scholarly journals Review of: Tamoxifen and TRAIL synergistically induce apoptosis in breast cancer cells

2008 ◽  
Vol 11 (2) ◽  
Author(s):  
Alison J. Butt

Citation of original article:C. Lagadec, E. Adriaenssens, R. A. Toillon, V. Chopin, R. Romon, F. Van Coppenolle, H. Hondermarck, X. Le Bourhis. Oncogene advance online publication, 3 September 2007; doi:10.1038/sj.onc.1210749.Abstract of the original article:Tamoxifen (TAM), is widely used as a single agent in adjuvant treatment of breast cancer. Here, we investigated the effects of TAM in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in estrogen receptor-α (ER-α)-positive and -negative breast cancer cells. We showed that cotreatment with TAM and TRAIL synergistically induced apoptosis regardless of ER-α status. By contrast, cotreatment did not affect the viability of normal breast epithelial cells. Cotreatment with TAM and TRAIL in breast cancer cells decreased the levels of antiapoptotic proteins including FLIPs and Bcl-2, and enhanced the levels of proapoptotic proteins such as FADD, caspase 8, tBid, Bax and caspase 9. Furthermore, cotreatment-induced apoptosis was efficiently reduced by FADD- or Bid-siRNA, indicating the implication of both extrinsic and intrinsic pathways in synergistic apoptosis induction. Importantly, cotreatment totally arrested tumor growth in an ER-α-negative MDA-MB-231 tumor xenograft model. The abrogation of tumor growth correlated with enhanced apoptosis in tumor tissues. Our findings raise the possibility to use TAM in combination with TRAIL for breast cancers, regardless of ER-α status.

2005 ◽  
Vol 280 (29) ◽  
pp. 27022-27028 ◽  
Author(s):  
Qingnan Li ◽  
Liyu Wu ◽  
Denise K. Oelschlager ◽  
Mei Wan ◽  
Cecil R. Stockard ◽  
...  

2014 ◽  
Vol 66 ◽  
pp. 158-165 ◽  
Author(s):  
Yeong-Seon Won ◽  
Ju-Hye Lee ◽  
Soon-Jae Kwon ◽  
Jae-Yong Kim ◽  
Ki-Hun Park ◽  
...  

2021 ◽  
Author(s):  
Yue Yang ◽  
yuanyuan Lu ◽  
Chunhua Zhang ◽  
Qianqian Guo ◽  
Ting Wang ◽  
...  

Abstract Background: Breast cancer stem cells (BCSCs) are positively correlated with metastasis, chemoresistance and recurrence of breast cancer.Methods: The stemness were analyzed by qPCR and western blot, flow cytometry, cell spheroid formation assay, and tumor xenograft model. The cell viability was performed by MTT. The transcriptome analysis was used to evaluate the influence of these three compounds. The migration was analyzed by wound-healing assay, transwell invasion analysis, and metastasis model. The iron concentration was analyzed by fluorescence microscopy analysis. The lipid peroxidation and ROS level were analyzed by flow cytometry. The results were presented as mean ± SD, the analysis was Student’s t-test by using GraphPad Prism 5 software. P-values less than 0.05 were considered to be statistically significant. (*p < 0.05, **p < 0.01, ***p < 0.001).Results: Here, we tried to screen out small-molecule compounds targeting BCSCs from the phenazine library established by us before. We focused on the compounds without affecting cell viability and screened out three potential compounds (CPUL119, CPUL129, CPUL149) that can significantly attenuate the stemness of breast cancer cells, as evident by the decrease of stemness marker expression, CD44+/CD24- subpopulation, mammary spheroid-formation ability and tumor-initiating capacity. Additionally, these compounds suppressed the metastatic ability of breast cancer cells in vitro and in vivo. Combined with the transcriptome-sequencing analysis in breast cancer cells with or without the treatment of these three compounds, respectively, it was found that ferroptosis was shown on the top of the most upregulated pathways by CPUL119, CPUL129 and CPUL149. Mechanistically, we found that CPUL119, CPUL129 and CPUL149 could trigger ferroptosis by accumulating and sequestering iron in lysosomes through interacting with iron, and by regulating the expression of proteins (IRP2, TfR1, ferritin) engaged in modulating iron transport and storage. Furthermore, inhibition of ferroptosis rescued the suppression of these three compounds on the stemness of breast cancer cells. Conclusions: This study suggests that CPUL119, CPUL129 and CPUL149 can specifically inhibit the stemness of breast cancer cells through ferroptosis and may be the potential compounds for breast cancer treatment.


2018 ◽  
Vol 115 (16) ◽  
pp. 4176-4181 ◽  
Author(s):  
Silvia Vega-Rubín-de-Celis ◽  
Zhongju Zou ◽  
Álvaro F. Fernández ◽  
Bo Ci ◽  
Min Kim ◽  
...  

Allelic loss of the autophagy gene, beclin 1/BECN1, increases the risk of patients developing aggressive, including human epidermal growth factor receptor 2 (HER2)-positive, breast cancers; however, it is not known whether autophagy induction may be beneficial in preventing HER2-positive breast tumor growth. We explored the regulation of autophagy in breast cancer cells by HER2 in vitro and the effects of genetic and pharmacological strategies to increase autophagy on HER2-driven breast cancer growth in vivo. Our findings demonstrate that HER2 interacts with Beclin 1 in breast cancer cells and inhibits autophagy. Mice with increased basal autophagy due to a genetically engineered mutation in Becn1 are protected from HER2-driven mammary tumorigenesis, and HER2 fails to inhibit autophagy in primary cells derived from these mice. Moreover, treatment of mice with HER2-positive human breast cancer xenografts with the Tat-Beclin 1 autophagy-inducing peptide inhibits tumor growth as effectively as a clinically used HER2 tyrosine kinase inhibitor (TKI). This inhibition of tumor growth is associated with a robust induction of autophagy, a disruption of HER2/Beclin 1 binding, and a transcriptional signature in the tumors distinct from that observed with HER2 TKI treatment. Taken together, these findings indicate that the HER2-mediated inhibition of Beclin 1 and autophagy likely contributes to HER2-mediated tumorigenesis and that strategies to block HER2/Beclin 1 binding and/or increase autophagy may represent a new therapeutic approach for HER2-positive breast cancers.


2017 ◽  
Author(s):  
◽  
Sandy Goyette

[ACCESS RESTRICTED TO THE UNIVERSITY OF MISSOURI AT AUTHOR'S REQUEST.] Clinical trials and studies show that post-menopausal women undergoing hormone replacement therapy containing a combination of estrogen and progestin (P) have an increased risk of breast cancer compared with women taking estrogen alone or placebo. Using animal models, we previously showed that both natural and synthetic P accelerate the development of breast tumors in vivo and increase their metastasis to lymph nodes. Our studies included an assessment of medroxyprogesterone acetate (MPA), a synthetic P that is widely used clinically. Having established the deleterious effects associated with P, we sought to better understand the mechanisms underlying breast tumor growth and metastasis. We show that exposure of human breast cancer cells to synthetic P causes the overexpression of several cancer stem cell (CSC) markers, which we further demonstrate to be functionally significant, since mammosphere formation increases. Based on our observations, we contend that exposure of breast cancer cells to synthetic P, including MPA, leads to an enrichment of CSCs, which would likely support the development of P-accelerated tumors in vivo. An enriched CSC pool greatly increases the likelihood that resistance to therapy will arise, as well as raising the chance of metastasis. Our findings suggest that clinicians may be able to combat P-dependent tumor growth by blocking PR-mediated induction of CSC markers by immunotherapy, tissue-selective anti-Ps, or through a combination approach involving both immunotherapy against CD44 and small-molecule targeting of PR. We carried out a series of studies to determine whether RO 48-8076, an oxidosqualene cyclase (OSC) inhibitor, might reduce P-induced CSC expansion. In previous studies, we showed that RO diminishes tumor formation in vivo and found that it exerts its potent anti-tumor effects in part through PR degradation. This in turn reduces P-induced expression of CD44, a process we showed to be PR-dependent. Importantly, treatment of hormone-responsive breast cancer cells with RO abolished MPA-induced mammosphere formation. We therefore contend that RO may represent a novel means by which to prevent MPA-induced CSC expansion. RO treatment may also represent a novel strategy of reducing resistance to anti-hormone therapies, given that survival of stem cells following chemotherapy is the major reason why such treatments fail. In addition to being a novel treatment option for hormone-dependent breast cancers, we show that RO might also be used to treat highly aggressive triple negative breast cancers (TNBC). Herein we present in vivo data showing that RO reduces the metastasis of TNBCs to the lungs. We hypothesize that by virtue of its ability to downregulate MetAP2 protein, RO might target the metastatic cascade by inhibiting angiogenesis and cell cycle progression, or by inducing apoptosis. Further mechanistic studies are needed to elucidate the importance of RO-mediated MetAP2 downregulation. We also demonstrate that RO inhibits TNBC cell migration and invasion, though the mechanism through which RO exerts these effects requires more in-depth study. Nevertheless, we show that RO, a compound hitherto considered as simply an inhibitor of cholesterol biosynthesis, clearly possesses potent anti-cancer effects. Its potential as an agent which might be used to combat both hormone-dependent and hormone-independent breast cancers, including TNBC, warrants further investigation.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Mingzhi Zhu ◽  
Yanyan Wang ◽  
Fang Wang ◽  
Lin Li ◽  
Xinguang Qiu

Abstract Background Circular RNAs (circRNAs) have been confirmed to be relevant to the 5-fluorouracil (5-FU) resistance of breast cancer. Nevertheless, how and whether circRNA F-box and leucine-rich repeat protein 5 (circFBXL5) regulates the 5-FU resistance of breast cancer is uncertain. This study aims to explore the function and mechanism of circFBXL5 in the 5-FU resistance of breast cancer. Methods Thirty nine paired breast cancer and normal tissues were harvested. circFBXL5, microRNA-216b (miR-216b) and high-mobility group AT-hook 2 (HMGA2) abundances were examined via quantitative reverse transcription polymerase chain reaction or western blot. Cell viability, 5-FU resistance, migration, invasion, and apoptosis were tested via cell counting kit-8 assay, wound healing analysis, transwell analysis, and flow cytometry. The relationship of miR-216b and circFBXL5 or HMGA2 was tested via dual-luciferase reporter analysis and RNA pull-down assay. The impact of circFBXL5 on breast cancer tumor growth in vivo was analyzed via xenograft model. Results circFBXL5 was highly expressed in breast cancer tissues and cells, and was more upregulated in 5-FU-resistant breast cancer cells. Function experiments showed that circFBXL5 knockdown inhibited the 5-FU resistance of breast cancer by inhibiting cell migration, invasion and promoting apoptosis. In the terms of mechanism, miR-216b could be sponged by circFBXL5, and its inhibitor could also reverse the influence of circFBXL5 silencing on the 5-FU resistance of breast cancer cells. In addition, HMGA2 was a target of miR-216b, and its overexpression also reversed the regulation of miR-216b overexpression on the 5-FU resistance of breast cancer. Furthermore, circFBXL5 interference declined breast cancer tumor growth in xenograft model. Conclusion Our data showed that circFBXL5 could promote the 5-FU resistance of breast cancer by regulating miR-216b/HMGA2 axis.


Sign in / Sign up

Export Citation Format

Share Document