scholarly journals MPPa-PDT suppresses breast tumor migration/invasion by inhibiting Akt-NF-κB-dependent MMP-9 expression via ROS

BMC Cancer ◽  
2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Liyi Huang ◽  
Haidan Lin ◽  
Qing Chen ◽  
Lehua Yu ◽  
Dingqun Bai

Abstract Background Breast cancer is one of the most commonly diagnosed cancers in women, with high morbidity and mortality. Tumor metastasis is implicated in most breast cancer deaths; thus, inhibiting metastasis may provide a therapeutic direction for breast cancer. In the present study, pyropheophorbide-α methyl ester-mediated photodynamic therapy (MPPa-PDT) was used to inhibit metastasis in MCF-7 breast cancer cells. Methods Uptake of MPPa was detected by fluorescence microscopy. Cell viability was evaluated by the Cell Counting Kit-8 (CCK-8). ROS generation was detected by 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA). The migration of cells was assessed by wound healing assay, and invasion ability was assessed by Matrigel invasion assay. Levels of MMP2 and MMP9 were measured by PCR. Akt, phospho-Akt (Ser473), phospho-NF-κB p65 (Ser536) and NF-κB p65 were measured by western blotting. The F-actin cytoskeleton was observed by immunofluorescence. Lung tissue was visualized by hematoxylin and eosin staining. Results Following MPPa-PDT, migration and invasion were decreased in the MCF-7 cells. MPPa-PDT downregulated the expression of MMP2 and MMP9, which are responsible for the initiation of metastasis. MPPa-PDT reduced the phosphorylation of Akt and NF-κB. MPPa-PDT also reduced the expression of F-actin in cytoskeleton in MCF-7 cells. These effects were blocked by the reactive oxygen species scavenger NAC or the Akt activator SC79, while the PI3K inhibitor LY294002 or the Akt inhibitor triciribine enhanced these effects. Moreover, MPPa-PDT inhibited tumor metastasis and destroyed F-actin in vivo. Conclusion Taken together, these results demonstrate that MPPa-PDT inhibits the metastasis of MCF-7 cells both in vitro and in vivo and may be involved in the Akt/NF-κB-dependent MMP-9 signaling pathway. Thus, MPPa-PDT may be a promising treatment to inhibit metastasis.

2019 ◽  
Author(s):  
Liyi Huang ◽  
Haidan Lin ◽  
Qing Chen ◽  
Lehua Yu ◽  
Dingqun Bai

Abstract Background: breast cancer is the most commonly women cancer and most breast cancer deaths are related to tumor metastasis. Therefore, inhibiting metastasis may provide a therapeutic treatment for breast cancer. In the present study, pyropheophorbide-α methyl ester mediated photodynamic therapy (MPPa-PDT) was used to inhibit metastasis in breast cancer cells MCF-7. Methods: Uptake of MPPa was detected by fluorescence microscope. Cell viability was evaluated by CCK-8. Generation of ROS were detected by DCFH-DA. Migration of cells was assessed by wound healing assay and invasion ability was assessed by Matrigel invasion assay. Levels of MMP2 and MMP9 were measured by PCR. Akt, Phospho-Akt, Phospho-NF-kB p65 and NF-kB p65 were measured by western blotting. F-actin cytoskeleton was observed by immunofluorescence. Lung organs were stained with Hematoxylin and Eosin. Results: Following MPPa-PDT, migration and invasion were decreased in the MCF-7 cells. MPPa-PDT down-regulated expression of MMP2 and MMP9 which is responsible for metastasis. MPPa-PDT reduced the phosphorylation of AKT and NF-κB. MPPa-PDT also destroyed cytoskeleton F-actin in MCF-7 cells. These effects were blocked by the reactive oxygen scavenger NAC or AKT activator SC79 while PI3K inhibitor LY294002 or AKT inhibitor Triciribine increased these effects. Moreover, MPPa-PDT inhibited tumor metastasis and destroyed F-actin in vivo. Conclusion: taken together, these results demonstrated that MPPa-PDT inhibits metastasis of MCF-7 cells both in vitro and vivo, and that may involve in AKT-NF-κB-dependent MMP-9 signaling pathway. Thus, MPPa-PDT may be a promising therapeutic treatment to inhibit metastasis.


2021 ◽  
Vol 71 (2) ◽  
pp. 245-266
Author(s):  
Qingfang Zeng ◽  
Cairong Luo ◽  
Junlae Cho ◽  
Donna Lai ◽  
Xiangchun Shen ◽  
...  

AbstractTryptanthrin is an indole quinazoline alkaloid from the indigo-bearing plants, such as Isatis indigotica Fort. Typically, this natural compound shows a variety of pharmacological activities such as antitumor, antibacterial, anti-inflammatory and antioxidant effects. This study was conducted to assess the antitumor activity of tryptanthrin in breast cancer models both in vitro and in vivo, and to explore the important role of the inflammatory tumor microenvironment (TME) in the antitumor effects of tryptanthrin. Human breast adenocarcinoma MCF-7 cells were used to assess the antitumor effect of tryptanthrin in vitro. MTT assay and colony formation assay were carried out to monitor the antiproliferative effect of tryptanthrin (1.56~50.0 μmol L−1) on inhibiting the proliferation and colony formation of MCF-7 cells, respectively. The migration and invasion of MCF-7 cells were evaluated by wound healing assay and Transwell chamber assay, respectively. Moreover, the 4T1 murine breast cancer model was established to examine the pharmacological activity of tryptanthrin, and three groups with different doses of tryptanthrin (25, 50 and 100 mg kg−1) were set in study. Additionally, tumor volumes and organ coefficients were measured and calculated. After two weeks of tryptanthrin treatment, samples from serum, tumor tissue and different organs from tumor-bearing mice were collected, and the enzyme-linked immunosorbent assay (ELISA) was performed to assess the regulation of inflammatory molecules in mouse serum. Additionally, pathological examinations of tumor tissues and organs from mice were evaluated through hematoxylin and eosin (H&E) staining. The expression of inflammatory proteins in tumor tissues was measured by immunohistochemistry (IHC) and Western blotting. Tryptanthrin inhibited the proliferation, migration and invasion of MCF-7 cells, up-regulated the protein level of E-cadherin, and down-regulated those of MMP-2 and Snail, as suggested by the MCF-7 cell experiment. According to the results from in vivo experiment, tryptanthrin was effective in inhibiting tumor growth, and it showed favorable safety without inducing the fluctuations of body mass and organ coefficient (p > 0.05). In addition, tryptanthrin also suppressed the expression levels of NOS1, COX-2 and NF-κB in mouse tumor tissues, and regulated those of IL-2, IL-10 and TNF-α in the serum of tumor cells-transplanted mice. Tryptanthrin exerted its anti-breast cancer activities through modulating the inflammatory TME both in vitro and in vivo.


2019 ◽  
Vol 166 (6) ◽  
pp. 485-493 ◽  
Author(s):  
Anyun Mao ◽  
Maojian Chen ◽  
Qinghong Qin ◽  
Zhijie Liang ◽  
Wei Jiang ◽  
...  

Abstract It has been generally confirmed that zinc finger and BTB domain containing 7A (ZBTB7A) plays an important role in the occurrence and progression of malignant tumours, but the promotion or inhibition effect is related to tumour type. The mechanism between ZBTB7A and breast cancer is not well understood, so further research is needed. In this study, we first investigated the expression of ZBTB7A in tissue samples of clinical breast cancer patients, MDA-MB-231, MCF-7 and MCF-10A cells. Second, we overexpressed the ZBTB7A in MCF-7 cells and silenced the ZBTB7A in MDA-MB-231 cells using lentivirus transfection technology, respectively, and verified the effect of ZBTB7A on migration and invasion of breast cancer cell lines through in vitro cell function experiments, such as wound-healing assay, migration and invasion assay, quantitative real time reverse transcriptase (qRT-PCR) and western blot. Then, the correlation between the above influences, epithelial–mesenchymal transition (EMT) and NF-κB was analysed. Finally, in vivo tumour transplantation model in nude mice was established to verified the effect of ZBTB7A on metastasis of breast cancer MDA-MB-231 cells. In conclusion, ZBTB7A is highly expressed in cancer tissue, breast cancer cell line MDA-MB-231 and MCF-7. Meanwhile, the high expression of ZBTB7A may promote cell migration, invasion and tumour metastasis, which may be related to EMT events by regulating NF-κB.


Author(s):  
Lihua Wang ◽  
Canwei Wang ◽  
Zheying Tao ◽  
Liqian Zhao ◽  
Zheng Zhu ◽  
...  

Abstract Background Breast cancer is the most prevalent cancer among women worldwide. WZ35, an analog of curcumin, has been demonstrated to remarkably improve the pharmacokinetic profiles in vivo compared with curcumin. WZ35 exhibits promising antitumor activity in gastric cancer, HCC, colon cancer. However, antitumor effects of WZ35 in breast cancer and its underlying molecular mechanisms remain unclear. Methods CCK8, Flow cytometry and transwell assays were used to measure cell proliferation, cell cycle arrest, apoptosis, cell migration and invasion. We constructed xenograft mouse model and lung metastasis model to assess the antitumor activities of WZ35 in vivo. To explore the underlying molecular mechanisms of WZ35, we performed a series of overexpression and knockdown experiments. The cellular oxygen consumption rates (OCRs) was measured to assess mitochondrial dysfunction. Results We found that treatment of breast cancer cells with WZ35 exerts stronger anti-tumor activities than curcumin both in vitro and in vivo. Mechanistically, our research showed that WZ35 induced reactive oxygen species (ROS) generation and subsequent YAP mediated JNK activation in breast cancer cells. Abrogation of ROS production markedly attenuated WZ35 induced anti-tumor activities as well as YAP and JNK activation. In addition, ROS mediated YAP and JNK activation induced mitochondrial dysfunction in breast cancer cells. Conclusion Our study showed that novel anti-cancer mechanisms of WZ35 in breast cancer cells and ROS-YAP-JNK pathway might be a potential therapeutic target for the treatment of breast cancer patients.


2019 ◽  
Author(s):  
Liyi Huang ◽  
Haidan Lin ◽  
Qing Chen ◽  
Lehua Yu ◽  
dingqun bai

Abstract Abstract Background: Breast cancer is one of the most commonly diagnosed cancers in women, with high morbidity and mortality. Tumor metastasis is implicated in most breast cancer deaths; thus, inhibiting metastasis may provide a therapeutic direction for breast cancer. In the present study, pyropheophorbide-α methyl ester-mediated photodynamic therapy (MPPa-PDT) was used to inhibit metastasis in MCF-7 breast cancer cells. Methods: Uptake of MPPa was detected by fluorescence microscopy. Cell viability was evaluated by the Cell Counting Kit-8 (CCK-8). ROS generation was detected by 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA). The migration of cells was assessed by wound healing assay, and invasion ability was assessed by Matrigel invasion assay. Levels of MMP2 and MMP9 were measured by PCR. Akt, phospho-Akt (Ser473), phospho-NF-κB p65 (Ser536) and NF-κB p65 were measured by western blotting. The F-actin cytoskeleton was observed by immunofluorescence. Lung tissue was visualized by hematoxylin and eosin staining. Results: Following MPPa-PDT, migration and invasion were decreased in the MCF-7 cells. MPPa-PDT downregulated the expression of MMP2 and MMP9, which are responsible for the initiation of metastasis. MPPa-PDT reduced the phosphorylation of Akt and NF-κB. MPPa-PDT also reduced and destroyed the F-actin cytoskeleton in MCF-7 cells. These effects were blocked by the reactive oxygen species scavenger NAC or the Akt activator SC79, while the PI3K inhibitor LY294002 or the Akt inhibitor triciribine enhanced these effects. Moreover, MPPa-PDT inhibited tumor metastasis and destroyed F-actin in vivo. Conclusion: Taken together, these results demonstrate that MPPa-PDT inhibits the metastasis of MCF-7 cells both in vitro and in vivo and may be involved in the Akt/NF-κB-dependent MMP-9 signaling pathway. Thus, MPPa-PDT may be a promising treatment to inhibit metastasis. Key words: photodynamic therapy, reactive oxygen species, breast tumor, migration, invasion


2021 ◽  
Vol 12 (7) ◽  
Author(s):  
Xiaohui Zhang ◽  
Fangyuan Li ◽  
Yidong Zhou ◽  
Feng Mao ◽  
Yan Lin ◽  
...  

AbstractLong noncoding ribonucleic acids (LncRNAs) have been found to be involved in the proliferation, apoptosis, invasion, migration, and other pathological processes of triple-negative breast cancer (TNBC). Expression of the lncRNA actin filament-associated protein 1 antisense RNA1 (AFAP1-AS1) has been found to be significantly higher in TNBC than in other subtypes or in normal tissue samples, but the specific mechanism by which AFAP1-AS1 affects the occurrence and development of TNBC is yet to be revealed. In this study, we used Cell Counting Kit-8 (CCK-8), colony formation, wound healing migration, Transwell invasion, and nude mouse xenograft assays to confirm the role of AFAP1-AS1 in the proliferation, migration of TNBC cells in vitro and in vivo. In addition, we performed bioinformatics analyses, reverse transcriptase quantitative polymerase chain reaction (RT-qPCR), western blot (WB), and dual-luciferase reporter assays (dual-LRA) to confirm interaction among AFAP1-AS1, micro-RNA 2110 (miR-2110), and Sp1 transcription factor (Sp1). We found that silencing AFAP1-AS1 and Sp1 or upregulating miR-2110 suppressed the proliferation, migration, and invasion of MDA–MB-231 and MDA–MB-468 cells in vitro as well as tumor growth in vivo. Mechanistically, the dual-LRA highlighted that miR-2110 was an inhibitory target of AFAP1-AS1, and that AFAP1-AS1 functioned as a miR-2110 sponge to increase Sp1 expression. AFAP1-AS1 silencing led to a reduction in Sp1 mRNA and protein levels, which could be reversed by joint transfection with miR-2110 inhibitor. Our findings demonstrated that AFAP1-AS1 could modulate the progression of breast cancer cells and affect tumorigenesis in mice by acting as a miR-2110 sponge, resulting in regulation of Sp1 expression. Therefore, AFAP1-AS1 could play a pivotal role in the treatment of TNBC.


2021 ◽  
Author(s):  
Aijun Chen ◽  
Shushu Zhang ◽  
Dandan Zhang ◽  
Xingjiang Hu ◽  
Nana Xu ◽  
...  

Abstract Many studies have shown that hederacolchiside A1 (HA1) is an important anticancer saponin, although its mechanism of action and in vivo investigations are still lacking. Our previous results revealed that HA1 may have the potential to treat breast cancer. Therefore, we attempted to verify the potential anti-breast cancer effect of HA1 in vitro and in vivo. MTT, flow cytometry, DCFH-DA fluorescence microscopy, and western blotting were used to evaluate the activities and mechanisms of action of HA1. Athymic nude mice were used to demonstrate the antitumor activity of HA1 in vivo. HA1 exhibited significant cytotoxic effects on HepG2, MCF-7, MDA-MB-231, SKBr-3, HT-29, and HCT-116 cells, especially MCF-7 cells. HA1 blocked the sub-G1 and G0/G1 phases, induced apoptosis, promoted reactive oxygen species (ROS) generation, and decreased the mitochondrial membrane potential of MCF-7 cells. HA1 upregulated Bax and downregulated Bcl-2 levels and activated caspase-9 and caspase-3 in MCF-7 cells Meanwhile, HA1 inhibited the phosphorylation of JAK2/STAT3 in MCF-7 cells. In addition, 50 and 100 mg/kg HA1 significantly inhibited the growth of transplanted tumors with inhibition rates of 46.95 ± 26.72% and 48.45 ± 22.36%, respectively. This preliminary study demonstrated that HA1 could inhibit proliferation and induce the apoptosis of MCF-7 cells via ROS-mediated activation of the mitochondrial apoptotic pathway and JAK2/STAT3 inactivation. HA1 may therefore be developed as a novel agent for breast cancer therapy.


2021 ◽  
Author(s):  
Shuo Zhang ◽  
Lei Zhou ◽  
XiaoYan Jiang ◽  
HongHui Ni ◽  
ShuiYing Yang ◽  
...  

Abstract Background: Breast cancer is the leading cause of cancer-related death in women worldwide. Hirudin has been shown to inhibit the growth and metastasis of several types of cancers in experimental tumor models. However, whether hirudin exerts antitumor effects on breast adenocarcinoma cells has not yet been investigated. The objective of this study was to evaluate the antitumor effects and explore the underlying mechanisms of hirudin in breast adenocarcinoma MCF-7 cells. Methods: The viability of MCF-7 cells was assayed by Cell Counting Kit-8. The adhesion ability of the cells was evaluated by cell adhesion assay. Besides, cell migration was detected by wound healing assay. Cell invasion was examined using Transwell chamber assay. The underlying molecular mechanism was investigated by immunofluorescence. In addition, In vivo zebrafish xenograft model was used to verify the proliferation and metastasis of hirudin on MCF-7 cells. Results: The results showed that hirudin significantly inhibited the cell viability and suppressed cell adhesion, migration, invasion compared with the control group. Importantly, hirudin significantly decreased the expression of CHD1L, MDM2 protein, and increased the expression of p53 protein. Moreover, the zebrafish xenograft study revealed that hirudin inhibited the proliferation and metastasis of MCF-7 cells in vivo. Conclusion: The present findings demonstrate that hirudin suppressed metastasis of MCF-7 cells and the mechanism may involve with the CHD1L/MDM2/p53 axis. Hirudin is a promising antineoplastic agent for the treatment of breast cancer with significant antimetastatic activities. Keywords: Breast cancer, Hirudin, Metastasis, Zebrafish, CHD1L


2020 ◽  
Author(s):  
Chuang Qi ◽  
Xianxiong Qin ◽  
Zuozhi Zhou ◽  
Yan Wang ◽  
Qin Yang ◽  
...  

Abstract Background Circ_0072995 is a novel identified circRNA and has been identified to involve in the metastasis of breast cancer. However, the detailed function and mechanism of circ_0072995 in the biological property of breast cancer cell remain vague. Methods The expression of circ_0072995, microRNA (miR)-149-5p and serine hydroxymethyltransferase 2 (SHMT2) mRNA was detected using quantitative real-time polymerase chain reaction. Western blot was used to detect levels of SHMT2, hexokinase-2 (HK-2), lactate dehydrogenase a chain (LDHA), glucose transporter 1 (GLUT1) and phosphoinositide 3-kinase (PI3K)/p-protein kinase B (AKT) pathway/mammalian target of rapamycin (mTOR) pathway-related protein. Cell proliferation, apoptosis, migration, and invasion were analyzed using cell counting kit-8 assay, flow cytometry, caspase-3 activity analysis, cell adhesion assay and transwell assay, respectively. Glucose metabolism was calculated by measuring glucose uptake, lactate production, and adenosine triphosphate (ATP) levels. The interaction between miR-149-5p and circ_0072995 or SHMT2 was confirmed by dual-luciferase reporter assay. In vivo tumorigenesis was performed using the murine xenograft model. Results Circ_0072995 and SHMT2 were up-regulated in breast cancer tissues and cell lines, and knockdown of circ_0072995 or SHMT2 suppressed cell malignant properties and anaerobic glycolysis; importantly, SHMT2 overexpression attenuated the anticancer action of si-Circ_0072995 in breast cancer. Besides, we also found miR-149-5p directly bound to circ_0072995 or SHMT2 in breast cancer cells, and circ_0072995 promoted the expression of SHMT2 by competitively binding to miR-149-5p. Moreover, circ_0072995 activated PI3K/AKT/mTOR pathway via elevating SHMT2 through miR-149-5p in vitro and in vivo. Additionally, xenograft tumors analysis showed circ_0072995 silence suppressed tumor growth via regulating SHMT2 and miR-149-5p. Conclusion This study demonstrated that circ_0072995 promoted cell malignant phenotypes and anaerobic glycolysis in breast cancer via up-regulating SHMT2 through sponging miR-149-5p, and activated PI3K/AKT/mTOR pathway via miR-149-5p/ SHMT2 axis, indicating a promising molecular target for breast cancer treatment.


Sign in / Sign up

Export Citation Format

Share Document