-
Ononin inhibits triple-negative breast cancer lung metastasis by targeting the EGFR-mediated PI3K/Akt/mTOR pathway.
The spreading of cancer cells from the primary tumor site to other parts of the body, known as metastasis, is the leading cause of cancer recurrence and mortality in patients with triple-negative breast cancer (TNBC). Overexpression of epidermal growth factor receptor (EGFR) is observed in approximately 70% of TNBC patients. EGFR is crucial for promoting tumor metastasis and associated with poor prognosis. Therefore, it is vital to identify effective therapeutic strategies targeting EGFR inhibition. Ononin, an isoflavonoid found in various plants, such as clover and soybeans, has been shown to have anticancer properties in several cancers. In the present study, we aimed to investigate the effects of ononin on TNBC lung metastasis and the associated molecular pathways. We used various assays, including cell viability, colony formation, Transwell, wound healing, ELISA, Western blotting, and staining techniques, to achieve this objective. The results demonstrated that ononin effectively suppressed cellular proliferation and induced apoptosis, as evidenced by the cell viability assay, colony formation assay, and expression of apoptosis markers, and reduced the metastatic capabilities of TNBC cells. These effects were achieved through the direct suppression of cell adhesion, invasiveness and motility. Furthermore, in TNBC xenograft lung metastatic models, ononin treatment significantly reduced tumor growth and lung metastasis. Additionally, ononin reversed the epithelial-mesenchymal transition (EMT) by downregulating the expression of EMT markers and matrix metalloproteinases, as confirmed by Western blot analysis. Furthermore, ononin treatment reduced EGFR phosphorylation and suppressed the PI3K, Akt, and mTOR signaling pathways, which was further confirmed using EGFR agonists or inhibitors. Importantly, ononin treatment did not exert any toxic effects on liver or kidney function. In conclusion, our findings suggest that ononin is a safe and potentially therapeutic treatment for TNBC metastasis that targets the EGFR-mediated PI3K/Akt/mTOR pathway. Further studies are warranted to validate its efficacy and explore its potential clinical applications.
Ganesan K
,Xu C
,Wu J
,Du B
,Liu Q
,Sui Y
,Song C
,Zhang J
,Tang H
,Chen J
... -
《-》
-
Ginsenoside 20(S)-protopanaxadiol inhibits triple-negative breast cancer metastasis in vivo by targeting EGFR-mediated MAPK pathway.
Metastasis is the primary cause of cancer recurrence and cancer related mortality in triple-negative breast cancer (TNBC). EGFR overexpression is in 50-75% TNBC and EGFR-mediated signaling has potential as an attractive therapeutic target in some specific subtypes of breast cancer due to its significant association with tumor metastasis and poor prognosis. Therefore, identification of promising therapeutic strategies targeting EGFR with higher specificity toward cancer metastasis is urgently needed. 20(S)-protopanaxadiol (PPD), one of the major active metabolites from Panax ginseng, has been widely reported to possess pleiotropic anticancer activities in various cancers. In this study, we investigated the effect of PPD against cancer metastasis and the related molecular mechanisms in TNBC in vitro and in vivo. PPD (>30 μM) suppressed cell proliferation by arresting cell cycle in G0/1 phase and triggering cells apoptosis as shown by cell viability assay, flow cytometry analysis and colony formation assay, whereas lower dose of PPD (<20 μM) decreased metastatic potential of MDA-MB-231 and SUM159 cells through direct inhibition of cell adhesion, motility and invasiveness. In TNBC xenograft and syngeneic models, PPD treatment markedly decreased tumor growth and lung metastasis. PPD reversed epithelial-mesenchymal transition (EMT), decreased the expression and activity of matrix metalloproteinases (MMPs) while increased the expression of tissue inhibitors of metalloproteinases (TIMPs) as shown by Western blot and gelatin zymography. Cell signaling pathways that control the expression or activation of these processes were investigated by Western blot and ELISA assay. PPD treatment reduced the phosphorylation of EGFR and down-regulated the activation ERK1/2, p38 and JNK signaling, which was further validated by using the agonists or inhibitors of EGFR and MAP kinases family. Collectively, these findings suggest that PPD holds therapeutic potential against the tumor metastasis of TNBC via targeting EGFR-mediated MAPK pathway.
Peng B
,He R
,Xu Q
,Yang Y
,Hu Q
,Hou H
,Liu X
,Li J
... -
《-》
-
Rhizoma Amorphophalli inhibits TNBC cell proliferation, migration, invasion and metastasis through the PI3K/Akt/mTOR pathway.
Triple-negative breast cancer (TNBC) often presents with a high histological grade and high malignancy, which greatly contribute to patient morbidity and mortality. Rhizoma Amorphophalli exhibits many biological and pharmacological activities, but its potential as a therapeutic agent for the treatment of metastatic TNBC patients remains poorly understood.
The aim of this study was to determine whether Rhizoma Amorphophalli inhibits metastasis in the human TNBC MDA-MB-231 cell line.
CCK-8 and colony formation assays were adopted for the analysis of cell activity and cell proliferation, respectively. Flow cytometry was used for cell cycle analysis. Wound healing and transwell assays were performed to assess cell migration and invasion, respectively. PI3K/Akt/mTOR signaling pathways were analyzed through western blotting. Breast cancer cell metastasis to the lung in a xenograft model was evaluated by in vivo fluorescence imaging. A GC-MS analysis was performed to determine the main components of the petroleum ether fraction from the ethanol extract of Rhizoma Amorphophalli (abbreviated RhA).
RhA significantly reduced breast cancer cell viability and proliferation. The flow cytometry analysis indicated that RhA induced MDA-MB-231 cell arrest at the S phase. Additionally, RhA decreased MDA-MB-231 cell migration and invasion and inhibited the PI3K/Akt/mTOR signaling pathway. In addition, mice treated with RhA exhibited a significant reduction in tumor infiltration and a decrease in breast cancer cell metastasis to the lung. The GC-MS analysis results showed that RhA contained a large number of unsaturated fatty acids, such as octadecadienoic acid (linoleic acid), octadecatrienoic acid (linolenic acid), and oleate, which might represent the anticancer components of the extract.
The results of this study suggest that RhA has potential as a therapeutic candidate for metastatic TNBC treatment.
Wu C
,Qiu S
,Liu P
,Ge Y
,Gao X
... -
《-》
-
Sophoraflavanone G suppresses the progression of triple-negative breast cancer via the inactivation of EGFR-PI3K-AKT signaling.
Sophoraflavanone G (SG), a prenylated flavonoid extracted from Sophora flavescens, has been found to possess antitumor activity in several types of human cancer. However, the biological functions and molecular mechanism of SG in triple-negative breast cancer (TNBC) are required to be investigated. On the basis of network pharmacology methods and molecular docking technology, estimated glomerular filtration rate (EGFR) was identified as a potential target, and phosphatidylinositol 3-kinase-protein kinase B (PI3K-AKT) signaling was demonstrated as an important signaling pathway for SG to treat breast cancer. TNBC cells (BT-549 and MDA-MB-231) were used to determine the effects of SG in vitro. Cell Counting Kit, 5-ethynyl-2'-deoxyuridine, and colony formation assays confirmed the proliferation inhibition of SG on TNBC cells. Moreover, SG administration promoted cell apoptosis by affecting Bax, Bcl-2, and cleaved caspase-3 expression. SG treatment also enhanced oxidative stress of TNBC cells by inducing reactive oxygen species production, increasing malondialdehyde (MDA) level, and decreasing superoxide dismutase activity. Additionally, SG suppressed cell migration, invasion, and epithelial-mesenchymal transition process. Inactivated EGFR-PI3K-AKT signaling was observed in TNBC cells after treatment with SG. Furthermore, the inhibitory effects of SG on cell proliferation and metastasis and the promotive effects of SG on cell apoptosis and oxidative stress were significantly attenuated due to the overexpression of EGFR. Mice experiments revealed the suppression of SG on tumor growth and EGFR-PI3K-AKT signaling. Together, SG repressed the proliferation and metastasis, and induced apoptosis and oxidative stress in TNBC by targeting the EGFR-PI3K-AKT signaling pathway. SG might serve as a promising therapeutic agent to combat TNBC.
Cheng W
,Liu D
,Guo M
,Li H
,Wang Q
... -
《-》
-
Calycosin inhibits triple-negative breast cancer progression through down-regulation of the novel estrogen receptor-α splice variant ER-α30-mediated PI3K/AKT signaling pathway.
Triple-negative breast cancer (TNBC) is a heterogeneous carcinoma characterized by the most aggressive phenotype among all breast cancer subtypes. However, therapeutic options for TNBC patients have limited clinical efficacy due to lack of specific target and efficient targeted therapeutics.
To investigate the biological characteristics of a novel estrogen receptor (ER)-α splice variant ER-α30 in breast cancer cells, and its possible role in the anticancer effects of calycosin, a typical phytoestrogen derived from the herbal plant Astragalus membranaceus, against TNBC. This may also provide a better understanding of the inhibitory activity of calycosin on TNBC progression.
Breast cancer tissues and para-cancer tissues were collected and analyzed for the expression levels of ER-α30 using immunohistochemistry (IHC), and its expression in two TNBC cell lines (MDA-MB-231 and BT-549) was detected by western blot and qRT-PCR assays. Then the alteration of cell viability, apoptosis, migration, invasion and epithelial-mesenchymal transition (EMT) in response to overexpression or knockdown of ER-α30 was separately determined by CCK-8, Hoechst 33258, wound healing, transwell and western blot assays in two TNBC cell lines. Next, the anticancer effects of calycosin on MDA-MB-231 cells were evaluated through CCK-8, colony formation, flow cytometry, Hoechst 33258 and western blot assays, along with the role of ER-α30 in these effects and the possible downstream targets of ER-α30. In addition, the in vivo experiments were carried out using MDA-MB-231 xenograft model intraperitoneally treated with calycosin. The volume and weight of xenograft tumor were measured to evaluate the in vivo anticancer activities of calycosin, while the corresponding changes of ER-α30 expression in tumor tissues were detected by IHC.
It was demonstrated that the novel ER-α splice variant ER-α30 was primarily distributed in the nucleus of TNBC cells. Compared with normal breast tissues, ER-α30 expression was found in significantly higher levels in breast cancer tissues of ER- and progesterone receptor (PR)-negative subtype, so did in TNBC cell lines (MDA-MB-231 and BT-549) when compared to normal breast cell line MCF10A. Moreover, ER-α30 overexpression strikingly enhanced cell viability, migration, invasion and EMT progression and reduced apoptosis in TNBC cells, whereas shRNA-mediated knockdown of ER-α30 revealed the opposite results. Notably, calycosin suppressed the expression of ER-α30 in a dose-dependent manner, accompanied with the inhibition of TNBC growth and metastasis. A similar finding was observed for the xenografts generated from MDA-MB-231 cells. The treatment with calycosin suppressed the tumor growth and decreased ER-α30 expression in tumor tissues. Furthermore, this inhibition by calycosin was more pronounced in ER-α30 knockdown cells. Meanwhile, we found a positive relationship between ER-α30 and the activity of PI3K and AKT, which could also be inactivated by calycosin treatment.
For the first time, it is demonstrated that the novel estrogen receptor-α splice variant ER-α30 could function as pro-tumorigenic factor in the context of TNBC by participating in cell proliferation, apoptosis, invasion and metastasis, thus it may serve as a potential therapeutic target for TNBC therapy. Calycosin could reduce the activation of ER-α30-mediated PI3K/AKT pathway, thereby inhibited TNBC development and progression, suggesting that calycosin may be a potential therapeutic option for TNBC.
Li Y
,Hu S
,Chen Y
,Zhang X
,Gao H
,Tian J
,Chen J
... -
《-》