Abstract
Statistics provided by GLOBOCAN list gastric cancer as the sixth most common, with a mortality ranking of third highest for the year 2020. In China, a herb called Rabdosia rubescens (Hemsl.) H.Hara, has been used by local residents for the treatment of digestive tract cancer for hundreds of years. Oridonin, the main ingredient of the herb, has a curative effect for gastric cancer, but the mechanism has not been previously clarified. This study mainly aimed to investigate the role of TNF‐alpha/Androgen receptor/TGF‐beta signalling pathway axis in mediating the proliferation inhibition of oridonin on gastric cancer SGC‐7901 cells. MTT assay, cell morphology observation assay and fluorescence assay were adopted to study the efficacy of oridonin on cell proliferation. The network pharmacology was used to predict the pathway axis regulated by oridonin. Western blot assay was adopted to verify the TNF‐α/Androgen receptor/TGF‐β signalling pathway axis regulation on gastric cancer by oridonin. The results showed Oridonin could inhibit the proliferation of gastric cancer cells, change cell morphology and cause cell nuclear fragmentation. A total of 11signaling pathways were annotated by the network pharmacology, among them, Tumour necrosis factor alpha (TNF‐α) signalling pathway, androgen receptor (AR) signalling pathway and transforming growth factor (TGF‐β) signalling pathway account for the largest proportion. Oridonin can regulate the protein expression of the three signalling pathways, which is consistent with the results predicted by network pharmacology. These findings indicated that oridonin can inhibit the proliferation of gastric cancer SGC‐7901 cells by regulating the TNF‐α /AR /TGF‐β signalling pathway axis.
Keywords: AR signalling pathway, gastric cancer, oridonin, TGF‐β signalling pathway, TNF‐α signalling pathway
1. INTRODUCTION
Rabdosia rubescens (Hemsl.) H.Hara, a small shrubs of the genus Camellia in the labiaceae, is naturally distributed in the Yellow River and Yangtze River basins of China. Local residents have been treating cancer with Rabdosia rubescens (Hemsl.) H.Hara for decades in the Taihang and Wangwu mountain areas, located in the Henan Province in the People's republic of China. In 1972, Chinese researchers discovered that Rabdosia rubescens (Hemsl.) H.Hara has a unique anti‐cancer effect on cardiac cancer, liver cancer and oesophageal cancer. In the 1980s, further research and development of Rabdosia rubescens (Hemsl.) H.Hara was carried out and the main anti‐cancer components of Rabdosia rubescens (Hemsl.) H.Hara were identified as oridonin(purity >99.5%, buy from National Institutes for Food and Drug Control). The 2000 and the more recent 2020 edition of ‘Pharmacopoeia of the People's Republic of China’ officially included Rabdosia rubescens (Hemsl.) H.Hara. Rabdosia rubescens (Hemsl.) H.Hara has been shown to remove heat and help detoxify, promote blood circulation, relieve pain and has also been used for the treatment of sore throats and lumps. Meanwhile, the 2020 edition of pharmacopoeia has also included Rabdosia rubescens (Hemsl.) H.Hara based tablets. 1
Oridonin (Figure 2A), a 7,20‐Epoxy‐ent‐Kauren Diterpenoid, is an effective ingredient with anti‐cancer activity. 2 , 3 The chemical formula is C20H28O6, the molecular weight is 364.44, the melting point is between 248 and 250°C, the compound is colourless or light yellow and displays a physical structure similar to needle‐like crystals. The compound tastes bitter, is slightly soluble in water and completely soluble in methanol, ethanol, dimethyl sulfoxide and other organic solvents. The enkeone system, the D‐ring core pharmacophore, is closely related to the biological activity of ordonin and its anticancer activity may be lost if the enkeone system is removed. 4 In order to improve the bioavailability of oridonin, a variety of preparations such as liposomes 5 , 6 microspheres 7 and nanoparticles 8 , 9 have been developed.
FIGURE 2.
Effect of oridonin on proliferation of SGC‐7901 cells. (A) 2D chemical structural formula of oridonin. (B) The growth curve. (C) Cell morphology. a. Control; b. 0.35 μM ADR; c 30 μM oridonin; d 60 μM oridonin; e 90 μM oridonin. (D) Cell nuclear morphology. a. Control; b. 0.35 μM ADR; c. 30 μM oridonin; d. 60 μM oridonin; e. 90 μM oridonin.
Oridonin has previously been investigated for its therapeutic effects on the following types of caners: colon, 2 , 3 , 6 , 10 , 11 breast, 12 , 13 pancreatic, 14 , 15 lung, 16 , 17 , 18 gallbladder, 19 prostate, 20 ovarian, 21 oesophageal, 22 oral, 23 , 24 bladder, 25 liver 26 and other cancers. It has been reported that the inhibitory effect of oridonin on proliferation of gastric cancer is mainly related to inducing apoptosis and promoting autophagy, 27 , 28 , 29 without further in‐depth discussions. Our previous study found that oridonin can block the G2/M phase of gastric cancer cells 30 and induce apoptosis 31 by affecting the expression of cyclinD1/CDK4 and cyclinA/CDK2 proteins.
Network pharmacology is a theory developed by Hopkins, 32 , 33 a British pharmacologist, in 2007 and is considered a new interdisciplinary subject. Based on systems biology and multidirectional pharmacology, network pharmacology gives a new idea for drug research from the perspective of a multi‐target strategy. 11 Through the analysis and integration of big data, potential drug targets and signalling pathways can be predicted more comprehensively. In this study, we first confirmed that oridonin could inhibit the proliferation of gastric cancer cells. Then, the network pharmacology method was adopted to predict the signal pathway of oridonin for gastric cancer. Finally, experimental studies were carried out to study the predicted pathways and comprehensively reveal the mechanism of action of oridonin on gastric cancer. The research flow chart of this paper is shown in Figure 1.
FIGURE 1.
The schematic diagram of the research methodology and workflow of the present study of oridonin acting on gastric cancer.
2. MATERIALS AND METHODS
2.1. Cell line
Human gastric cancer SGC‐7901 cells were obtained from Heilongjiang Provincial Key Laboratory of Tumour Prevention and Antitumor Drug Research.
2.2. Chemicals and reagents
Oridonin (>98%) was purchased from National Institutes for Food and Drug Control of China. Adriamycin was purchased from Pharmacia Italia S.p.A., Gaggiano, Italy. Dimethyl sulfoxide (DMSO), trypsin, Tris, glycine, acrylamide, methylene diacrylamide were purchased from Sigma‐Aldrich. RPMI‐1640 cell culture medium was purchased from Gibco. Fetal bovine serum (FBS) was obtained from Sijiqing Hangzhou Bio Engineering Co., Ltd. Antibodies for caspase‐8, caspase‐3, caspase‐7, MEKK1, MEK7, JNK, IKBα, NF‐κB, AR, TGF‐β, Smad 2/3, TNF‐α, STEAP, Smad‐4, β‐actin and the secondary antibodies were purchased from Wanleibio.
2.3. In vitro cell viability assay
SGC‐7901 cells at the logarithmic growth stage were added into a 96‐well plate with 3.0 × 103 cells/well. After incubation in the carbon dioxide incubator with 5% CO2 for 24 h, oridonin were added to cell suspensions and then incubated for another 72 h. This was followed by adding 0.5 mg/mL MTT solution to the suspensions, then incubated for 4 h in the carbon dioxide incubator. After this time, the liquid was poured out and 150 μL of DMSO was added to the suspensions, after which the optical density (OD) was detected by a microplate reader. The wavelength used for measurement was 570 nm, the IC50 value and inhibition rate were calculated. 31 , 34
2.4. Cell morphological and nuclei morphologicalchanges
Cell morphology was observed by inverted microscope after treating cells with oridonin. Morphological changes of the cell nuclei were examined by fluorescence analysis with a Hoechst 33258 stain solution. SGC‐7901 cells treated with oridonin for 24 h were fixed in 4% paraformaldehyde. Then, the cells were exposed to the Hoechst33258 stain solution for 30 min. Finally, the cells were washed three times in cold PBS, and the morphology of the cell nuclei was observed using a fluorescence microscope.
2.5. Analysis of oridonin on gastric cancer by network pharmacology
2.5.1. Potential targets of oridonin
The 2D (Figure 2A) and 3D structures of oridonin were obtained from PubChem (https://pubchem.ncbi.nlm.nih.gov/). The mol2 file of oridonin was obtained from ZINC (https://zinc.docking.org). The structural information was used to look for potential targets of oridonin from PharmMapper (http://lilab.ecust.edu.cn/, updated May 29, 2018). Uniport (http://www.uniprot.org/) was adopted to standardize the nomenclature.
2.5.2. Related targets of gastric cancer
The gastric cancer‐related targets were obtained from five databases: Drugbank (https://www.drugbank.ca/, updated April 2, 2018), The Genetic Association Database (GAD, https://geneticassociationdb.nih.gov, updated August 18, 2014), Kyoto Encyclopedia of Genes and Genomes(KEGG, http://www.genome.jp/kegg, updated June 1, 2018), Online Mendelian Inheritance in Man (OMIM, http://www.omim.org, updated July 1, 2018), the Therapeutic Target Database(TTD, https://db.idrblab.org/ttd/, updated September 15, 2017).
2.5.3. Protein–protein interaction analysis(PPI) and hub gene screening
The PPI of targets was generated by Bisogenet, a plug‐in of Cytoscape. Hub gene was screened using six parameters, including betweenness centrality (BC), closeness centrality (CC), degree centrality (DC), eigenvector centrality (EC), local average connectivity‐based method (LAC) and network centrality (NC).
2.5.4. Pathway and biological functional enrichment analysis of Hub targets
Enrichment analysis of the Hub targets was performed by ClueGo, a plug‐in of Cytoscape (https://cytoscape.org), pV <0.05, Pathway = wikipathway. Visualisation was organized and imported using Cytoscape. Biological processes (BP) and pathways were visualized to show the relations between oridonin and gastric cancer targets.
2.6. Western blot assay 31 , 35
SGC‐7901 cells treated with oridonin were collected, and lysed with buffer (50 mM Tris‐Cl, pH 8.0, 120 mM NaCl, 50 mM NaF, 200 μM sodium vanadate, 0.5% NP‐40, 10 mM PMSF, 2 μg/mL aprotinin 0.2 μL, 10 μg/mL leupeptin 10 μL). The solution of the cell being lysed was centrifuged at 12000 × g at 4°C and the supernatant was collected. Protein concentration of the lysates were detected by Bradford assay. SDS‐PAGE was performed until the protein bands were completely separated, and then the proteins were transferred onto NC membranes. After blocking, the corresponding primary and secondary antibodies were added. The bands were measured by Gel Imaging System (GE, ImageQuant LAS500), and all the bands were repeated for three times.
2.7. Statistical analysis
Data are represented by mean ± standard deviation. The statistical analysis of data were calculated using Student's t‐test. p < 0.05 is believed as statistically significant.
3. RESULTS
3.1. Inhibition effect of oridonin on SGC‐7901 cells proliferation
Oridonin showed an inhibitory effect on SGC‐7901 cells and the proliferation inhibition rate increased gradually with the extension of drug treatment time (24, 48, 72 h) (Table 1, Figure 2B). The IC50 value was 6.55 × 101 μM (Table 2). Cell morphology studies showed that cells in the control group showed up as irregular polygons with clear edges, large cell density, uniform arrangement and almost no suspended cells present (Figure 2C a). However, with the increase of the dosage of oridonin, the number of living cells (polygonal cells) gradually decreases and the number of dead cells (round cells) gradually increased (Figure 2C c,d,e). The morphology of the nuclei in the control was irregular oval, with moderate fluorescence intensity (Figure 2D a), while the cells nuclei treated with oridonin showed irregular shapes such as pyknosis, fragmentation, renal shape et al., and the fluorescence emitted by the cells is highlighted, which are characteristics of dying cells (Figure 2D c,d,e).
TABLE 1.
Effect of oridonin on inhibition rate of the gastric cancer SGC‐7901 cell(x ± s, n = 6).
Group | Dosage (μΜ) | Inhibition rate (%) | ||
---|---|---|---|---|
24 h | 48 h | 72 h | ||
Control | 0 | — | — | — |
Oridonin | 40 | 10.49 ± 1.83** | 24.20 ± 5.13** | 19.31 ± 1.57** |
50 | 12.29 ± 3.51** | 28.68 ± 4.39** | 28.44 ± 1.36** | |
60 | 14.22 ± 3.66** | 33.87 ± 4.86** | 37.22 ± 3.33** | |
70 | 15.25 ± 2.19** | 40.69 ± 3.63** | 48.71 ± 3.98** | |
80 | 15.83 ± 2.50** | 46.80 ± 7.81** | 63.52 ± 6.10** | |
90 | 21.94 ± 5.60** | 63.24 ± 5.56** | 81.80 ± 2.94** | |
100 | 29.47 ± 3.59** | 76.85 ± 2.94** | 89.78 ± 0.94** | |
110 | 40.03 ± 3.92** | 84.19 ± 1.20** | 91.26 ± 0.67** | |
120 | 49.81 ± 1.43** | 85.68 ± 0.54** | 91.61 ± 0.31** |
Note: compared with the control.
p < 0.05
p < 0.01.
TABLE 2.
The IC50 of oridonin on the gastric cancer SGC‐7901 cell.
Groups | IC50 (μM) |
---|---|
Adr | 1.88 × 10−1 |
Oridonin | 6.55 × 101 |
3.2. Potential targets of oridonin
The Pharmapper Database was adopted to identify the potential targets of oridonin, and the top 300 targets were selected as relevant. After processing, a total of 293 targets meeting the standards were obtained (Table S1).
3.3. Gastric cancer targets
Through five different databases, 180 targets relating to gastric cancer was identified (Table S3). A total of 18 targets were found by Drugbank, 56 targets were sought out from GAD, 103 targets were got from KEGG, 12 targets were mined from OMIM and 7 targets were fished using TTD.
3.4. Common targets fishing of oridonin in the treatment of gastric cancer and Hub target screening
By means of PPI, 7046 potential targets of oridonin (Figure3Aa, Table S2) and 5613 targets of gastric cancer related diseases (Figure 3Ab, Table S4) were obtained. Then a total of 4342 common targets of oridonin for gastric cancer were fished (Table S5). DC, BC, CC, EC, LAC, NC and other parameters were adopted to find 4342 common targets of oridonin for gastric cancer successively and finally 358 oridonin Hub targets for gastric cancer were obtained (Figure 3Ae, Table S6).
FIGURE 3.
The fishing of Hub targets and the enrichment analysis of oridonin on gastric cancer. (A) The fishing of Hub targets. a. 7046 targets‐related of oridonin with 166346 edges; b 5613 target‐related of gastric cancer with 139501 edges; c 432 common targets of oridonin on gastric cancer with 119558 edges; d 1063 common targets (DC > 66) with 48485 edges; e 385 Hub targets with 15418 edges. (B) the pathway enrichment analysis of the Hub targets. a. Dot plot. b. Pie plot.
3.5. Oridonin‐Targets‐Gastric Cancer network construction
A network depicting Oridonin‐Targets‐Gastric Cancer was constructed to observe the anti‐gastric cancer mechanism of oridonin (Figure 4). The network consists of 453 nodes (273 oridonin targets, 160 gastric cancer‐related targets and 20 overlapping targets) and 473 edges. The 20 overlapped targets are the potential targets for the treatment of gastric cancer with oridonin, which suggested a possible mechanism for the treatment of on gastric cancer with oridonin.
FIGURE 4.
Oridonin‐target‐gastric Cancer networks. oridonin targets (red), gastric cancer targets (yellow), and overlapped targets (purple) accounted for 273, 160 and 20, respectively.
3.6. Pathway enrichment analysis of the Hub targets
The 358 Hub targets of oridonin on gastric cancer were adopted to fish related pathways by wikipathway. 11 related pathways were enriched, among which the three pathways with the largest weight were TNF‐α signalling pathway, androgen receptor signalling pathway and TGF‐β signalling pathway (Figure 3B b). The results showed the anti‐gastric cancer effect of oridonin is potentially associated with the three signalling pathways.
3.7. Effects of oridonin on TNF‐α signalling pathway in SGC‐7901 cells
After oridonin intervention, the expression of TNF‐α protein increased with the increase of oridonin dosage (Figure 5A). The protein expressions of caspase‐8, caspase‐3 and caspase‐7 decreased and cleaved‐caspase‐8, cleaved‐caspase‐3, and cleaved‐caspase‐7 protein expression levels increased with an increasing dosage. These results suggest that oridonin can activate intracellular expression of cleaved‐caspase‐8, cleaved‐caspase‐3 and cleaved‐caspase‐7 proteins (Figure 5B,C).
FIGURE 5.
Effects of oridonin on expression of TNF‐α signalling pathway related proteins in human gastric cancer SGC‐7901 cells. (A)TNF‐α protein expression. (B) caspase‐8 protein expression. (C) caspase‐3 and caspase‐7 protein expression. (D) MEKK1, MEK7 and JNK protein expression; (E) NF‐κΒ protein expression.
The expression of MEK7 protein did not change significantly, while the expression of MEKK1, p‐MEK7 and p‐JNK protein gradually increased and the expression of JNK protein gradually decreased with the increase of oridonin dosage (Figure 5D).With the increase of oridonin dosage, the protein expression of IKBα and NF‐κB decreased gradually, while the protein expression of p‐IKB α increased gradually (Figure 5E).
3.8. Effects of oridonin on AR signalling pathway in SGC‐7901 cells
The protein expression of AR in cytoplasm was increased, while it was decreased in the nucleus, and the protein expression of STEAP1 in cells was also decreased, suggesting that oridonin could promote the flow of AR protein in the nucleus to the cytoplasm. It demonstrated the translocation of AR was induced by oridonin (Figure 6).
FIGURE 6.
Effects of oridonin on expression of AR signalling pathway related proteins in human gastric cancer SGC‐7901 cells.
3.9. Effects of oridonin on the TGF‐β signalling pathway in SGC‐7901 cells
The expression levels of TGF‐β and p‐Smad 2/3 protein were decreased and the Smad4 protein were increased, but the Smad2/3 protein were not changed by oridonin, The results showed that oridonin could regulate down the TGF‐β expression, inhibiting the activation of Smad2/3 protein and activating Smad4 protein (Figure 7).
FIGURE 7.
Effects of oridonin on expression of TGF‐β signalling pathway related proteins in human gastric cancer SGC‐7901 cells. (A) TGF‐β protein expression. (B) Smad2/3, p‐Smad2/3 and Smad4 protein expression.
4. DISCUSSION
Rabdosia rubescens (Hemsl.) H.Hara has been used as a herb for many years in China. In Henan or Shanxi Provinces of China, local villagers often drink its decoction as a health promoting tea, which has a good therapeutic effect on chronic pharyngitis and tonsillitis. Villagers also treated cardia cancer by taking decoction of Rabdosia rubescens (Hemsl.) H.Hara for many years and had a good therapeutic effect. As one of the active components of Rabdosia rubescens (Hemsl.) H.Hara, oridonin has shown obvious anti‐cancer activity on various cancer cells, such as digestive system malignant tumours, 6 , 26 breast cancer 12 , 13 and prostate cancer, 20 to name just a few. Angela M. Wong et al. compared the activity of the extract of Rabdosia rubescens (Hemsl.) H.Hara and oridonin on the anti‐tumour. The results showed that five times the pure oridonin more than it in the extract was required to obtain equivalent proliferation inhibition, which showed that oridonin is one of the anti‐cancer active compound in the Rabdosia rubescens (Hemsl.) H.Hara. 36
At present, the relevant literature on the antitumor effects of oridonin are focused on the following aspects: inducing apoptosis by arresting the cell cycle, 37 inducing DNA damage, 38 , 39 inducing autophagy, 15 inhibiting tumour angiogenesis 3 and inhibiting the migration of tumour cells. 18 , 19 , 40 It is relatively single‐sided, and lacks comprehensive and multi‐angle research. The network pharmacology has broken the barriers in the study of traditional Chinese medicine, which makes it easier for us to apply the technology of experimental science to study traditional Chinese medicine. 34 It provides a good idea for comprehensively elucidating the mechanism of drug action for wider applications. 41
Oridonin can induce apoptosis of SGC‐7901 cells, inhibit cell proliferation and arrest cells in G2/M phase. 30 , 31 Other mechanisms however, still remain unclear. In this study, the results confirmed that oridonin can inhibit the proliferation of SGC‐7901 cells. Then, using the network pharmacology method, the proliferation of gastric cancer was inhibited by the oridonin through 11 signalling pathways, among which TNF‐α, AR and TGF‐β signalling pathways were the three most important signalling pathways. Finally, western blotting was adopted to determine the regulation of oridonin on the pathway axis composed of the three major signalling pathways and to explain the mechanism of oridonin in the treatment of gastric cancer.
A growth curve study found that oridonin could inhibit the proliferation of human gastric cancer SGC‐7901 cells. The IC50 value is 65.5 μM. The morphological study indicated that oridonin could damage the cell membrane and reduce the number of living cells. The observation of nuclear morphology proved that oridonin caused the typical nuclear morphology of cell death, such as nuclear fragmentation and dense staining.
In this study, through the study of network pharmacology, it was predicted that the proliferation inhibition effect of oridonin was related to the regulation of TNF‐α, AR and TGF‐β signalling pathways. Therefore, this finding was then experimentally verified in the following study.
TNF‐α is involved in many physiological and pathological processes, such as cellular proliferation or differentiation, regulation of immunologic, vascular invasion and destruction of tumour vasculature. 42 TNF‐α binds to TNFR to activate caspase‐8, which activates downstream caspase‐3 and caspase‐7 protein through a cascade reaction, thereby inducing apoptosis; NF‐κB usually binds to inhibitory factor IκB in an inactive state in cells. Stimulation of NF‐κB results in the degradation of IκB and activation of NF‐κB. JNK is activated by the pro‐inflammatory factor TNF‐α. The activation program is that MAPKKK activates MAPKK, and MAPKK activates MAPK, then MAPK activates JNK signalling pathway. 43 In this study, we found that oridonin up‐regulated TNF‐α protein expression and stimulated downstream caspase, JNK and NF‐κB signalling pathways. Activing caspase‐8, caspase‐3 and caspase‐7 can induce apoptosis. MAPK is an important signalling system that mediates cell biological responses. JNK is one of four MAPK signalling systems (ERK, JNK, P38, MAPK) 44 , 45 , 46 in mammalian cells. MAPK signal transduction is completed through the MAPKKK‐MAPKK‐MAPK pathway, while MEKK1 is the MAPKKK within the cytoplasm. Through the MAPKKK‐MAPKK mode, MKK7 is activated 47 and then the JNK pathway is activated. 48 In this study, the results showed that oridonin increased the expression of MEKK 1, p‐MEK 7 and p‐JNK proteins in cells, suggesting that TNF‐α activated MEKK 1 and further activated MEK 7 under the action of oridonin. MEK 7 specifically activated JNK and promoted apoptosis; The expression of p‐IKBα was up‐regulated and NF‐κB was down‐regulated with the increase of oridonin dosage, suggesting that phosphorylation of IKBα was induced after TNF‐α stimulation. IKBα was an inhibitory factor of NF‐κB, so the activity of NF‐κB was inhibited, and the activity of promoting proliferation and inhibiting apoptosis was decreased. These findings showed that the inhibitory effect of oridonin on SGC‐7901 cells is related to TNF‐α signalling pathway, which is achieved by regulating caspase family proteins, NF‐κB signalling family proteins and promoting the protein expression of JNK signalling pathway.
AR is the androgen receptor, and AR signalling pathway is related to many hormone‐dependent diseases such as prostate cancer, 49 breast cancer 50 , 51 , 52 and ovarian cancer, 53 as well as malignant tumours, for example gastric cancer, 54 , 55 lung cancer, 56 , 57 bladder cancer, 58 , 59 pancreatic cancer, 60 , 61 liver cancer 62 and kidney cancer. 63 The distribution of AR in cells is closely related to the survival of tumours. The combination of androgen and AR occurs in the nucleus, so the distribution and metastasis of AR in cytoplasm and nucleus are closely related to the proliferation of tumours. STEAP1 protein downstream of AR signalling pathway exists at the junction of cell membranes and acts as a ‘communicator’ transmitting information between cells. 64 , 65 The results showed that oridonin up‐regulated cytoplasmic AR protein expression, down‐regulated nuclear AR protein and downstream STEAP 1 protein. In other words, oridonin can regulate the distribution of AR between the cytoplasm and nucleus, inhibit the transport of AR from cytoplasm to nucleus, or promote the flow of AR from nucleus to cytoplasm and then affect the binding amount of androgen to AR in nucleus, thus affecting the proliferation of SGC‐7901 cells.
TGF‐β is a growth transformation factor, not only involved in tumour proliferation, 66 invasion, 67 , 68 migration, 69 adhesion, 70 , 71 induce cell cycle arrest 72 , 73 and apoptosis, 74 , 75 but also through up‐regulation of cell growth inhibitory factors and inflammatory factors, so as to achieve the purpose of inhibiting tumour proliferation. Smads are intracellular signal transduction molecules, which can transfer TGF‐β signal from the cell membrane to the nucleus. When its function is abnormal, TGF‐β signal transduction will also be affected. TGF‐β binds to its receptor and phosphorylates. The activated TβRI can specifically recognize downstream Smad2 and Smad3, bind to and activate them, and the activated Smad2 and Smad3 dissociate from TβRI and then form heterologous complex with tumour suppressor gene Smad4. It is then transferred into the nucleus to affect the expression of target genes. 76 , 77 Oridonin could down‐regulate TGF‐β and p‐smad2/3 protein expression and up‐regulate Smad4 protein expression, thereby reducing downstream Smad2/3 activity, activating the Smad4 protein and inhibiting the proliferation of cancer cells. 78 Hence, SGC‐7901 cell proliferation inhibited is related to its regulation of the TGF‐β signalling pathway.
Tumour‐related signalling pathways are not only independent but also indivisible. They interweave, interact and influence each other to jointly affect the tumours and participate in the regulation of tumour cell proliferation, differentiation, invasion, metastasis, angiogenesis, immune response as well as other mechanisms. JNK is not only activated by TNF‐α, but also closely related to the caspase family, NF‐κB pathway and TGF‐β pathway. JNK mediates the exogenous death receptor pathway initiated by caspase‐8, and its active substance promotes the release of mitochondrial pro‐apoptotic protein jBID, which promotes the release of caspase‐8 inhibitors on the TNFR1 complex. Thus, the inhibition of caspase‐8 was decreased, and apoptosis was induced. 79 JNK is also activated by TGF‐β in many cancer cell lines, and the TGF‐β/Smad is upstream of JNK. MEKK1, a member of the MAPKKK family, directly binds to and activates the NF‐κB inhibitory kinase signals IKKα and IKKβ, leading to phosphorylation. MEKK1 is also an upstream kinase of IκB, selectively phosphorylating the Ser32 and Ser36 sites of IκB, which together activate the downstream NF‐κB to accelerate proliferation and reduce apoptosis. 80 , 81 That is, TNF‐α pathway, androgen receptor pathway and TGF‐β pathway are independent but related to each other and the three pathways constitute a TNF‐α/AR/TGF‐β signalling pathway axis.
5. CONCLUSIONS
In conclusion, in this study, the signal pathway of oridonin inhibiting gastric cancer proliferation was predicted by network pharmacology. Experimental studies confirmed that the proliferation inhibition of oridonin on SGC‐7901 cell was related to TNF‐α/AR/TGF‐β signalling pathway axis, which confirmed the prediction of network pharmacology, and elucidated the mechanism of oridonin on gastric cancer (Figure 8).
FIGURE 8.
Potential signalling pathway of oridonin on gastric cancer constructed in the present research.
AUTHOR CONTRIBUTIONS
Shiyong Gao: Conceptualization (lead); funding acquisition (equal); investigation (equal); software (equal); writing – original draft (equal). Huixin Tan: Data curation (equal); project administration (equal); writing – original draft (equal); writing – review and editing (equal). Dan Li: Investigation (equal); methodology (equal); writing – review and editing (equal).
FUNDING INFORMATION
Natural Science Foundation in Heilongjiang Province, Grant/Award Number: LH2021H002; 2021 Harbin University of Commerce Teacher “Innovation” Project, Grant/Award Number: LH2021H002; Outstanding Young Talents Project of the Central Government's Fund for Local Universities' Reform and Development, Grant/Award Number: 2020YQ12; Science and Technology Project of Heilongjiang Provincial Health and Family Planning Commission, Grant/Award Number: 2017–132; General Project of Education Department of Heilongjiang Province, Grant/Award Number: 12541571.
CONFLICT OF INTEREST STATEMENT
The authors declare that they have no conflict of interest.
Supporting information
Table S1.
Table S2.
Table S3.
Table S4.
Table S5.
Table S6.
ACKNOWLEDGEMENTS
No applicable.
Gao S, Tan H, Li D. Oridonin suppresses gastric cancer SGC‐7901 cell proliferation by targeting the TNF‐alpha/androgen receptor/TGF‐beta signalling pathway axis. J Cell Mol Med. 2023;27:2661‐2674. doi: 10.1111/jcmm.17841
Shiyong Gao and Huixin Tan contributed equally.
Shiyong Gao and Huixin Tan should be considered joint first author.
Contributor Information
Shiyong Gao, Email: sygao2002@163.com.
Huixin Tan, Email: thxydsy@163.com.
DATA AVAILABILITY STATEMENT
The data that supports the findings of this study are available in the supplementary material of this article
REFERENCES
- 1. Commission NP . Pharmacopoeia of People's Republic of China (2020 Edition). China Medical Science and Technology Press; 2020. [Google Scholar]
- 2. Bu H, Liu D, Zhang G, Chen L, Song Z. AMPK/mTOR/ULK1 Axis‐mediated pathway participates in apoptosis and autophagy induction by Oridonin in colon cancer DLD‐1 cells. Onco Targets Ther. 2020;13:8533‐8545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Zhou J, Li Y, Shi X, et al. Oridonin inhibits tumor angiogenesis and induces vessel normalization in experimental colon cancer. J Cancer. 2021;12(11):3257‐3264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Ding CY, Zhang YS, Chen HJ, et al. Oridonin ring A‐based diverse constructions of enone functionality: identification of novel dienone analogues effective for highly aggressive breast cancer by inducing apoptosis. J Med Chem. 2013;56(21):8814‐8825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Dai W, Zhang D, Duan C, et al. Preparation and characteristics of oridonin‐loaded nanostructured lipid carriers as a controlled‐release delivery system. J Microencapsul. 2010;27(3):234‐241. [DOI] [PubMed] [Google Scholar]
- 6. Gao C, Zhang L, Tang Z, Fang Z, Ye X, Yu W. Preparation, characterization, and anti‐colon cancer activity of oridonin‐loaded long‐circulating liposomes. Pharm Dev Technol. 2021;26(10):1073‐1078. [DOI] [PubMed] [Google Scholar]
- 7. He J, Cao Y, Yuan Q. Preparation and Photocytotoxicity in vitro of Oridonin‐porphyrin‐chitosan microspheres. Chin J Org Chem. 2017;37(3):759‐766. [Google Scholar]
- 8. Li C, Zhang D, Guo H, et al. Preparation and characterization of galactosylated bovine serum albumin nanoparticles for liver‐targeted delivery of oridonin. Int J Pharm. 2013;448(1):79‐86. [DOI] [PubMed] [Google Scholar]
- 9. Zhang DR, Tan TW, Gao L. Preparation of oridonin‐loaded solid lipid nanoparticles and studies on them in vitro and in vivo. Nanotechnology. 2006;17(23):5821‐5828. [Google Scholar]
- 10. Wen D, Yang YS, Gao DZ, Wang Z, Jiang QW, Zhao XF. Oridonin enhances the anti‐metastasis effect of oxaliplatinliplatin on colorectal cancer liver metastasis. Bull Exp Biol Med. 2021;172(1):26‐32. [DOI] [PubMed] [Google Scholar]
- 11. Zhang D, Zhou Q, Huang DD, et al. ROS/JNK/c‐Jun axis is involved in oridonin‐induced caspase‐dependent apoptosis in human colorectal cancer cells. Biochem Biophys Res Commun. 2019;513(3):594‐601. [DOI] [PubMed] [Google Scholar]
- 12. Xia SX, Zhang XL, Li CH, Guan HL. Oridonin inhibits breast cancer growth and metastasis through blocking the notch signaling. Saudi Pharm J. 2017;25(4):638‐643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Zhao Y, Xiao WW, Peng WQ, et al. Oridonin‐loaded nanoparticles inhibit breast cancer progression through regulation of ROS‐related Nrf2 signaling pathway. Front Bioeng Biotechnol. 2021;9:600579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Lou SM, Xu J, Wang BL, et al. Downregulation of lncRNA AFAP1‐AS1 by oridonin inhibits the epithelial‐to‐mesenchymal transition and proliferation of pancreatic cancer cells. Acta Biochim Biophys Sin. 2019;51(8):814‐825. [DOI] [PubMed] [Google Scholar]
- 15. Zhao X, Zhang Q, Wang YY, et al. Oridonin induces autophagy‐mediated cell death in pancreatic cancer by activating the c‐Jun N‐terminal kinase pathway and inhibiting phosphoinositide 3‐kinase signaling. Ann Transl Med. 2021;9(13):1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Fan XL, Wang T, Ji ZY, Li QP, Shen HY, Wang J. Synergistic combination therapy of lung cancer using lipid‐layered cisplatin and oridonin co‐encapsulated nanoparticles. Biomed Pharmacother. 2021;141:11180. [DOI] [PubMed] [Google Scholar]
- 17. Li S, Shi D, Zhang L, Yang F, Cheng G. Oridonin enhances the radiosensitivity of lung cancer cells by upregulating Bax and downregulating Bcl‐2. Exp Ther Med. 2018;16(6):4859‐4864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Xu L, Bi Y, Xu Y, et al. Oridonin inhibits the migration and epithelial‐to‐mesenchymal transition of small cell lung cancer cells by suppressing FAK‐ERK1/2 signalling pathway. J Cell Mol Med. 2020;24(8):4480‐4493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Chen K, Ye J, Qi L, et al. Oridonin inhibits hypoxia‐induced epithelial‐mesenchymal transition and cell migration by the hypoxia‐inducible factor‐1alpha/matrix metallopeptidase‐9 signal pathway in gallbladder cancer. Anticancer Drugs. 2019;30(9):925‐932. [DOI] [PubMed] [Google Scholar]
- 20. Lu J, Chen X, Qu S, et al. Oridonin induces G2/M cell cycle arrest and apoptosis via the PI3K/Akt signaling pathway in hormone‐independent prostate cancer cells. Oncol Lett. 2017;13(4):2838‐2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Wang Y, Zhu Z. Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway. Arch Med Sci. 2019;15(4):1017‐1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Jiang JH, Pi J, Jin H, Cai JY. Oridonin‐induced mitochondria‐dependent apoptosis in esophageal cancer cells by inhibiting PI3K/AKT/mTOR and Ras/Raf pathways. J Cell Biochem. 2019;120(3):3736‐3746. [DOI] [PubMed] [Google Scholar]
- 23. Yang IH, Shin JA, Lee KE, Kim J, Cho NP, Cho SD. Oridonin induces apoptosis in human oral cancer cells via phosphorylation of histone H2AX. Eur J Oral Sci. 2017;125(6):438‐443. [DOI] [PubMed] [Google Scholar]
- 24. Yang J, Ren X, Zhang L, Li Y, Cheng B, Xia J. Oridonin inhibits oral cancer growth and PI3K/Akt signaling pathway. Biomed Pharmacother. 2018;100:226‐232. [DOI] [PubMed] [Google Scholar]
- 25. Che X, Zhan J, Zhao F, et al. Oridonin promotes apoptosis and restrains the viability and migration of bladder cancer by impeding TRPM7 expression via the ERK and AKT signaling pathways. Biomed Res Int. 2021;2021:4340950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Zhang HP, Li GQ, Guo WZ, et al. Oridonin synergistically enhances JQ1‐triggered apoptosis in hepatocellular cancer cells through mitochondrial pathway. Oncotarget. 2017;8(63):106833‐106843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. He Z, Xiao X, Li S, et al. Oridonin induces apoptosis and reverses drug resistance in cisplatin resistant human gastric cancer cells. Oncol Lett. 2017;14(2):2499‐2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Wang Y, Lv H, Dai C, Wang X, Yin Y, Chen Z. Oridonin dose‐dependently modulates the cell senescence and apoptosis of gastric cancer cells. Evid Based Complement Alternat Med. 2021;2021:5023536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Yang Q, Ma W, Yu K, et al. Oridonin suppresses human gastric cancer growth in vitro and in vivo via inhibition of VEGF, integrin beta3, and PCNA. Biol Pharm Bull. 2020;43(7):1035‐1045. [DOI] [PubMed] [Google Scholar]
- 30. Gao SY, Li J, Qu XY, Zhu N, Ji YB. Downregulation of Cdk1 and cyclinB1 expression contributes to oridonin‐induced cell cycle arrest at G2/M phase and growth inhibition in SGC‐7901 gastric cancer cells. Asian Pac J Cancer Prev. 2014;15(15):6437‐6441. [DOI] [PubMed] [Google Scholar]
- 31. Gao S, Tan H, Zhu N, et al. Oridonin induces apoptosis through the mitochondrial pathway in human gastric cancer SGC‐7901 cells. Int J Oncol. 2016;48(6):2453‐2460. [DOI] [PubMed] [Google Scholar]
- 32. Hopkins AL. Network pharmacology. Nat Biotechnol. 2007;25(10):1110‐1111. [DOI] [PubMed] [Google Scholar]
- 33. Hopkins AL. Network pharmacology: the next paradigm in drug discovery. Nat Chem Biol. 2008;4(11):682‐690. [DOI] [PubMed] [Google Scholar]
- 34. Zhang R, Zhu X, Bai H, Ning K. Network pharmacology databases for traditional Chinese medicine: review and assessment. Front Pharmacol. 2019;10:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Tan H, Gao S, Zhuang Y, et al. R‐Phycoerythrin induces SGC‐7901 apoptosis by arresting cell cycle at S phase. Mar Drugs. 2016;14(9):166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Wong AM, Zhang Y, Kesler K, et al. Genomic and in vivo evidence of synergy of a herbal extract compared to its most active ingredient: Rabdosia rubescens vs. oridonin. Exp Ther Med. 2010;1(6):1013‐1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Zhu HQ, Zhang C, Guo ZY, et al. Oridonin induces Mdm2‐p60 to promote p53‐mediated apoptosis and cell cycle arrest in neuroblastoma. Cancer Med. 2019;8(11):5313‐5326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Park H, Jeong YJ, Han NK, Kim JS, Lee HJ. Oridonin enhances radiation‐induced cell death by promoting DNA damage in non‐small cell lung cancer cells. Int J Mol Sci. 2018;19(8):2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Xu ZZ, Fu WB, Jin Z, Guo P, Wang WF, Li JM. Reactive oxygen species mediate oridonin‐induced apoptosis through DNA damage response and activation of JNK pathway in diffuse large B cell lymphoma. Leuk Lymphoma. 2016;57(4):888‐898. [DOI] [PubMed] [Google Scholar]
- 40. Pagliara V, Donadio G, De Tommasi N, et al. Bioactive ent‐kaurane diterpenes oridonin and irudonin prevent cancer cells migration by interacting with the Actin cytoskeleton controller Ezrin. Int J Mol Sci. 2020;21(19):7186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Kibble M, Saarinen N, Tang J, Wennerberg K, Makela S, Aittokallio T. Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products. Nat Prod Rep. 2015;32(8):1249‐1266. [DOI] [PubMed] [Google Scholar]
- 42. Mahdavi Sharif P, Jabbari P, Razi S, Keshavarz‐Fathi M, Rezaei N. Importance of TNF‐alpha and its alterations in the development of cancers. Cytokine. 2020;130:155066. [DOI] [PubMed] [Google Scholar]
- 43. Muthusamy V, Piva TJ. The UV response of the skin: a review of the MAPK, NFkappaB and TNFalpha signal transduction pathways. Arch Dermatol Res. 2010;302(1):5‐17. [DOI] [PubMed] [Google Scholar]
- 44. Huang G, Shi LZ, Chi H. Regulation of JNK and p38 MAPK in the immune system: signal integration, propagation and termination. Cytokine. 2009;48(3):161‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Korcheva V, Wong J, Corless C, Iordanov M, Magun B. Administration of ricin induces a severe inflammatory response via nonredundant stimulation of ERK, JNK, and P38 MAPK and provides a mouse model of hemolytic uremic syndrome. Am J Pathol. 2005;166(1):323‐339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Sui X, Kong N, Ye L, et al. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 2014;344(2):174‐179. [DOI] [PubMed] [Google Scholar]
- 47. Muniyappa H, Das KC. Activation of c‐Jun N‐terminal kinase (JNK) by widely used specific p38 MAPK inhibitors SB202190 and SB203580: a MLK‐3‐MKK7‐dependent mechanism. Cell Signal. 2008;20(4):675‐683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Ferrer I, Friguls B, Dalfo E, Planas AM. Early modifications in the expression of mitogen‐activated protein kinase (MAPK/ERK), stress‐activated kinases SAPK/JNK and p38, and their phosphorylated substrates following focal cerebral ischemia. Acta Neuropathol. 2003;105(5):425‐437. [DOI] [PubMed] [Google Scholar]
- 49. Hong X, Mao L, Xu L, Hu Q, Jia R. Prostate‐specific membrane antigen modulates the progression of prostate cancer by regulating the synthesis of arginine and proline and the expression of androgen receptors and Fos proto‐oncogenes. Bioengineered. 2022;13(1):995‐1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Anestis A, Zoi I, Papavassiliou AG, Karamouzis MV. Androgen receptor in breast cancer‐clinical and preclinical research insights. Molecules. 2020;25(2):358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Hanamura T, Christenson JL, O'Neill KI, et al. Secreted indicators of androgen receptor activity in breast cancer pre‐clinical models. Breast Cancer Res. 2021;23(1):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Kono M, Fujii T, Lim B, Karuturi MS, Tripathy D, Ueno NT. Androgen receptor function and androgen receptor‐targeted therapies in breast cancer: a review. JAMA Oncol. 2017;3(9):1266‐1273. [DOI] [PubMed] [Google Scholar]
- 53. Chung WM, Chen L, Chang WC, Su SY, Hung YC, Ma WL. Androgen/androgen receptor signaling in ovarian cancer: molecular regulation and therapeutic potentials. Int J Mol Sci. 2021;22(14):7748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Peng L, Li Y, Wei S, et al. LAMA4 activated by androgen receptor induces the cisplatin resistance in gastric cancer. Biomed Pharmacother. 2020;124:109667. [DOI] [PubMed] [Google Scholar]
- 55. Xia N, Cui J, Zhu M, Xing R, Lu Y. Androgen receptor variant 12 promotes migration and invasion by regulating MYLK in gastric cancer. J Pathol. 2019;248(3):304‐315. [DOI] [PubMed] [Google Scholar]
- 56. Grant L, Banerji S, Murphy L, et al. Androgen receptor and Ki67 expression and survival outcomes in non‐small cell lung cancer. Horm Cancer. 2018;9(4):288‐294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Zhou J, Wang H, Sun Q, et al. miR‐224‐5p‐enriched exosomes promote tumorigenesis by directly targeting androgen receptor in non‐small cell lung cancer. Mol Ther Nucleic Acids. 2021;23:1217‐1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Sikic D, Taubert H, Wirtz RM, et al. High androgen receptor mRNA expression is associated with improved outcome in patients with high‐risk non‐muscle‐invasive bladder cancer. Life (Basel). 2021;11(7):642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Teramoto Y, Jiang G, Goto T, et al. Androgen receptor signaling induces cisplatin resistance via down‐regulating GULP1 expression in bladder cancer. Int J Mol Sci. 2021;22(18):10030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Ho AL. Androgen receptor pathway in salivary gland cancer. J Clin Oncol. 2021;39(36):4069‐4072. [DOI] [PubMed] [Google Scholar]
- 61. Locati LD, Cavalieri S, Bergamini C, et al. Abiraterone acetate in patients with castration‐resistant, androgen receptor‐expressing salivary gland cancer: a phase II trial. J Clin Oncol. 2021;39(36):4061‐4068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Feng MW, Hanley KL, Feng GS. Androgen receptor, neovascularization and liver cancer metastasis. J Hepatol. 2021;75(4):768‐769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Bialek J, Piwonka M, Kawan F, Fornara P, Theil G. Differential expression of the androgen receptor, splice variants and relaxin 2 in renal cancer. Life (Basel). 2021;11(8):731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Whiteland H, Spencer‐Harty S, Morgan C, et al. A role for STEAP2 in prostate cancer progression. Clin Exp Metastasis. 2014;31(8):909‐920. [DOI] [PubMed] [Google Scholar]
- 65. Yang D, Holt GE, Velders MP, Kwon ED, Kast WM. Murine six‐transmembrane epithelial antigen of the prostate, prostate stem cell antigen, and prostate‐specific membrane antigen: prostate‐specific cell‐surface antigens highly expressed in prostate cancer of transgenic adenocarcinoma mouse prostate mice. Cancer Res. 2001;61(15):5857‐5860. [PubMed] [Google Scholar]
- 66. Guo Y, Zhu J, Xu X, et al. TGF‐beta/YB‐1/Atg7 axis promotes the proliferation of hepatic progenitor cells and liver fibrogenesis. Biochim Biophys Acta Mol Basis Dis. 2022;1868(1):166290. [DOI] [PubMed] [Google Scholar]
- 67. Yeung TL, Leung CS, Wong KK, et al. TGF‐beta modulates ovarian cancer invasion by upregulating CAF‐derived versican in the tumor microenvironment. Cancer Res. 2013;73(16):5016‐5028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Yuan JH, Yang F, Wang F, et al. A long noncoding RNA activated by TGF‐beta promotes the invasion‐metastasis cascade in hepatocellular carcinoma. Cancer Cell. 2014;25(5):666‐681. [DOI] [PubMed] [Google Scholar]
- 69. Pang MF, Georgoudaki AM, Lambut L, et al. TGF‐beta1‐induced EMT promotes targeted migration of breast cancer cells through the lymphatic system by the activation of CCR7/CCL21‐mediated chemotaxis. Oncogene. 2016;35(6):748‐760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Boguslawska J, Rodzik K, Poplawski P, et al. TGF‐beta1 targets a microRNA network that regulates cellular adhesion and migration in renal cancer. Cancer Lett. 2018;412:155‐169. [DOI] [PubMed] [Google Scholar]
- 71. Wang Y, Yang H, Su X, et al. TGF‐beta1/SMOC2/AKT and ERK axis regulates proliferation, migration, and fibroblast to myofibroblast transformation in lung fibroblast, contributing with the asthma progression. Hereditas. 2021;158(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Nepon‐Sixt BS, Alexandrow MG. TGF beta 1 cell cycle arrest is mediated by inhibition of MCM assembly in Rb‐deficient conditions. Mol Cancer Res. 2019;17(1):277‐288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Takahashi K, Inoue KA, Kaida A, et al. TGF‐beta‐induced cell cycle arrest is associated with increased migration and metastasis of oral squamous carcinoma cells. Cancer Sci. 2021;112:418. [Google Scholar]
- 74. Gudino V, Cammareri P, Billard CV, Myant KB. Negative regulation of TGFbeta‐induced apoptosis by RAC1B enhances intestinal tumourigenesis. Cell Death Dis. 2021;12(10):873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Li Z, Liu H, Zhang Y, Tan H. The effect of propofol on the proliferation and apoptosis of hepatocellular carcinoma cells through TGF‐Beta1/Smad2 signaling pathway. Bioengineered. 2021;12(1):4581‐4592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Akhurst RJ. TGF‐beta antagonists: why suppress a tumor suppressor? J Clin Invest. 2002;109(12):1533‐1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Javelaud D, Alexaki VI, Mauviel A. Transforming growth factor‐beta in cutaneous melanoma. Pigment Cell Melanoma Res. 2008;21(2):123‐132. [DOI] [PubMed] [Google Scholar]
- 78. Gough NR, Xiang X, Mishra L. TGF‐beta signaling in liver, pancreas, and gastrointestinal diseases and cancer. Gastroenterology. 2021;161(2):434‐452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Park SM, Kim S, Choi JS, et al. TGF‐beta inhibits Fas‐mediated apoptosis of a follicular dendritic cell line by down‐regulating the expression of Fas and caspase‐8: counteracting role of TGF‐beta on TNF sensitization of Fas‐mediated apoptosis. J Immunol. 2005;174(10):6169‐6175. [DOI] [PubMed] [Google Scholar]
- 80. Baumann B, Weber CK, Troppmair J, et al. Raf induces NF‐kappaB by membrane shuttle kinase MEKK1, a signaling pathway critical for transformation. Proc Natl Acad Sci U S A. 2000;97(9):4615‐4620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lee CK, Lee EY, Kim YG, Mun SH, Moon HB, Yoo B. Alpha‐lipoic acid inhibits TNF‐alpha induced NF‐kappa B activation through blocking of MEKK1‐MKK4‐IKK signaling cascades. Int Immunopharmacol. 2008;8(2):362‐370. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Table S1.
Table S2.
Table S3.
Table S4.
Table S5.
Table S6.
Data Availability Statement
The data that supports the findings of this study are available in the supplementary material of this article