients and DEGs of GBM were matched using a Venn diagram with Venny 2.1 [12], and the intersecting genes were obtained as potential target genes for YQQYJDF in the treatment of GBM.

Enrichment analysis

The “clusterProfiler”, “org.Hs.eg.db”, “enrichplot” and “pathview” packages in R software with p value cutoff = 0.05 and q value cutoff = 0.05 were used to perform Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) [13] signal pathway analysis for these target genes. The “ggplot2” package was used to visualize the results. The network graph of the relationship between target genes and pathways was drawn using Cytoscape 3.9.1 software (Cytoscape Consortium, USA).

Protein‒protein interaction (PPI) analysis

The STRING database [14] was used to perform a PPI analysis to reveal the interaction network relationship of target genes at the translation level. The network graph of PPI was drawn by Cytoscape 3.9.1 software (Cytoscape Consortium, USA).

Prediction of the core target genes of YQQYJDF for GBM treatment

The core target genes of YQQYJDF for the treatment of GBM were obtained by using the Venn diagram to intersect genes closely related to the patient’s prognosis obtained from WGCNA with the target genes of the active ingredients of YQQYJDF, the DEGs of GBM and the genes with a degree value greater than 10 in the PPI network.

Molecular docking

The core active ingredients of YQQYJDF were obtained by matching the core target genes of YQQYJDF for GBM treatment and the target genes of YQQYJDF active ingredients. The 2D structure of the active ingredient small molecule ligand was obtained from the Drugbank database [15]. The 3D structures of the proteins of the core target genes were obtained from the RCSB protein data bank database [16]. Molecular docking was performed with AutoDockTools software 1.5.7 (Molecular Graphics Laboratory, The Scripps Research Institute). It is generally believed that the smaller the binding energy, the more stable the docking modules. A binding energy value less than -5.0 kcal/mol indicates that there is a good affinity between the receptor and the ligand, and a value below -7.0 kcal/mol indicates that there is a very strong affinity between the receptor and the ligand. Visualization of molecular docking results was performed with PyMOL software 2.5.4 (Schrödinger, LLC.).

Cell culture and preparation of drugs

The U87MG human glioblastoma cell line was purchased from the Cell Resource Center of the Chinese Academy of Sciences (Shanghai, China). U87 cells were cultured in Dulbecco’s modified Eagle’s medium (Gibco; Thermo Fisher Scientific,Inc. USA) with 10% fetal bovine serum (FBS) (Gibco; Thermo Fisher Scientific,Inc. USA) at 37 °C in the presence of 5% CO2. Luteolin, quercetin and kaempferol were purchased from Sigma‒Aldrich (St. Louis, USA). Stigmasterol was purchased from Avanti Polar Lipids, Inc. (Alabaster, USA). All drugs were dissolved in dimethyl sulfoxide (DMSO) and stored at -20 °C.

Cell proliferation assay

The Cell Counting Kit-8 (CCK-8) (Elabscience, China) assay was used to analyze the proliferation of glioma cells. U87 cells were seeded at a density of 5 × 103 cells/well in 24-well plates. The cells were incubated in medium containing 40 μmol/L luteolin, quercetin, kaempferol or stigmasterol. The control group was treated with an equal volume of the solvent (DMSO) in the culture medium. After 24 h, glioma cells were then analyzed by CCK-8 assay according to the manufacturer’s instructions. Briefly, 50 μL CCK-8 solution was added to each well. After incubating at 37 °C for 4 h, the absorbance value at 450 nm was measured.

Matrigel invasion assay

U87 cells were seeded at a density of 5 × 104 cells/well in Transwell chambers (BD Biosciences, Bedford, USA) precoated with 50 μl Matrigel (BD Biosciences) diluted with culture medium. Serum-free culture medium containing 40 μmol/l luteolin, quercetin, kaempferol or stigmasterol was used to incubate the U87 cells and medium containing 20% FBS was used in the lower chamber as the chemoattractant. After 24 h, noninvasive cells were removed with cotton swabs, and the invasive cells were dyed with 0.1% crystal violet (Sigma‒Aldrich) and analyzed directly under an inverted fluorescent microscope (magnification, × 200, IX51; Olympus Corporation, Japan). The average cell number in five random visual fields was considered to represent the number of invasive cells of each group.

Statistical analysis

Values are presented as the mean ± standard deviation. Data were analyzed using SPSS 25.0 software (SPSS, Inc. USA). One-way analysis of variance was used to compare the groups, and the least significant difference post hoc test was performed to further assess intergroup differences. A P value < 0.05 indicated a statistically significant difference.

Results

DEGs between GBM tissues and normal tissues

As shown in Fig. 1, 3100 DEGs were identified between normal tissues and GBM tissues from the TCGA-GBM cohort; 1415 of the DEGs were upregulated and 1685 of the DEGs were downregulated in GBM.

Identification of GBM clinical module genes by WGCNA

Expression data for 3100 DEGs and the OS data of GBM patients were used to construct a scale-free network by WGCNA. As shown in Fig. 2, nine modules were identified; including black, blue, magenta, red, yellow, brown, green, gray, pink and turquoise. According to the results of the module-trait relationship analysis, a total of 236 genes in the magenta and red modules (shown in Table 1 and Supplementary Table 1) were considered to be closely related to the prognosis of GBM patients (P < 0.05).
Table 1
WGCNA color modules related to the prognosis of GBM patients
Module
Number of genes
P value
Magenta
59
0.01
Red
176
0.03
Sum
235
 

Active ingredients and potential target genes of YQQYJDF

After screening, a total of 119 active ingredients and 252 corresponding target genes of YQQYJDF were obtained (Supplementary Table 2).

Prediction of YQQYJDF targets in the treatment of GBM

As shown in Fig. 3 and Supplementary Table 3, after taking the intersection of YQQYJDF potential target genes and DEGs of GBM, a total of 73 YQQYJDF potential targets in the treatment of GBM were obtained.

Enrichment analysis of potential targets of YQQYJDF in the treatment of GBM

A total of 807 GO items were obtained by GO enrichment analysis of 73 potential YQQYJDF targets in the treatment of GBM; There were 656 enriched biological process (BP) terms, 57 enriched cellular component (CC) terms and 94 enriched molecular function (MF) terms. Figure 4A, B and C show the top 10 enriched terms in each of the three categories. The length of the line in the figures represents the number of genes enriched in this term, and the color represents the significance of the enrichment. The results showed that the mechanism of YQQYJDF in the treatment of GBM might be related to several biological processes such as regulation of membrane potential, the G protein-coupled receptor signaling pathway, the serotonin receptor signaling pathway, the response to hypoxia, and leukocyte migration. Related molecular functions included neurotransmitter receptor activity, G protein-coupled receptor activity, serotonin receptor activity, etc. Moreover, some cellular components, such as postsynaptic membrane, synaptic membrane, dendrite membrane and membrane raft, were involved.
Sixty-eight pathways were obtained through KEGG pathway enrichment analysis. The top 20 pathways are shown in Fig. 4D. The length of the line represents the number of genes enriched in the pathway; the color represents the significance of the enrichment. The results showed that the potential targets of YQQYJDF in the treatment of GBM were related to the pathways that regulate neuroactive ligand‒receptor interactions, malignant tumors, the cell cycle, and cellular senescence.

PPI network analysis

As shown in Fig. 5A, the PPI network showed the interaction of potential target genes at the protein level. Figure 5B shows the 28 genes with a PPI network degree greater than 10. The darker the color is, the higher the degree value, indicating that the corresponding protein is more important in the network.

Selection of the core target genes of YQQYJDF for GBM treatment

As shown in Fig. 6, after taking the intersection of YQQYJDF potential target genes, WGCNA screening genes, genes with a degree value greater than 10 in the PPI network and DEGs of GBM, five core target genes of YQQYJDF that are very important in the PPI network and closely related to the prognosis of glioma patients were obtained (Table 2).
Table 2
The core target genes of YQQYJDF for GBM treatment
Gene symbol
Gene name
MMP1
Interstitial collagenase
MMP2
Matrix metalloproteinase-2
MMP9
Matrix metalloproteinase-9
PLAU
Urokinase-type plasminogen activator
SELE
E-selectin

Molecular docking results

Molecular docking of proteins encoded by five core target genes with the corresponding core active ingredients showed that their binding energies were all less than -5.0 kcal/mol, indicating that there are good binding effects between the target proteins and active ingredients (Table 3). The visualized results of molecular docking are shown in Fig. 7.
Table 3
The molecular docking results
Target
Active ingredient
Molecule ID
Binding energy (kcal/mol)
MMP1
Luteolin
MOL000006
-10.1
MMP2
Luteolin
MOL000006
-7.8
MMP9
Luteolin
MOL000006
-10.0
MMP1
Quercetin
MOL000098
-10.1
MMP2
Quercetin
MOL000098
-7.0
MMP9
Quercetin
MOL000098
-10.0
PLAU
Quercetin
MOL000098
-8.0
SELE
Quercetin
MOL000098
-5.3
MMP1
Kaempferol
MOL000422
-9.7
SELE
Kaempferol
MOL000422
-5.1
PLAU
Stigmasterol
MOL000449
-6.1
MMP1 Matrix metalloproteinase-1/ Interstitial collagenase, MMP2 Matrix metalloproteinase-2, MMP9 Matrix metalloproteinase-9, PLAU Urokinase-type plasminogen activator, SELE E-selectin

The core active ingredients of YYQQJDF inhibit the proliferation of glioma cells

After treatment with the core active ingredients of YYQQJDF, the proliferation ability of U87 glioma cells was detected by CCK-8 assay. As shown in Fig. 8, after 24 h of intervention, four core active ingredients luteolin, quercetin, kaempferol and stigmasterol significantly inhibited the proliferation of U87 glioma cells (F = 52.075, P < 0.01).

The core active ingredients of YYQQJDF inhibit the invasion of glioma cells

Following treatment with the four core active ingredients of YYQQJDF for 24 h, the number of invading glioma cells was significantly reduced compared with that in the control group (F = 25.648, P < 0.01) (Fig. 9).

Discussion

Although TCM has been used to treat various diseases for thousands of years, it is difficult to gain recognition in the field of modern medicine due to theoretical differences and the lack of direct experimental evidence [17]. For example, the principles of TCM such as the concept of “vital energy”, are esoteric and difficult to validate under modern medicine methods. The diagnostic and treatment methods of TCM are also fundamentally different from modern medicine [18]. Therefore, research on the active ingredients and biological functions of TCM has always been the focus of TCM pharmacological research [19]. The results of these studies are helpful to reveal the mechanism of TCM in treating diseases [20, 21]. Recently, network pharmacological analysis has been increasingly applied to TCM research. Network pharmacology can be used to search for the relationships among herbs, active ingredients, diseases and target genes, predict key active ingredients, targets and possible mechanisms of TCM in the treatment of diseases, and provide clues for further research [7, 8]. In the present study, network pharmacological methods were used to analyze the potential mechanisms of YQQYJDF in treating GBM.
According to the results, five core target genes and four corresponding active ingredients were obtained. Among the five core target genes, MMP-1 is considered closely related to the malignant degree of glioma [22] and the prognosis of glioma patients [23]. Similar to other members of the MMP family, MMP-1 can not only target the extracellular matrix, but also activate other bioactive molecules and activate downstream proinvasive and prooncogenic signaling mechanisms [24]. Inhibition of MMP1 expression can significantly inhibit glioma proliferation and invasion [25]. MMP-2 and MMP-9 are often discussed together due to their similar mechanisms and signaling pathways. As two key members of the MMP family, they are closely related to the migration, proliferation, vascular mimicry formation, and tumor growth of glioma [26, 27]. Many interventions can inhibit glioma growth by inhibiting the expression of MMP-2 and MMP-9 [28, 29]. PLAU participates in a series of cell physiological activities, such as migration and invasion [30]. Abnormal expression of PLAU is associated with the development of glioma and the prognosis of glioma patients[31]. Upregulation of PLAU also promotes the migration of glioma cells [32]. SELE is a selectin cell adhesion molecule expressed on endothelial cells induced by cytokines, that plays an important role in inflammation [33]. At present, SELE is considered to be related to the progression, metastasis and development of various tumors [34, 35]. Few studies have examined the relationship between SELE and glioma. A study found that SELE can promote glioma metastasis [36].
In YQQYJDF, the core active ingredients corresponding to these five target genes are luteolin, quercetin, kaempferol and stigmasterol. Luteolin is a natural flavone compound that is considered to have antitumor effects [37, 38]. Some studies have confirmed that luteolin can significantly inhibit glioma cell proliferation, migration, and invasion and induce their apoptosis [39, 40]. These effects may be related to the activation of the MAPK pathway [39] or disruption of the function of RNA binding proteins [40]. Quercetin is another recognized small molecule compound with antitumor effects [41, 42]. This natural compound can affect a variety of cellular and molecular processes of tumors, such as apoptosis, metastasis, and autophagy [43]. The antiglioma effect of quercetin involves multiple mechanisms, including apoptosis induction, metastasis and invasion inhibition, and cytotoxicity induction [4446]. Kaempferol has also shown antitumor effects in many studies[47]. Studies have shown that kaempferol can inhibit the proliferation, metabolism and migration of glioma cells in vitro and inhibit tumor growth in vivo [48, 49]. These effects may be related to programmed cell death and oxidative stress [48, 50]. Different from the other three active ingredients, stigmasterol is a sterol compound, that has the main function of maintaining the structure and physiology of cell membranes [51]. There are few studies on the effect of stigmasterol on glioma. In one study, researchers found that stigmasterol had cytotoxic effects on glioma cells [52]. The results of the present study verified that these four core active ingredients can significantly inhibit the proliferation and invasion of glioma cells. These core active ingredients may play an important role in the antiglioma effects of YDDYJDF.
In addition to the five core target genes, 68 other potential target genes of YQQYJDF for GBM treatment were obtained in this study. According to the results of the GO analysis, many of the biological processes and molecular functions involving in these target genes are related to the activation of the G protein-coupled receptor (GPCR) signaling pathway and the regulation of hypoxia. GPCRs and their downstream signaling targets play a central role in the initiation and progression of tumors [53]. They can regulate the growth and survival of tumor cells, and provide nutrients and routes for tumor metastasis by inducing cytoskeletal changes and angiogenesis [54]. GPCR signaling pathways are also important for the tumorigenesis and development of glioma [55]. Therefore, they are also considered potential targets for the treatment of glioma [56]. Hypoxia can promote glioma growth, angiogenesis, and invasion, which is achieved mainly through the hypoxia-inducible factor 1 (HIF-1) pathway to upregulate target genes such as vascular endothelial growth factors (VEGF), VEGF receptors (VEGFR), and MMPs [57]. The mechanism of YQQYJDF in the treatment of GBM may be related to regulating the biological processes involved in the GPCR signaling pathway and hypoxia. In the KEGG analysis, the neuroactive ligand‒receptor pathway, cellular senescence pathway, calcium signaling pathway, cell cycle pathway, p53 signaling pathway and other signaling pathways were considered to be related to YQQYJDF treatment of GBM. As a classical tumor-related signaling pathway, the p53 signaling pathway has long been a focus of research. Many interventions have been proven to regulate the biological behavior of glioma from different aspects through the p53 signaling pathway [58, 59]. The cell cycle pathway participates in cell cycle regulation as a downstream pathway of many signaling pathways [60, 61]. In glioma, interventions can regulate the cell cycle pathway through Akt, PI3K and other signaling pathways to regulate the glioma cell cycle and apoptosis [62, 63]. The calcium signaling pathway is also crucial for the behavior of glioma cells [64]. The calcium signal may regulate glioma cell motility, which is important in the early stages of tumor development [64]. Similar to the cell cycle pathway, cellular senescence is regulated by upstream pathways [65]. Many interventions can inhibit glioma cell proliferation through the cellular senescence pathway [66, 67]. The neuroactive ligand‒receptor pathway involves various neuroactive substances, such as acetylcholine, dopamine, and 5-hydroxytryptamine (5-HT) [68]. Acetylcholine participates in the regulation of astrocyte differentiation into glioma cells [69]. Dopamine can induce glioma cell apoptosis and inhibit tumor growth by regulating mitochondrial apoptosis and anti-inflammatory signaling pathways [70]. 5-HT can induce the expression of glial cell line-derived neurotrophic factor (GDNF) in glioma cells [71], which is an important prerequisite for the initiation and development of glioma [72]. However, the above biological processes and signaling pathways involved in the possible mechanism of YQQYJDF treatment of GBM need to be further verified in vivo and in vitro. In addition, according to the GO analysis and KEGG results, the mechanism of YQQYJDF in treating GBM may involve other biological processes and signaling pathways, but this needs to be verified in further studies. These results may provide potential new research directions.
There are still some limitations to this study. First, the ingredients and target genes in this study are mainly from databases. Due to the limitations of data sources and differences in screening strategies, some ingredients or target genes may be omitted. Second, this is a study based on network pharmacology and molecular docking, and the results have been preliminarily verified in vitro. Further in vitro and in vivo experiments are needed to verify these possible mechanisms. These shortcomings will be addressed in further research.

Conclusions

In conclusion, the present study involved the prediction of the possible active ingredients and targets of YQQYJDF in the treatment of GBM using network pharmacology and the analysis of its possible mechanism. These results may provide a basis and ideas for further research.

Acknowledgements

Not applicable

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Shergalis A, Bankhead A 3rd, Luesakul U, Muangsin N, Neamati N. Current challenges and opportunities in treating glioblastoma. Pharmacol Rev. 2018;70(3):412–45.PubMedPubMedCentral Shergalis A, Bankhead A 3rd, Luesakul U, Muangsin N, Neamati N. Current challenges and opportunities in treating glioblastoma. Pharmacol Rev. 2018;70(3):412–45.PubMedPubMedCentral
2.
Zurück zum Zitat Zhang F, Lv H, Zhang X. Erinacerins, novel glioma inhibitors from hericium erinaceus, induce apoptosis of u87 cells through Bax/Capase-2 pathway. Anticancer Agents Med Chem. 2020;20(17):2082–8.PubMed Zhang F, Lv H, Zhang X. Erinacerins, novel glioma inhibitors from hericium erinaceus, induce apoptosis of u87 cells through Bax/Capase-2 pathway. Anticancer Agents Med Chem. 2020;20(17):2082–8.PubMed
3.
Zurück zum Zitat Li T, Xiao Y, Wang Z, Xiao H, Liu H. The mechanism study of common flavonoids on antiglioma based on network pharmacology and molecular docking. Evid Based Complement Alternat Med. 2022;2022:2198722.PubMedPubMedCentral Li T, Xiao Y, Wang Z, Xiao H, Liu H. The mechanism study of common flavonoids on antiglioma based on network pharmacology and molecular docking. Evid Based Complement Alternat Med. 2022;2022:2198722.PubMedPubMedCentral
4.
Zurück zum Zitat Wang H, Luo W, Ma ST, Zhao XP, Fan XX, Wang K, Yang YF. Medication rules of TCM in treating glioma based on data mining. W J Tradit Chinese Med. 2021;34(04):71–4. Wang H, Luo W, Ma ST, Zhao XP, Fan XX, Wang K, Yang YF. Medication rules of TCM in treating glioma based on data mining. W J Tradit Chinese Med. 2021;34(04):71–4.
5.
Zurück zum Zitat Wang L, Wang S, Yang T, Tong YP, Kang YZ, Wang AW, Wan JL, Zhao TY, Cheng CC, Fan YP. Analysis of TCM syndromes and medication characteristics of 32 patients with brainstem glioma. Chin J Tradit Chinese Med Pharm. 2017;32(11):4943–5. Wang L, Wang S, Yang T, Tong YP, Kang YZ, Wang AW, Wan JL, Zhao TY, Cheng CC, Fan YP. Analysis of TCM syndromes and medication characteristics of 32 patients with brainstem glioma. Chin J Tradit Chinese Med Pharm. 2017;32(11):4943–5.
6.
Zurück zum Zitat Zhou S, Bi J, Zhao W, Zhao J, Wan H, Wang S. Study on the mechanism of fentanyl in pain treatment based on network pharmacology. J Healthc Eng. 2022;2022:4139200.PubMedPubMedCentral Zhou S, Bi J, Zhao W, Zhao J, Wan H, Wang S. Study on the mechanism of fentanyl in pain treatment based on network pharmacology. J Healthc Eng. 2022;2022:4139200.PubMedPubMedCentral
7.
Zurück zum Zitat Zheng L, Jiang H, Li R, Song L, Chen R, Dong H. The pharmacological mechanisms of xiaochaihutang in treating breast cancer based on network pharmacology. Contrast Media Mol Imaging. 2022;2022:3900636.PubMedPubMedCentral Zheng L, Jiang H, Li R, Song L, Chen R, Dong H. The pharmacological mechanisms of xiaochaihutang in treating breast cancer based on network pharmacology. Contrast Media Mol Imaging. 2022;2022:3900636.PubMedPubMedCentral
8.
Zurück zum Zitat Zou M, Zhu Y. Exploring the molecular mechanism of Tong Xie Yao Fang in treating ulcerative colitis using network pharmacology and molecular docking. Evid Based Complement Alternat Med. 2022;2022:8141443.PubMedPubMedCentral Zou M, Zhu Y. Exploring the molecular mechanism of Tong Xie Yao Fang in treating ulcerative colitis using network pharmacology and molecular docking. Evid Based Complement Alternat Med. 2022;2022:8141443.PubMedPubMedCentral
11.
13.
17.
Zurück zum Zitat Cheung F. TCM: made in China. Nature. 2011;480(7378):S82–3.PubMed Cheung F. TCM: made in China. Nature. 2011;480(7378):S82–3.PubMed
18.
Zurück zum Zitat Wang J, Wong YK, Liao F. What has traditional Chinese medicine delivered for modern medicine? Expert Rev Mol Med. 2018;20:e4.PubMed Wang J, Wong YK, Liao F. What has traditional Chinese medicine delivered for modern medicine? Expert Rev Mol Med. 2018;20:e4.PubMed
19.
Zurück zum Zitat Yang Y, Zhang Z, Li S, Ye X, Li X, He K. Synergy effects of herb extracts: pharmacokinetics and pharmacodynamic basis. Fitoterapia. 2014;92:133–47.PubMed Yang Y, Zhang Z, Li S, Ye X, Li X, He K. Synergy effects of herb extracts: pharmacokinetics and pharmacodynamic basis. Fitoterapia. 2014;92:133–47.PubMed
20.
Zurück zum Zitat Wu S, Sun Z, Guo Z, Li P, Mao Q, Tang Y, Chen H, Peng H, Wang S, Cao Y. The effectiveness of blood-activating and stasis-transforming traditional Chinese medicines (BAST) in lung cancer progression-a comprehensive review. J Ethnopharmacol. 2023;314:116565.PubMed Wu S, Sun Z, Guo Z, Li P, Mao Q, Tang Y, Chen H, Peng H, Wang S, Cao Y. The effectiveness of blood-activating and stasis-transforming traditional Chinese medicines (BAST) in lung cancer progression-a comprehensive review. J Ethnopharmacol. 2023;314:116565.PubMed
21.
Zurück zum Zitat Yan F, Li F, Liu J, Ye S, Zhang Y, Jia J, Li H, Chen D, Mo X. The formulae and biologically active ingredients of Chinese herbal medicines for the treatment of atopic dermatitis. Biomed Pharmacother. 2020;127:110142.PubMed Yan F, Li F, Liu J, Ye S, Zhang Y, Jia J, Li H, Chen D, Mo X. The formulae and biologically active ingredients of Chinese herbal medicines for the treatment of atopic dermatitis. Biomed Pharmacother. 2020;127:110142.PubMed
22.
Zurück zum Zitat Stojic J, Hagemann C, Haas S, Herbold C, Kuhnel S, Gerngras S, Roggendorf W, Roosen K, Vince GH. Expression of matrix metalloproteinases MMP-1, MMP-11 and MMP-19 is correlated with the WHO-grading of human malignant gliomas. Neurosci Res. 2008;60(1):40–9.PubMed Stojic J, Hagemann C, Haas S, Herbold C, Kuhnel S, Gerngras S, Roggendorf W, Roosen K, Vince GH. Expression of matrix metalloproteinases MMP-1, MMP-11 and MMP-19 is correlated with the WHO-grading of human malignant gliomas. Neurosci Res. 2008;60(1):40–9.PubMed
23.
Zurück zum Zitat Zhang Y, Zhan H, Xu W, Yuan Z, Lu P, Zhan L, Li Q. Upregulation of matrix metalloproteinase-1 and proteinase-activated receptor-1 promotes the progression of human gliomas. Pathol Res Pract. 2011;207(1):24–9.PubMed Zhang Y, Zhan H, Xu W, Yuan Z, Lu P, Zhan L, Li Q. Upregulation of matrix metalloproteinase-1 and proteinase-activated receptor-1 promotes the progression of human gliomas. Pathol Res Pract. 2011;207(1):24–9.PubMed
24.
Zurück zum Zitat Anand M, Van Meter TE, Fillmore HL. Epidermal growth factor induces matrix metalloproteinase-1 (MMP-1) expression and invasion in glioma cell lines via the MAPK pathway. J Neurooncol. 2011;104(3):679–87.PubMed Anand M, Van Meter TE, Fillmore HL. Epidermal growth factor induces matrix metalloproteinase-1 (MMP-1) expression and invasion in glioma cell lines via the MAPK pathway. J Neurooncol. 2011;104(3):679–87.PubMed
25.
Zurück zum Zitat Tung JN, Ko CP, Yang SF, Cheng CW, Chen PN, Chang CY, Lin CL, Yang TF, Hsieh YH, Chen KC. Inhibition of pentraxin 3 in glioma cells impairs proliferation and invasion in vitro and in vivo. J Neurooncol. 2016;129(2):201–9.PubMed Tung JN, Ko CP, Yang SF, Cheng CW, Chen PN, Chang CY, Lin CL, Yang TF, Hsieh YH, Chen KC. Inhibition of pentraxin 3 in glioma cells impairs proliferation and invasion in vitro and in vivo. J Neurooncol. 2016;129(2):201–9.PubMed
26.
Zurück zum Zitat Liang C, Shangguan J, Yang L, Guo S. Downregulation of astrocyte elevated gene-1 expression inhibits the development of vasculogenic mimicry in gliomas. Exp Ther Med. 2021;21(1):22.PubMed Liang C, Shangguan J, Yang L, Guo S. Downregulation of astrocyte elevated gene-1 expression inhibits the development of vasculogenic mimicry in gliomas. Exp Ther Med. 2021;21(1):22.PubMed
27.
Zurück zum Zitat Liang C, Yang L, Guo S. All-trans retinoic acid inhibits migration, invasion and proliferation, and promotes apoptosis in glioma cells in vitro. Oncol Lett. 2015;9(6):2833–8.PubMedPubMedCentral Liang C, Yang L, Guo S. All-trans retinoic acid inhibits migration, invasion and proliferation, and promotes apoptosis in glioma cells in vitro. Oncol Lett. 2015;9(6):2833–8.PubMedPubMedCentral
28.
Zurück zum Zitat Liang C, Yang L, Guo SW, Li RC. Downregulation of astrocyte elevated gene-1 expression combined with all-trans retinoic acid inhibits development of vasculogenic mimicry and angiogenesis in glioma. Curr Med Sci. 2022;42(2):397–406.PubMed Liang C, Yang L, Guo SW, Li RC. Downregulation of astrocyte elevated gene-1 expression combined with all-trans retinoic acid inhibits development of vasculogenic mimicry and angiogenesis in glioma. Curr Med Sci. 2022;42(2):397–406.PubMed
29.
Zurück zum Zitat Aroui S, Najlaoui F, Chtourou Y, Meunier AC, Laajimi A, Kenani A, Fetoui H. Naringin inhibits the invasion and migration of human glioblastoma cell via downregulation of MMP-2 and MMP-9 expression and inactivation of p38 signaling pathway. Tumour Biol. 2016;37(3):3831–9.PubMed Aroui S, Najlaoui F, Chtourou Y, Meunier AC, Laajimi A, Kenani A, Fetoui H. Naringin inhibits the invasion and migration of human glioblastoma cell via downregulation of MMP-2 and MMP-9 expression and inactivation of p38 signaling pathway. Tumour Biol. 2016;37(3):3831–9.PubMed
30.
Zurück zum Zitat Gao Y, Ma X, Lu H, Xu P, Xu C. PLAU is associated with cell migration and invasion and is regulated by transcription factor YY1 in cervical cancer. Oncol Rep. 2023;49(2):25.PubMed Gao Y, Ma X, Lu H, Xu P, Xu C. PLAU is associated with cell migration and invasion and is regulated by transcription factor YY1 in cervical cancer. Oncol Rep. 2023;49(2):25.PubMed
31.
Zurück zum Zitat Li J, Fan H, Zhou X, Xiang Y, Liu Y. Prognostic significance and gene co-expression network of PLAU and PLAUR in gliomas. Front Oncol. 2021;11:602321.PubMed Li J, Fan H, Zhou X, Xiang Y, Liu Y. Prognostic significance and gene co-expression network of PLAU and PLAUR in gliomas. Front Oncol. 2021;11:602321.PubMed
32.
Zurück zum Zitat Kaphle P, Li Y, Yao L. The mechanical and pharmacological regulation of glioblastoma cell migration in 3D matrices. J Cell Physiol. 2019;234(4):3948–60.PubMed Kaphle P, Li Y, Yao L. The mechanical and pharmacological regulation of glioblastoma cell migration in 3D matrices. J Cell Physiol. 2019;234(4):3948–60.PubMed
33.
Zurück zum Zitat Silva M, Videira PA, Sackstein R. E-selectin ligands in the human mononuclear phagocyte system: implications for infection, inflammation, and immunotherapy. Front Immunol. 2017;8:1878.PubMed Silva M, Videira PA, Sackstein R. E-selectin ligands in the human mononuclear phagocyte system: implications for infection, inflammation, and immunotherapy. Front Immunol. 2017;8:1878.PubMed
34.
Zurück zum Zitat Kang SA, Blache CA, Bajana S, Hasan N, Kamal M, Morita Y, Gupta V, Tsolmon B, Suh KS, Gorenstein DG, et al. The effect of soluble E-selectin on tumor progression and metastasis. BMC Cancer. 2016;16:331.PubMedPubMedCentral Kang SA, Blache CA, Bajana S, Hasan N, Kamal M, Morita Y, Gupta V, Tsolmon B, Suh KS, Gorenstein DG, et al. The effect of soluble E-selectin on tumor progression and metastasis. BMC Cancer. 2016;16:331.PubMedPubMedCentral
35.
Zurück zum Zitat Borentain P, Carmona S, Mathieu S, Jouve E, El-Battari A, Gerolami R. Inhibition of E-selectin expression on the surface of endothelial cells inhibits hepatocellular carcinoma growth by preventing tumor angiogenesis. Cancer Chemother Pharmacol. 2016;77(4):847–56.PubMed Borentain P, Carmona S, Mathieu S, Jouve E, El-Battari A, Gerolami R. Inhibition of E-selectin expression on the surface of endothelial cells inhibits hepatocellular carcinoma growth by preventing tumor angiogenesis. Cancer Chemother Pharmacol. 2016;77(4):847–56.PubMed
36.
Zurück zum Zitat Gu Q, Chen X, Zhou L, Liu X. Exosome EpCAM promotes the metastasis of glioma by targeting the CD44 signaling molecule on the surface of glioma cells. Adv Clin Exp Med. 2020;29(11):1277–82.PubMed Gu Q, Chen X, Zhou L, Liu X. Exosome EpCAM promotes the metastasis of glioma by targeting the CD44 signaling molecule on the surface of glioma cells. Adv Clin Exp Med. 2020;29(11):1277–82.PubMed
37.
Zurück zum Zitat Hao X, Zu M, Ning J, Zhou X, Gong Y, Han X, Meng Q, Li D, Ding S. Antitumor effect of luteolin proven by patient-derived organoids of gastric cancer. Phytother Res. 2023; Online ahead of print. Hao X, Zu M, Ning J, Zhou X, Gong Y, Han X, Meng Q, Li D, Ding S. Antitumor effect of luteolin proven by patient-derived organoids of gastric cancer. Phytother Res. 2023; Online ahead of print.
38.
Zurück zum Zitat Aljohani H, Khodier AE, Al-Gayyar MM. Antitumor activity of luteolin against ehrlich solid carcinoma in rats via blocking Wnt/beta-Catenin/SMAD4 pathway. Cureus. 2023;15(5):e39789.PubMedPubMedCentral Aljohani H, Khodier AE, Al-Gayyar MM. Antitumor activity of luteolin against ehrlich solid carcinoma in rats via blocking Wnt/beta-Catenin/SMAD4 pathway. Cureus. 2023;15(5):e39789.PubMedPubMedCentral
39.
Zurück zum Zitat You Y, Wang R, Shao N, Zhi F, Yang Y. Luteolin suppresses tumor proliferation through inducing apoptosis and autophagy via MAPK activation in glioma. Onco Targets Ther. 2019;12:2383–96.PubMedPubMedCentral You Y, Wang R, Shao N, Zhi F, Yang Y. Luteolin suppresses tumor proliferation through inducing apoptosis and autophagy via MAPK activation in glioma. Onco Targets Ther. 2019;12:2383–96.PubMedPubMedCentral
40.
Zurück zum Zitat Yi C, Li G, Ivanov DN, Wang Z, Velasco MX, Hernandez G, Kaundal S, Villarreal J, Gupta YK, Qiao M, et al. Luteolin inhibits Musashi1 binding to RNA and disrupts cancer phenotypes in glioblastoma cells. Rna Biol. 2018;15(11):1420–32.PubMedPubMedCentral Yi C, Li G, Ivanov DN, Wang Z, Velasco MX, Hernandez G, Kaundal S, Villarreal J, Gupta YK, Qiao M, et al. Luteolin inhibits Musashi1 binding to RNA and disrupts cancer phenotypes in glioblastoma cells. Rna Biol. 2018;15(11):1420–32.PubMedPubMedCentral
41.
Zurück zum Zitat Fernandez-Palanca P, Fondevila F, Mendez-Blanco C, Tunon MJ, Gonzalez-Gallego J, Mauriz JL. Antitumor effects of quercetin in hepatocarcinoma in vitro and in vivo models: a systematic review. Nutrients. 2019;11(12):2875.PubMedPubMedCentral Fernandez-Palanca P, Fondevila F, Mendez-Blanco C, Tunon MJ, Gonzalez-Gallego J, Mauriz JL. Antitumor effects of quercetin in hepatocarcinoma in vitro and in vivo models: a systematic review. Nutrients. 2019;11(12):2875.PubMedPubMedCentral
42.
Zurück zum Zitat Wu Q, Needs PW, Lu Y, Kroon PA, Ren D, Yang X. Different antitumor effects of quercetin, quercetin-3’-sulfate and quercetin-3-glucuronide in human breast cancer MCF-7 cells. Food Funct. 2018;9(3):1736–46.PubMed Wu Q, Needs PW, Lu Y, Kroon PA, Ren D, Yang X. Different antitumor effects of quercetin, quercetin-3’-sulfate and quercetin-3-glucuronide in human breast cancer MCF-7 cells. Food Funct. 2018;9(3):1736–46.PubMed
43.
Zurück zum Zitat Tamtaji OR, Razavi ZS, Razzaghi N, Aschner M, Barati E, Mirzaei H. Quercetin and glioma: which signaling pathways are involved? Curr Mol Pharmacol. 2022;15(7):962–8.PubMed Tamtaji OR, Razavi ZS, Razzaghi N, Aschner M, Barati E, Mirzaei H. Quercetin and glioma: which signaling pathways are involved? Curr Mol Pharmacol. 2022;15(7):962–8.PubMed
44.
Zurück zum Zitat Pan HC, Jiang Q, Yu Y, Mei JP, Cui YK, Zhao WJ. Quercetin promotes cell apoptosis and inhibits the expression of MMP-9 and fibronectin via the AKT and ERK signalling pathways in human glioma cells. Neurochem Int. 2015;80:60–71.PubMed Pan HC, Jiang Q, Yu Y, Mei JP, Cui YK, Zhao WJ. Quercetin promotes cell apoptosis and inhibits the expression of MMP-9 and fibronectin via the AKT and ERK signalling pathways in human glioma cells. Neurochem Int. 2015;80:60–71.PubMed
45.
Zurück zum Zitat Chen B, Li X, Wu L, Zhou D, Song Y, Zhang L, Wu Q, He Q, Wang G, Liu X, et al. Quercetin suppresses human glioblastoma migration and invasion via GSK3beta/beta-catenin/ZEB1 signaling pathway. Front Pharmacol. 2022;13:963614.PubMedPubMedCentral Chen B, Li X, Wu L, Zhou D, Song Y, Zhang L, Wu Q, He Q, Wang G, Liu X, et al. Quercetin suppresses human glioblastoma migration and invasion via GSK3beta/beta-catenin/ZEB1 signaling pathway. Front Pharmacol. 2022;13:963614.PubMedPubMedCentral
46.
Zurück zum Zitat Ersoz M, Erdemir A, Derman S, Arasoglu T, Mansuroglu B. Quercetin-loaded nanoparticles enhance cytotoxicity and antioxidant activity on C6 glioma cells. Pharm Dev Technol. 2020;25(6):757–66.PubMed Ersoz M, Erdemir A, Derman S, Arasoglu T, Mansuroglu B. Quercetin-loaded nanoparticles enhance cytotoxicity and antioxidant activity on C6 glioma cells. Pharm Dev Technol. 2020;25(6):757–66.PubMed
47.
Zurück zum Zitat Wang J, Fang X, Ge L, Cao F, Zhao L, Wang Z, Xiao W. Antitumor, antioxidant and anti-inflammatory activities of kaempferol and its corresponding glycosides and the enzymatic preparation of kaempferol. PLoS One. 2018;13(5):e0197563.PubMedPubMedCentral Wang J, Fang X, Ge L, Cao F, Zhao L, Wang Z, Xiao W. Antitumor, antioxidant and anti-inflammatory activities of kaempferol and its corresponding glycosides and the enzymatic preparation of kaempferol. PLoS One. 2018;13(5):e0197563.PubMedPubMedCentral
48.
Zurück zum Zitat Chen S, Ma J, Yang L, Teng M, Lai ZQ, Chen X, He J. Anti-glioblastoma activity of kaempferol via programmed cell death induction: involvement of autophagy and pyroptosis. Front Bioeng Biotechnol. 2020;8:614419.PubMedPubMedCentral Chen S, Ma J, Yang L, Teng M, Lai ZQ, Chen X, He J. Anti-glioblastoma activity of kaempferol via programmed cell death induction: involvement of autophagy and pyroptosis. Front Bioeng Biotechnol. 2020;8:614419.PubMedPubMedCentral
49.
Zurück zum Zitat Santos BL, Oliveira MN, Coelho PL, Pitanga BP, da Silva AB, Adelita T, Silva VD, Costa Mde F, El-Bacha RS, Tardy M, et al. Flavonoids suppress human glioblastoma cell growth by inhibiting cell metabolism, migration, and by regulating extracellular matrix proteins and metalloproteinases expression. Chem Biol Interact. 2015;242:123–38.PubMed Santos BL, Oliveira MN, Coelho PL, Pitanga BP, da Silva AB, Adelita T, Silva VD, Costa Mde F, El-Bacha RS, Tardy M, et al. Flavonoids suppress human glioblastoma cell growth by inhibiting cell metabolism, migration, and by regulating extracellular matrix proteins and metalloproteinases expression. Chem Biol Interact. 2015;242:123–38.PubMed
50.
Zurück zum Zitat Sharma V, Joseph C, Ghosh S, Agarwal A, Mishra MK, Sen E. Kaempferol induces apoptosis in glioblastoma cells through oxidative stress. Mol Cancer Ther. 2007;6(9):2544–53.PubMed Sharma V, Joseph C, Ghosh S, Agarwal A, Mishra MK, Sen E. Kaempferol induces apoptosis in glioblastoma cells through oxidative stress. Mol Cancer Ther. 2007;6(9):2544–53.PubMed
51.
Zurück zum Zitat Ferrer A, Altabella T, Arro M, Boronat A. Emerging roles for conjugated sterols in plants. Prog Lipid Res. 2017;67:27–37.PubMed Ferrer A, Altabella T, Arro M, Boronat A. Emerging roles for conjugated sterols in plants. Prog Lipid Res. 2017;67:27–37.PubMed
52.
Zurück zum Zitat Sharma N, Sharma A, Bhatia G, Landi M, Brestic M, Singh B, Singh J, Kaur S, Bhardwaj R. Isolation of phytochemicals from bauhinia variegata L. bark and their in vitro antioxidant and cytotoxic potential. Antioxidants (Basel). 2019;8(10):492.PubMed Sharma N, Sharma A, Bhatia G, Landi M, Brestic M, Singh B, Singh J, Kaur S, Bhardwaj R. Isolation of phytochemicals from bauhinia variegata L. bark and their in vitro antioxidant and cytotoxic potential. Antioxidants (Basel). 2019;8(10):492.PubMed
53.
Zurück zum Zitat Li S, Huang S, Peng SB. Overexpression of G protein-coupled receptors in cancer cells: involvement in tumor progression. Int J Oncol. 2005;27(5):1329–39.PubMed Li S, Huang S, Peng SB. Overexpression of G protein-coupled receptors in cancer cells: involvement in tumor progression. Int J Oncol. 2005;27(5):1329–39.PubMed
54.
Zurück zum Zitat O’Hayre M, Degese MS, Gutkind JS. Novel insights into G protein and G protein-coupled receptor signaling in cancer. Curr Opin Cell Biol. 2014;27:126–35.PubMedPubMedCentral O’Hayre M, Degese MS, Gutkind JS. Novel insights into G protein and G protein-coupled receptor signaling in cancer. Curr Opin Cell Biol. 2014;27:126–35.PubMedPubMedCentral
55.
Zurück zum Zitat Frenster JD, Kader M, Kamen S, Sun J, Chiriboga L, Serrano J, Bready D, Golub D, Ravn-Boess N, Stephan G, et al. Expression profiling of the adhesion G protein-coupled receptor GPR133 (ADGRD1) in glioma subtypes. Neurooncol Adv. 2020;2(1):1–9. Frenster JD, Kader M, Kamen S, Sun J, Chiriboga L, Serrano J, Bready D, Golub D, Ravn-Boess N, Stephan G, et al. Expression profiling of the adhesion G protein-coupled receptor GPR133 (ADGRD1) in glioma subtypes. Neurooncol Adv. 2020;2(1):1–9.
56.
Zurück zum Zitat Cherry AE, Stella N. G protein-coupled receptors as oncogenic signals in glioma: emerging therapeutic avenues. Neuroscience. 2014;278:222–36.PubMed Cherry AE, Stella N. G protein-coupled receptors as oncogenic signals in glioma: emerging therapeutic avenues. Neuroscience. 2014;278:222–36.PubMed
57.
Zurück zum Zitat Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ, Van Meir EG. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol. 2005;7(2):134–53.PubMedPubMedCentral Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ, Van Meir EG. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol. 2005;7(2):134–53.PubMedPubMedCentral
58.
Zurück zum Zitat Hu S, Cai J, Fang H, Chen Z, Zhang J, Cai R. RPS14 promotes the development and progression of glioma via p53 signaling pathway. Exp Cell Res. 2022;423(1):113451.PubMed Hu S, Cai J, Fang H, Chen Z, Zhang J, Cai R. RPS14 promotes the development and progression of glioma via p53 signaling pathway. Exp Cell Res. 2022;423(1):113451.PubMed
59.
Zurück zum Zitat Wang T, Li X, Sun SL. EX527, a Sirt-1 inhibitor, induces apoptosis in glioma via activating the p53 signaling pathway. Anticancer Drugs. 2020;31(1):19–26.PubMed Wang T, Li X, Sun SL. EX527, a Sirt-1 inhibitor, induces apoptosis in glioma via activating the p53 signaling pathway. Anticancer Drugs. 2020;31(1):19–26.PubMed
60.
62.
Zurück zum Zitat Liu P, Zhu C, Luo J, Lan S, Su D, Wang Q, Wei Z, Cui W, Xu C, Yang X. Par6 regulates cell cycle progression through enhancement of Akt/PI3K/GSK-3beta signaling pathway activation in glioma. FASEB J. 2020;34(1):1481–96.PubMed Liu P, Zhu C, Luo J, Lan S, Su D, Wang Q, Wei Z, Cui W, Xu C, Yang X. Par6 regulates cell cycle progression through enhancement of Akt/PI3K/GSK-3beta signaling pathway activation in glioma. FASEB J. 2020;34(1):1481–96.PubMed
63.
Zurück zum Zitat Zhao ZQ, Wu XJ, Cheng YH, Zhou YF, Ma XM, Zhang J, Heng XY, Feng F. TROAP regulates cell cycle and promotes tumor progression through Wnt/beta-Catenin signaling pathway in glioma cells. CNS Neurosci Ther. 2021;27(9):1064–76.PubMedPubMedCentral Zhao ZQ, Wu XJ, Cheng YH, Zhou YF, Ma XM, Zhang J, Heng XY, Feng F. TROAP regulates cell cycle and promotes tumor progression through Wnt/beta-Catenin signaling pathway in glioma cells. CNS Neurosci Ther. 2021;27(9):1064–76.PubMedPubMedCentral
64.
Zurück zum Zitat Wypych D, Pomorski P. Calcium signaling in glioma cells: the role of nucleotide receptors. Adv Exp Med Biol. 2020;1202:67–86.PubMed Wypych D, Pomorski P. Calcium signaling in glioma cells: the role of nucleotide receptors. Adv Exp Med Biol. 2020;1202:67–86.PubMed
65.
66.
Zurück zum Zitat Subburayan K, Thayyullathil F, Pallichankandy S, Rahman A, Galadari S. Par-4-dependent p53 up-regulation plays a critical role in thymoquinone-induced cellular senescence in human malignant glioma cells. Cancer Lett. 2018;426:80–97.PubMed Subburayan K, Thayyullathil F, Pallichankandy S, Rahman A, Galadari S. Par-4-dependent p53 up-regulation plays a critical role in thymoquinone-induced cellular senescence in human malignant glioma cells. Cancer Lett. 2018;426:80–97.PubMed
67.
Zurück zum Zitat Jinno-Oue A, Shimizu N, Hamada N, Wada S, Tanaka A, Shinagawa M, Ohtsuki T, Mori T, Saha MN, Hoque AS, et al. Irradiation with carbon ion beams induces apoptosis, autophagy, and cellular senescence in a human glioma-derived cell line. Int J Radiat Oncol Biol Phys. 2010;76(1):229–41.PubMed Jinno-Oue A, Shimizu N, Hamada N, Wada S, Tanaka A, Shinagawa M, Ohtsuki T, Mori T, Saha MN, Hoque AS, et al. Irradiation with carbon ion beams induces apoptosis, autophagy, and cellular senescence in a human glioma-derived cell line. Int J Radiat Oncol Biol Phys. 2010;76(1):229–41.PubMed
68.
Zurück zum Zitat Adkins DE, Khachane AN, McClay JL, Aberg K, Bukszar J, Sullivan PF, van den Oord EJ. SNP-based analysis of neuroactive ligand-receptor interaction pathways implicates PGE2 as a novel mediator of antipsychotic treatment response: data from the CATIE study. Schizophr Res. 2012;135(1–3):200–1.PubMed Adkins DE, Khachane AN, McClay JL, Aberg K, Bukszar J, Sullivan PF, van den Oord EJ. SNP-based analysis of neuroactive ligand-receptor interaction pathways implicates PGE2 as a novel mediator of antipsychotic treatment response: data from the CATIE study. Schizophr Res. 2012;135(1–3):200–1.PubMed
69.
Zurück zum Zitat Ozawa A, Kadowaki E, Horio T, Sakaue M. Acetylcholine suppresses the increase of glia fibrillary acidic protein expression via acetylcholine receptors in cAMP-induced astrocytic differentiation of rat C6 glioma cells. Neurosci Lett. 2019;698:146–53.PubMed Ozawa A, Kadowaki E, Horio T, Sakaue M. Acetylcholine suppresses the increase of glia fibrillary acidic protein expression via acetylcholine receptors in cAMP-induced astrocytic differentiation of rat C6 glioma cells. Neurosci Lett. 2019;698:146–53.PubMed
70.
Zurück zum Zitat Lan YL, Wang X, Xing JS, Yu ZL, Lou JC, Ma XC, Zhang B. Anti-cancer effects of dopamine in human glioma: involvement of mitochondrial apoptotic and anti-inflammatory pathways. Oncotarget. 2017;8(51):88488–500.PubMedPubMedCentral Lan YL, Wang X, Xing JS, Yu ZL, Lou JC, Ma XC, Zhang B. Anti-cancer effects of dopamine in human glioma: involvement of mitochondrial apoptotic and anti-inflammatory pathways. Oncotarget. 2017;8(51):88488–500.PubMedPubMedCentral
71.
Zurück zum Zitat Tsuchioka M, Takebayashi M, Hisaoka K, Maeda N, Nakata Y. Serotonin (5-HT) induces glial cell line-derived neurotrophic factor (GDNF) mRNA expression via the transactivation of fibroblast growth factor receptor 2 (FGFR2) in rat C6 glioma cells. J Neurochem. 2008;106(1):244–57.PubMed Tsuchioka M, Takebayashi M, Hisaoka K, Maeda N, Nakata Y. Serotonin (5-HT) induces glial cell line-derived neurotrophic factor (GDNF) mRNA expression via the transactivation of fibroblast growth factor receptor 2 (FGFR2) in rat C6 glioma cells. J Neurochem. 2008;106(1):244–57.PubMed
72.
Zurück zum Zitat Zhang L, Wang D, Han X, Tang F, Gao D. Mechanism of methylation and acetylation of high GDNF transcription in glioma cells: a review. Heliyon. 2019;5(6):e01951.PubMedPubMedCentral Zhang L, Wang D, Han X, Tang F, Gao D. Mechanism of methylation and acetylation of high GDNF transcription in glioma cells: a review. Heliyon. 2019;5(6):e01951.PubMedPubMedCentral
Metadaten
Titel
Network pharmacology -based study on the mechanism of traditional Chinese medicine in the treatment of glioblastoma multiforme
verfasst von
Chen Liang
Binbin Zhang
Ruichun Li
Shiwen Guo
Xiaoxuan Fan
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2023
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-023-04174-7

Weitere Artikel der Ausgabe 1/2023

BMC Complementary Medicine and Therapies 1/2023 Zur Ausgabe