Plaque formation assay by crystal violet staining and cell proliferation assay by MTS. (A) A549 cells in 12-well plates treated with 1 MOI, 10 MOI, and 100 MOI of ATV and 0.4 µg cisplatin for 24, 48, 72 h and then stained with 0.4% crystal violet after plaque phenotypes formed. (B) The suppression of proliferation of A549 cells treated by 0.1, 0.2, 0.4, or 0.8 µg of cisplatin. (C) The suppression of proliferation of A549 cells infected with 1 MOI ATV or a combination of the 1 MOI ATV and 0.4 µg cisplatin. (D) The suppression of proliferation of A549 cells infected with 10 MOI ATV or a combination of the 10 MOI ATV and 0.4 µg cisplatin. (E) The suppression of proliferation of A549 cells infected with 100 MOI ATV or a combination of the 100 MOI ATV and 0.4 µg cisplatin. All values represent the mean ± SD. ** P<0.01 and * P<0.05. MOI, multiplicity of infection; SD, standard deviation; Ad5, human adenovirus serotype 5.

Plaque formation assay by crystal violet staining and cell proliferation assay by MTS. (A) A549 cells in 12-well plates treated with 1 MOI, 10 MOI, and 100 MOI of ATV and 0.4 µg cisplatin for 24, 48, 72 h and then stained with 0.4% crystal violet after plaque phenotypes formed. (B) The suppression of proliferation of A549 cells treated by 0.1, 0.2, 0.4, or 0.8 µg of cisplatin. (C) The suppression of proliferation of A549 cells infected with 1 MOI ATV or a combination of the 1 MOI ATV and 0.4 µg cisplatin. (D) The suppression of proliferation of A549 cells infected with 10 MOI ATV or a combination of the 10 MOI ATV and 0.4 µg cisplatin. (E) The suppression of proliferation of A549 cells infected with 100 MOI ATV or a combination of the 100 MOI ATV and 0.4 µg cisplatin. All values represent the mean ± SD. ** P<0.01 and * P<0.05. MOI, multiplicity of infection; SD, standard deviation; Ad5, human adenovirus serotype 5.

Source publication
Article
Full-text available
The effect of the combination of a recombinant adenovirus (ATV) expressing a specific apoptin protein and cisplatin on human lung cancer cells (A549 cells) was determined. The inhibitory effects of ATV and cisplatin, ATV alone, or cisplatin alone on the migration and invasion of A549 cells were evaluated in vitro using cell proliferation, wound hea...

Contexts in source publication

Context 1
... inhibition effect of A549 cells. The results for the inhibition of A549 cell proliferation are shown in Fig. 1A-E. The plaque formation assay is shown in Fig. 1A. The MTS assays showed that the proliferation of A549 cells was suppressed significantly at 24, 48 and 72 h by 0.8 µg cisplatin (P<0.01). However, this inhibitory effect may be caused by itself toxic. The proliferation of A549 cells was suppressed significantly at 24 and 48 h by 0.4 µg ...
Context 2
... inhibition effect of A549 cells. The results for the inhibition of A549 cell proliferation are shown in Fig. 1A-E. The plaque formation assay is shown in Fig. 1A. The MTS assays showed that the proliferation of A549 cells was suppressed significantly at 24, 48 and 72 h by 0.8 µg cisplatin (P<0.01). However, this inhibitory effect may be caused by itself toxic. The proliferation of A549 cells was suppressed significantly at 24 and 48 h by 0.4 µg cisplatin (P<0.05) and the inhibitory effect of ...
Context 3
... of A549 cells could be suppressed by ATV or ATV combined with cisplatin in a dose-and time-dependent manner. The inhibition rate of A549 cells was increased significantly by 0.4 µg (P<0.05) and 0.8 µg (P<0.01) cisplatin compared to 0.1 and 0.2 µg cisplatin, while the higher toxicity of 0.8 µg cisplatin made it unfit for subsequent experiments (Fig. 1B). The growth suppression of A549 cells was not increased by 1 MOI ATV. However, increased growth suppression of A549 cells was observed by 1 MOI ATV and 0.4 µg cisplatin, which reached a peak of 3.11-fold greater than that of the 1 MOI ATV-infected cells at 72 h (Fig. 1C). The growth suppression induced by 10 MOI ATV and 0.4 µg ...
Context 4
... higher toxicity of 0.8 µg cisplatin made it unfit for subsequent experiments (Fig. 1B). The growth suppression of A549 cells was not increased by 1 MOI ATV. However, increased growth suppression of A549 cells was observed by 1 MOI ATV and 0.4 µg cisplatin, which reached a peak of 3.11-fold greater than that of the 1 MOI ATV-infected cells at 72 h (Fig. 1C). The growth suppression induced by 10 MOI ATV and 0.4 µg cisplatin was significantly higher than that of the control group (P<0.01). Meanwhile, the combination of 10 MOI and 0.4 µg cisplatin caused increasing growth suppression from 24 to 72 h, which reached a peak of 2.5-fold greater than that of the 10 MOI ATV-infected cells at 72 h ...
Context 5
... h (Fig. 1C). The growth suppression induced by 10 MOI ATV and 0.4 µg cisplatin was significantly higher than that of the control group (P<0.01). Meanwhile, the combination of 10 MOI and 0.4 µg cisplatin caused increasing growth suppression from 24 to 72 h, which reached a peak of 2.5-fold greater than that of the 10 MOI ATV-infected cells at 72 h (Fig. 1D). The growth suppression rate of 100 MOI ATV infected-cells was also significantly higher than that of the control group (P<0.01). However, the growth suppression was not significantly different between the combination of 100 MOI ATV 100 with 0.4 µg and 100 MOI ATV (P>0.05) (Fig. 1E). Therefore, the combination of ATV at an MOI of 10 or ...
Context 6
... greater than that of the 10 MOI ATV-infected cells at 72 h (Fig. 1D). The growth suppression rate of 100 MOI ATV infected-cells was also significantly higher than that of the control group (P<0.01). However, the growth suppression was not significantly different between the combination of 100 MOI ATV 100 with 0.4 µg and 100 MOI ATV (P>0.05) (Fig. 1E). Therefore, the combination of ATV at an MOI of 10 or 100 with 0.4 µg cisplatin showed a synergistic effect on the inhibition of the growth of A549 ...

Similar publications

Article
Full-text available
Objective To examine the effects of pachyman in combination with vinorelbine and cisplatin on tumor growth and the expression of epidermal growth factor receptor (EGFR) and K-ras in a mouse model of lung cancer induced using the human lung cancer cell line A549, and to investigate the molecular mechanisms underlying the antitumor effects of pachyma...

Citations

... These four recombinant oncolytic adenoviruses have been confirmed in previous studies to have strong inhibitory effects on the growth of lung cancer, prostate cancer and liver cancer. 8,9 Bioluminescent imaging in vivo is a highly useful visualization technique that can be used to track cells and analyse tissue activity and gene behaviour in vivo. 10 Bioluminescence imaging in vivo is characterized by light scattering and is associated with a unique imaging advantage 11 ; since there is almost no endogenous luminescence in tissues and cells, the endogenous signal-to-noise ratio is low; therefore, background interference can be effectively eliminated, and the bioluminescence signal can be clearly observed in complex organisms. ...
Article
Full-text available
In this study, we compared the inhibitory effects of recombinant oncolytic adenovirus (Ad‐apoptin‐hTERTp‐E1a, Ad‐VT) with that of doxorubicin (DOX), a first‐line chemotherapy drug, and tamoxifen (TAM), an endocrine therapy drug, on the proliferation of breast cancer cells. We found that Ad‐VT could effectively inhibit the proliferation of breast cancer cells (p < 0.01); the inhibition rate of Ad‐VT on normal mammary epithelial MCF‐10A cells was less than 20%. DOX can effectively inhibit the proliferation of breast cancer cells and also has a strong inhibitory effect on MCF‐10A cells (p < 0.01). TAM also has a strong inhibitory effect on breast cancer cells, among which the oestrogen‐dependent MCF‐7 cell inhibition was stronger (p < 0.01), At higher concentrations, TAM also had a high rate of inhibition (>70%) on the proliferation of MCF‐10A cells. We also found that both recombinant adenovirus and both drugs could successfully induce tumour cell apoptosis. Further Western blot results showed that the recombinant adenovirus killed breast cancer cells through the endogenous apoptotic pathway. Analysis of the nude mouse subcutaneous breast cancer model showed that Ad‐VT significantly inhibited tumour growth (the luminescence rate of cancer cells was reduced by more than 90%) and improved the survival rate of tumour‐bearing mice (p < 0.01). Compared with DOX and TAM, Ad‐VT has a significant inhibitory effect on breast cancer cells, but almost no inhibitory effect on normal breast epithelial cells, and this inhibitory effect is mainly through the endogenous apoptotic pathway. These results indicate that Ad‐VT has significant potential as a drug for the treatment of breast cancer.
... In this study, we first analyzed the cytotoxic effect of apoptin on liver cancer cells through a variety of in vivo and in vitro experiments. We found that apoptin can significantly cause the death of liver cancer cells, and similar results were previously obtained with other tumor cell types [4,7,15,[17][18][19]. Moreover, we also found that the killing ability of apoptin of liver cancer cells is higher than that observed with other tumor cells, indicating that apoptin may have a higher potential therapeutic effect in the treatment of liver cancer. ...
Article
Full-text available
Background Apoptin, as a tumor-specific pro-apoptotic protein, plays an important anti-tumoral role, but its mechanism of autophagy activation and the interaction between autophagy and apoptosis have not been accurately elucidated. Here, we studied the mechanism of apoptin-induced apoptosis and autophagy and the interaction between two processes. Methods Using crystal violet staining and the CCK-8 assay, we analyzed the effect of apoptin in the inhibition of liver cancer cells in vitro and analyzed the effect of inhibiting liver cancer in vivo by establishing a nude mouse tumor model. Flow cytometry and fluorescence staining were used to analyze the main types of apoptin-induced apoptosis and autophagy. Subsequently, the relationship between the two events was also analyzed. Flow cytometry was used to analyze the effect of ROS on apoptin-mediated apoptosis and autophagy mediated by apoptin. The effect of ROS on two phenomena was analyzed. Finally, the role of key genes involved in autophagy was analyzed using gene silencing. Results The results showed that apoptin can significantly increase the apoptosis and autophagy of liver cancer cells, and that apoptin can cause mitophagy through the increase in the expression of NIX protein. Apoptin can also significantly increase the level of cellular ROS, involved in apoptin-mediated autophagy and apoptosis of liver cancer cells. The change of ROS may be a key factor causing apoptosis and autophagy. Conclusion The above results indicate that the increase in ROS levels after apoptin treatment of liver cancer cells leads to the loss of mitochondrial transmembrane potential, resulting in endogenous apoptosis and mitophagy through the recruitment of NIX. Therefore, ROS may be a key factor connecting endogenous apoptosis and autophagy induced by apoptin in liver cancer cells. Graphical abstract
... Hoechst, Annexin V, JC-1 staining, and Western blotting were employed to analyze the inhibition pathway of Ad-hTERT-E1a-Apoptin on tumor cells. Infection of tumor cells with Ad-hTERT-E-1a-Apoptin caused a marked increase in the activity levels of caspases-3, 6 and 7 66,[70][71][72][73][74][75][76][77][78][79][80][81] . Significant quantities of cytochrome c were detected in the cytosol of the Ad-hTERT-E1a-Apoptin-infected cancer cells 70,72,79 . ...
... Significant quantities of cytochrome c were detected in the cytosol of the Ad-hTERT-E1a-Apoptin-infected cancer cells 70,72,79 . Transwell assay showed that Ad-hTERT-E1a-Apoptin inhibited tumor migration and invasion 73,80,81 . In summary, the Ad-hTERT-E1a-Apoptin can promote tumor cell apoptosis through intrisic pathway. ...
... In addition, we also studied the antitumor effects of the combination of Ad-hTERT-E1a-Apoptin and chemotherapy drugs, including cisplatin, doxorubicin, gemcitabine, and peclitaxel 73,78,80,81 . Ad-hTERT-E1a-Apoptin can not only inhibit the growth of tumor cells and promote the apoptosis of tumor cells, but also has a significant synergistic effect on tumor inhibition with chemotherapy drugs. ...
Article
Full-text available
Apoptin is a small molecular weight protein derived from chicken anemia virus, which can induce the apoptosis of transformed cells and tumor cells and leave primary and nontransformed cells unharmed. Apoptin's cell localization depends on its own phosphorylation state and cell type. In tumor cells, phosphorylated apoptin enters the nucleus and induces apoptosis. While, in normal cells apoptin mainly exists in the cytoplasm. Apoptin, as a disordered protein in cells, interacts with many proteins in cell signal pathways to induce apoptosis of tumor cells. The specific mechanism of apoptosis induced by apoptin has not been completely elucidated. Therefore, apoptin has become a potential anticancer agent. This review summarizes the research results of apoptin in our laboratory and reveals the specific antitumor mechanism of apoptin expressed by oncolytic virus vector on a variety of tumor cells and mouse models.
Article
Lung cancer has a high morbidity rate worldwide due to its resistance to therapy. So new treatment options are needed to improve the outcomes of lung cancer treatment. This study aimed to evaluate the effectiveness of oncolytic viruses (OVs) as a new type of cancer treatment. In this study, 158 articles from PubMed and Scopus from 1994 to 2022 were reviewed on the effectiveness of OVs in the treatment of lung cancer. The oncolytic properties of eight categories of OVs and their interactions with treatment options were investigated. OVs can be applied as a promising immunotherapy option, as they are reproduced selectively in different types of cancer cells, cause tumor cell lysis and trigger efficient immune responses.
Preprint
Full-text available
Background: Apoptin, as a tumor-specific pro-apoptotic protein, apoptin plays an important role in the field of anti-tumor, but its autophagy activation mechanism and the interaction between autophagy and apoptosis have not been accurately elucidated. Here, we studied the mechanism of apoptosis and autophagy induced by apoptin and the interaction between autophagy and apoptosis. Methods: Through crystal violet staining and CCK-8 assay, we analyzed the effect of apoptin in inhibiting liver cancer in vitro, and also analyzed the effect of inhibiting liver cancer in vivo by establishing a nude mouse tumor model. Flow cytometry and fluorescence staining were used to analyze the main types of apoptosis and autophagy induced by apoptin. Subsequently, the relationship between apoptosis and autophagy induced by apoptin was analyzed. Then, flow cytometry was used to analyze the effect of ROS on apoptosis and autophagy mediated by apoptin. Then, the affect of ROS on apoptosis and autophagy mediated by apoptin was analyzed. Finally, the key genes leading to autophagy were analyzed by silencing different genes. Results: The results showed that apoptin can significantly increase the apoptosis and autophagy of liver cancer cells, and apoptin can cause mitophagy through the increase of NIX protein. Apoptin can also significantly reduce the level of cellular ROS, which is related to the autophagy and apoptosis of liver cancer cells caused by apoptin. The change of ROS may be a key factor causing apoptosis and autophagy. Conclusion: The above results indicate that the increase of ROS level after apoptin treatment of liver cancer cells leads to the loss of mitochondrial transmembrane potential, which leads to endogenous apoptosis and mitophagy while recruiting NIX. Therefore, ROS may be a key factor connecting endogenous apoptosis and mitophagy induced by apoptin in liver cancer cells.
Article
Background: To explore the suppressive effect of Apoptin-loaded oncolytic adenovirus (Ad-VT) on luciferase-labeled human melanoma cells in vitro and in vivo. Methods: The stable luciferase-expressing human melanoma cells A375-luc or M14-luc were obtained by transfecting the plasmid pGL4.51 and selection with G418, followed by luciferase activity, genetic stability and bioluminescence intensity assays. In vitro, the inhibitory effects of Ad-VT on A375-luc or M14-luc were evaluated using the MTS cell proliferation, FITC-Annexin V apoptosis detection, transwell migration, Matrigel invasion and scratch assays. The inhibition pathway in Ad-VT-infected A375-luc and M14-luc cells were analyzed by JC-1 staining and Western-blot detection of mitochondrial apoptosis-related proteins. In vivo, the suppressive effects of Ad-VT on A375-luc or M14-luc were assessed by living imaging technology, tumor volume, bioluminescence intensity, survival curves and immunohistochemical analysis of the tumors from the xenograft tumor model BALB/c nude mice. Results: The growth and migration of A375-luc and M14-luc were significantly inhibited by Ad-VT in vitro. The evaluations of A375-luc and M14-luc tumor models in BALB/c nude mice were successfully performed using living imaging technology. Ad-VT had an anti-tumor effect by reducing tumor growth and increasing survival in vivo. Ad-VT significantly changed the mitochondrial membrane potential by triggering the the mitochondrial release of apoptosis-related proteins, AIF (apoptosis inducing factor), ARTS (Apoptosis-Related Proteins), and Cyto-c (cytochrome c) from the mitochondria. Conclusion: Ad-VT reduced the mitochondrial membrane potential in A375-luc or M14-luc cells and induced the mitochondrial release of AIF, ARTS and Cyto-C. Ad-VT induced apoptosis in A375-luc or M14-luc cells via the mitochondrial apoptotic pathway.
Article
Full-text available
Apoptin is a protein that specifically induces apoptosis in tumor cells. The anti-tumorigenic functions of Apoptin, including autophagy activation and its interaction with apoptosis, have not been precisely elucidated. Here we investigate the main pathways of apoptin-mediated killing of human liver cancer cells, as well as its putative role in autophagy and apoptosis. The anti-proliferative effect of apoptin in liver cancer cells was analyzed in vitro by crystal violet staining and MTS detection, and also in vivo using a tumor-based model. The main pathway related to apoptin-induced growth inhibition in vitro was evaluated by flow cytometry and fluorescence staining. The relationship between apoptosis and autophagy on apoptin-treating cells was analyzed using apoptosis and autophagy inhibitors, mitochondrial staining, Annexin V-FITC/PI flow detection, LC3 staining, and western blotting. The effect of ROS toward the apoptosis and autophagy of apoptin-treating cells was also evaluated by ROS detection, Annexin V-FITC/PI flow detection, LC3 staining, and western blotting. Inhibition of apoptosis in apoptin-treating liver cancer cells significantly reduced the autophagy levels in vitro. The overall inhibition increased from 12 h and the effect was most obvious at 48 h. Inhibition of autophagy could increase apoptin-induced apoptosis of cells in a time-dependent manner, reaching its peak at 24 h. Apoptin significantly alters ROS levels in liver cancer cells, and this effect is directly related to apoptosis and autophagy. ROS appears to be the key factor linking apoptin-induced autophagy and apoptosis through the mitochondria in liver cancer cells. Therefore, evaluating the interaction between apoptin-induced apoptosis and autophagy is a promising step for the development of alternate tumor therapies.