Share this post on:

Been shown to be a critical mechanism underlying SIPS [12,15,17,18].Resveratrol-Induced Senescence in Cancer CellsFigure 6. RV induces Nox5 mRNA expression in LSCLC cells. (A) Cells were treated with 50 mM of RV or DMSO as vehicle control. Twenty-four hours after RV treatment, the expression levels of Nox1, Nox2, and Nox5 mRNAs were determined using real-time RT-PCR. (B) The expression levels of SOD1, SOD2 and TXN in A549 cells were determined by real-time RT-PCR. (C) The expression levels of Nox1, Nox2, and Nox5 in H460 cells are presented as fold change (mean 6 SEM). (D) The expression levels of SOD1, SOD2 and TXN in H460 cells are presented. **, p,0.001 vs. control. doi:10.1371/journal.pone.0060065.gHere we show that RV-induced premature senescence is associated with 23977191 increased expression of p53 and p21 in NSCLC cells, Calyculin A price suggesting that activation of the p53 21 pathway may play an important role in modulating RV-induced senescence in lung cancer cells. More importantly, it was also found that RV-induced senescence correlates well with a significant decrease in EF1A expression in A549 and H460 cells. These novel findings demonstrate, for the first time, that down-regulation of EF1A is involved in RV-induced premature senescence in lung cancer cells. Consistent with these observations, a recent study has suggested that decreased expression of EF1A is a potential biomarker of premature senescence [47]. However, further studies will be needed to define the exact role of EF1A in modulating RVinduced premature senescence in cancer cells. Many anticancer agents and ionizing radiation destroy tumor cells largely through the generation of ROS [48]. Moreover, increased ROS can CASIN trigger oxidative DNA damage and cause DNA DSBs, thus leading to premature senescence [37]. To determine the role of ROS in RV-induced premature senescence in lung cancer cells, we investigated the levels of ROS in RVtreated A549 and H460 cells using DCF-DA staining and flow cytometric assays. The data show that RV-induced senescence is associated with increased ROS production and DNA DSBs in lung cancer cells, suggesting that RV may induce premature senescence in lung cancer cells via ROS-mediated DNA damage. The important contribution of ROS to RV-induced DNA damage and premature senescence was further confirmed by the observations that inhibition of ROS production by NAC attenuates RVinduced DNA damage and senescence in NSCLC cells. Consistent with these observations, a pro-oxidant effect of RV was also observed in U937 leukemia cells and was characterized by the depletion of GSH and an increase in ROS production [49]. Moreover, previous studies by Hadi and coworkers also showed that RV could increase ROS generation and ROS-induced DNA damage in human peripheral lymphocytes [50,51]. Together, these findings demonstrate that low dose RV inhibits the growth of lung cancer cells via the induction of senescence through ROSmediated DNA damage. It is worth noting that there is evidence that RV can act as an ROS scavenger in normal cells to protect against ionizing radiation-induced oxidative stress and tissue injury [52], suggesting that RV may have differential effects on ROS production in normal versus cancer cells. Given that aberrant redox systems are frequently observed in many tumor cells [48,53,54], it is possible that RV may selectively suppress the growth of tumor cells with little or no toxicity to normal cells due to their differential redox status. In agreement wi.Been shown to be a critical mechanism underlying SIPS [12,15,17,18].Resveratrol-Induced Senescence in Cancer CellsFigure 6. RV induces Nox5 mRNA expression in LSCLC cells. (A) Cells were treated with 50 mM of RV or DMSO as vehicle control. Twenty-four hours after RV treatment, the expression levels of Nox1, Nox2, and Nox5 mRNAs were determined using real-time RT-PCR. (B) The expression levels of SOD1, SOD2 and TXN in A549 cells were determined by real-time RT-PCR. (C) The expression levels of Nox1, Nox2, and Nox5 in H460 cells are presented as fold change (mean 6 SEM). (D) The expression levels of SOD1, SOD2 and TXN in H460 cells are presented. **, p,0.001 vs. control. doi:10.1371/journal.pone.0060065.gHere we show that RV-induced premature senescence is associated with 23977191 increased expression of p53 and p21 in NSCLC cells, suggesting that activation of the p53 21 pathway may play an important role in modulating RV-induced senescence in lung cancer cells. More importantly, it was also found that RV-induced senescence correlates well with a significant decrease in EF1A expression in A549 and H460 cells. These novel findings demonstrate, for the first time, that down-regulation of EF1A is involved in RV-induced premature senescence in lung cancer cells. Consistent with these observations, a recent study has suggested that decreased expression of EF1A is a potential biomarker of premature senescence [47]. However, further studies will be needed to define the exact role of EF1A in modulating RVinduced premature senescence in cancer cells. Many anticancer agents and ionizing radiation destroy tumor cells largely through the generation of ROS [48]. Moreover, increased ROS can trigger oxidative DNA damage and cause DNA DSBs, thus leading to premature senescence [37]. To determine the role of ROS in RV-induced premature senescence in lung cancer cells, we investigated the levels of ROS in RVtreated A549 and H460 cells using DCF-DA staining and flow cytometric assays. The data show that RV-induced senescence is associated with increased ROS production and DNA DSBs in lung cancer cells, suggesting that RV may induce premature senescence in lung cancer cells via ROS-mediated DNA damage. The important contribution of ROS to RV-induced DNA damage and premature senescence was further confirmed by the observations that inhibition of ROS production by NAC attenuates RVinduced DNA damage and senescence in NSCLC cells. Consistent with these observations, a pro-oxidant effect of RV was also observed in U937 leukemia cells and was characterized by the depletion of GSH and an increase in ROS production [49]. Moreover, previous studies by Hadi and coworkers also showed that RV could increase ROS generation and ROS-induced DNA damage in human peripheral lymphocytes [50,51]. Together, these findings demonstrate that low dose RV inhibits the growth of lung cancer cells via the induction of senescence through ROSmediated DNA damage. It is worth noting that there is evidence that RV can act as an ROS scavenger in normal cells to protect against ionizing radiation-induced oxidative stress and tissue injury [52], suggesting that RV may have differential effects on ROS production in normal versus cancer cells. Given that aberrant redox systems are frequently observed in many tumor cells [48,53,54], it is possible that RV may selectively suppress the growth of tumor cells with little or no toxicity to normal cells due to their differential redox status. In agreement wi.

Share this post on:

Author: bet-bromodomain.