Journal of Traditional Chinese Medicine ›› 2024, Vol. 44 ›› Issue (5): 896-905.DOI: 10.19852/j.cnki.jtcm.20240806.001
Previous Articles Next Articles
ZHANG Guangshun1,2, XU Xiaonan3,4,5, XU Chuyun6, LIAO Guanghui1,2, XU Hao3,4,5, LOU Zhaohuan1,2(), ZHANG Guangji3,4,5()
Received:
2023-04-21
Accepted:
2023-08-29
Online:
2024-10-15
Published:
2024-08-06
Contact:
Prof. ZHANG Guangji, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. zgj@zcmu.edu.cn Telephone: +86-13335812880; +86-13957152511; Prof. LOU Zhaohuan, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China. zhaohuanlou@zcmu.edu.cn
Supported by:
ZHANG Guangshun, XU Xiaonan, XU Chuyun, LIAO Guanghui, XU Hao, LOU Zhaohuan, ZHANG Guangji. Actinidia chinensis polysaccharide interferes with the epithelial-mesenchymal transition of gastric cancer by regulating the nuclear transcription factor-κB pathway to inhibit invasion and metastasis[J]. Journal of Traditional Chinese Medicine, 2024, 44(5): 896-905.
Figure 1 ACPS improved tumor inflammatory response A: histopathology of gastric cancer transplanted tumor; A1: representative picture of the model group; A2: 5-FU group; A3: ACPS-L group; A4: ACPS-H group; HE staining, × 200; B: serum levels of IL-6 in each group; C: serum levels of TGF-β in each group. 5-FU: 5-fluorouracil; ACPS: actinidia chinensis polysaccharide; ACPS-L: actinidia chinensis polysaccharide low-dose group (50 mg·kg-1·d-1); ACPS-H: actinidia chinensis polysaccharide high-dose group (100 mg·kg-1·d-1). The drug was given intraperitoneally once a day for 28 d. IL-6: interleukin-6; TGF-β: transforming growth factor beta. The statistical method used in this figure is one-way analysis of variance. Results are presented as mean ± standard deviation (n = 6). aP<0.01 model group vs normal group; bP<0.01 vs model group; cP<0.05 model group vs normal group.
Figure 2 Scratch experiment was used to evaluate the effect of ACPS on the migration of BGC-823 cells A: migration at time points of 0, 6, 24, 48 h in scratch experiment; A1: control group at 0 h post-administration; A2: control group at 6 h post-administration; A3: control group at 24 h post-administration; A4: control group at 48 h post-administration; A5: EGF group at 0 h post-administration (100 ng/mL); A6: EGF group at 6 h post-administration; A7: EGF group at 24 h post-administration; A8: EGF group at 48 h post-administration; A9: ACPS group at 0 h post-administration (0.1 mg/mL); A10: ACPS group at 6 h post-administration; A11: ACPS group at 24 h post-administration; A12: ACPS group at 48 h post-administration; A13: EGF (100 ng/mL) + ACPS (0.1 mg/mL) group at 0 h post-administration; A14: EGF + ACPS group at 6 h post-administration; A15: EGF + ACPS group at 24 h post-administration; A16: EGF + ACPS group at 48 h post-administration; B: mobility of each group was compared at 48 h post-administration. EGF: epidermal growth factor; ACPS: Actinidia chinensis polysaccharide. The scratch experiment photos were collected under a 10 μm mirror. The statistical method used in this figure is one-way analysis of variance. Results are presented as mean ± standard deviation (n = 3). aP<0.01 vs control group.
Figure 3 Immunohistochemistry was used to detect the expression of MMP9, N-cadherin and p-NF-κB p65 A: representative picture of MMP9 protein expression in each group by immunohistochemistry (DAB staining, × 200); A1: model group; A2: 5-FU group; A3: ACPS-L dose group; A4: ACPS-H dose group; B: representative picture of N-cadherin protein expression in each group by immunohistochemistry (DAB staining, × 200); B1: model group; B2: 5-FU group; B3: ACPS-L dose group; B4: ACPS-H dose group; C: representative picture of p-NF-κB p65 protein expression in each group by immunohistochemistry (DAB staining, × 200); C1: model group; C2: 5-FU group; C3: ACPS-L dose group; A4: ACPS-H dose group; D: mean OD values of MMP9 in each group were compared by immunohistochemistry; E: mean OD values of N-cadherin in each group were compared by immunohistochemistry; F: mean OD values of p-NF-κB p65 in each group were compared by immunohistochemistry. 5-FU: 5-fluorouracil; ACPS: actinidia chinensis polysaccharide; ACPS-L: actinidia chinensis polysaccharide low-dose group (50 mg·kg-1·d-1); ACPS-H: actinidia chinensis polysaccharide high-dose group (100 mg·kg-1·d-1). The drug was given intraperitoneally once a day for 28 d. MMP-9: matrix metallopeptidase 9; N-cadherin: neural cadherin; P-NFκB p65: phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells p65 subunit. The statistical method used in this figure is one-way analysis of variance. Results are presented as mean ± standard deviation (n = 6). aP<0.01 vs model group.
Figure 4 ACPS promoted p65-NF-κB nuclear transfer in BGC-823 cells and reduced the mRNA expression of MMP2, ICAM2, and VCAM1 A: expression of p65-NFκB was detected using confocal technology. A1: control group; A2: EGF group (100 ng/mL); A3: ACPS group (0.1 mg/mL); A4: EGF + ACPS group; Blue fluorescence represents nuclei stained with DAPI; The red fluorescence represents p65-NFκB; B: qPCR was used to detect MMP2 genes in each group standardized with GAPDH; C: qPCR was used to detect ICAM2 genes in each group standardized with GAPDH; D: qPCR was used to detect VCAM1 genes in each group standardized with GAPDH. Drug intervention for 48 h. p65-NFκB: p65 subunit of nuclear factor kappa-light-chain-enhancer of activated B cells; DAPI: 6-diamidino-2-phenylindole; qPCR: quantitative Polymerase chain reaction; MMP2: matrix metalloproteinase-2; ICAM2: intercellular adhesion molecule 2; VCAM1: vascular cell adhesion molecule 1; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; EGF: epidermal growth factor; ACPS: actinidia chinensis polysaccharide. The scale bar of the above image is 25 μm. The statistical method used in this figure is one-way analysis of variance. Results are presented as mean ± standard deviation (n = 3). aP<0.01, EGF group vs Control group; bP<0.01, vs EGF group.
1. | Zang X, Jiang J, Gu J, et al. Circular RNA EIF4G 3 suppresses gastric cancer progression through inhibition of β-catenin by promoting δ-catenin ubiquitin degradation and upregulating SIK1. Mol Cancer 2022; 21: 141. |
2. |
Zhang Y, Liu C, Zhou L. Prognosis of gastric adenocarcinoma associated with girdin, Akt, and cortactin. Ann Saudi Med 2022; 42: 181-90.
DOI PMID |
3. | Zhou Y, Li X, Luo W, et al. Allicin in digestive system cancer: from biological effects to clinical treatment. Front Pharmacol 2022; 13: 903259. |
4. | Gao R, Lyu Y. Characterizing the Antitumor Effect of coptis chinensis and mume fructus against colorectal cancer based on pharmacological analysis. Evid Based Complement Alternat Med 2022; 2022: 9061752. |
5. | Cangiano LR, Henry DD, Ciriaco FM, et al. Triterpenes from Olea europaea modulate in vitro ruminal fermentation. Transl Anim Sci 2022; 6: txac056. |
6. | She Y, Zhao X, Wu P, et al. COL8A 1 Predicts the clinical prognosis of gastric cancer and is related to epithelial-mesenchymal transition. Biomed Res Int 2022; 2022: 7567447. |
7. |
Fu C, Wang J, Pallikkuth S, et al. EWI2 prevents EGFR from clustering and endocytosis to reduce tumor cell movement and proliferation. Cell Mol Life Sci 2022; 79: 389.
DOI PMID |
8. | Song J, Wei R, Huo S, et al. Metastasis related epithelial-mesenchymal transition signature predicts prognosis and response to immunotherapy in gastric cancer. Front Immunol 2022; 13: 920512. |
9. |
Sun Y, Du R, Shang Y, et al. Rho GTPase-activating protein 35 suppresses gastric cancer metastasis by regulating cytoskeleton reorganization and epithelial-to-mesenchymal transition. Bioengineered 2022; 13: 14605-15.
DOI PMID |
10. | Westrick RJ, Røjkjaer LP, Yang AY, et al. Deficiency of plasminogen activator inhibitor-2 results in accelerated tumor growth. J Thromb Haemost 2020; 18: 2968-75. |
11. | Wang Y, Liu F, Chen L, et al. Neutrophil extracellular traps (NETs) promote non-small cell lung cancer metastasis by suppressing lncRNA mir503HG to activate the NF-κB/NLRP 3 inflammasome pathway. Front Immunol 2022; 13: 867516. |
12. |
Mirzaei S, Saghari S, Bassiri F, et al. NF-κB as a regulator of cancer metastasis and therapy response: a focus on epithelial-mesenchymal transition. J Cell Physiol 2022; 237: 2770-95.
DOI PMID |
13. |
Huber MA, Azoitei N, Baumann B, et al. NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J Clin Invest 2004; 114: 569-81.
DOI PMID |
14. |
Gao Z, Deng G, Li Y, et al. Actinidia chinensis Planch prevents proliferation and migration of gastric cancer associated with apoptosis, ferroptosis activation and mesenchymal phenotype suppression. Biomed Pharmacother 2020; 126: 110092.
DOI PMID |
15. |
Zeng Q, Nie X, Li L, et al. Salidroside promotes sensitization to doxorubicin in human cancer cells by affecting the PI3K/Akt/HIF signal pathway and inhibiting the expression of tumor-resistance-related proteins. J Nat Prod 2022; 85: 196-204.
DOI PMID |
16. | Lu X, Qian CN, Mu YG, et al. Serum CCL2 and serum TNF-α--two new biomarkers predict bone invasion, post-treatment distant metastasis and poor overall survival in nasopharyngeal carcinoma. Eur J Cancer 2011; 47: 339-46. |
17. | Xie F, Zhou X, Li H, et al. USP8 promotes cancer progression and extracellular vesicle-mediated CD8+ T cell exhaustion by deubi-quitinating the TGF-β receptor TβRII. EMBO J 2022; 41: e108791. |
18. | Jiang H, Ma P, Duan Z, et al. Ginsenoside Rh4 suppresses metastasis of gastric cancer via SIX1-dependent TGF-β/Smad2/3 signaling pathway. Nutrients 2022; 14: 1564. |
19. |
Cheng H, Chen L, Fang Z, et al. STIM2 promotes the invasion and metastasis of breast cancer cells through the NFAT1/TGF-β1 pathway. Cell Mol Biol (Noisy-le-grand) 2022; 67: 55-61.
DOI PMID |
20. |
Bailly M. Connecting cell adhesion to the actin polymerization machinery: vinculin as the missing link? Trends Cell Biol 2003; 13: 163-5.
PMID |
21. |
Chen H, Cohen DM, Choudhury DM, et al. Spatial distribution and functional significance of activated vinculin in living cells. J Cell Biol 2005; 169: 459-70.
DOI PMID |
22. | Chan LP, Tseng YP, Wang HC, et al. Growth regulated oncogene-α contribute to EMT/MMPs pathway by binding its receptors in head and neck squamous cell carcinoma. Life sciences 2022; 306: 120791. |
23. |
Alba J, Barcia R, Gutiérrez-Berzal J, et al. Could inhibition of metalloproteinases be used to block the process of metastasis? Cell Biochem Funct 2022; 40: 600-7.
DOI PMID |
24. |
Nishimura S, Yamamoto Y, Sugimoto A, et al. Lipocalin-2 negatively regulates epithelial-mesenchymal transition through matrix metalloprotease-2 downregulation in gastric cancer. Gastric cancer 2022; 25: 850-61.
DOI PMID |
25. | Yu C, Chen W, Cai Y, et al. The lncRNA ZNF667-AS 1 inhibits propagation, invasion, and angiogenesis of gastric cancer by silencing the expression of N-Cadherin and VEGFA. J Oncol 2022; 2022: 3579547. |
26. | Loh CY, Chai JY, Tang TF, et al. The E-cadherin and N-cadherin switch in epithelial-to-mesenchymal transition: signaling, therapeutic Implications, and Challenges. Cells 2019; 8: 1118. |
27. |
Chauhan A, Islam AU, Prakash H, et al. Phytochemicals targeting NF-κB signaling: potential anti-cancer interventions. J Pharm Anal 2022; 12: 394-405.
DOI PMID |
28. | Wang W, Zhou L, Sun Z, et al. Down-regulation of tripartite motif containing 59 (TRIM59) blocks the NF-κB signaling pathway and inhibits the invasion and migration of nasopharyngeal carcinoma cells. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 2022; 38: 419-24. |
29. |
Sha M, Ye J, Zhang LX, et al. Celastrol induces apoptosis of gastric cancer cells by miR-21 inhibiting PI3K/Akt-NF-κB signaling pathway. Pharmacology 2014; 93: 39-46.
DOI PMID |
30. | Liao X, Che X, Zhao W, et al. The β-adrenoceptor antagonist, propranolol, induces human gastric cancer cell apoptosis and cell cycle arrest via inhibiting nuclear factor κB signaling. Oncol Rep 2010; 24: 1669-76. |
31. | Sohma I, Fujiwara Y, Sugita Y, et al. Parthenolide, an NF-κB inhibitor, suppresses tumor growth and enhances response to chemotherapy in gastric cancer. Cancer Genomics Proteomics 2011; 8: 39-47. |
32. | Zhang S, Xie B, Wang L, et al. Macrophage-mediated vascular permeability via VLA4/VCAM1 pathway dictates ascites development in ovarian cancer. J Clin Invest 2021; 131; e140315. |
33. | Bronikowska J, Kłósek M, Janeczko T, et al. The modulating effect of methoxy-derivatives of 2'-hydroxychalcones on the release of IL-8, MIF, VCAM-1 and ICAM-1 by colon cancer cells. Biomed Pharmacother 2022; 145: 112428. |
34. |
Melero I, Gabari I, Corbí AL, et al. An anti-ICAM-2 (CD102) monoclonal antibody induces immune-mediated regressions of transplanted ICAM-2-negative colon carcinomas. Cancer Res 2002; 62: 3167-74.
PMID |
Viewed | ||||||
Full text |
|
|||||
Abstract |
|
|||||
Sponsored by China Association of Chinese Medicine
& China Academy of Chinese Medical Sciences
16 Nanxiaojie, Dongzhimen Nei, Beijing, China. 100700 Email: jtcmen@126.com
Copyright 2020 Journal of Traditional Chinese Medicine. All rights reserved.