Figure 3 - available via license: CC BY-NC
Content may be subject to copyright.
Effects of rapamycin and YM155 on the OSCC-induced angiogenesis ability. (a) The CAM assays as well as Matrigel plug assay were performed to examine the effects of rapamycin and YM155 on the OSCC-induced angiogenesis ability in vivo. The eggs and the nude mice were divided into five groups as before: Control group, conditioned medium without YM155 and rapamycin group, conditioned medium with YM155 single group, conditioned medium with rapamycin single group, conditioned medium with rapamycin and YM155 group. A volume of 100 nM of rapamycin and 60 nM of YM155 was used. (b) All data are presented as mean ± SD from three different experiments performed in duplicate and then analyzed by Graphpad (**p < 0.01 vs the control group (no drug added); † † p < 0.01 the drug combination group vs the YM155 single group). 

Effects of rapamycin and YM155 on the OSCC-induced angiogenesis ability. (a) The CAM assays as well as Matrigel plug assay were performed to examine the effects of rapamycin and YM155 on the OSCC-induced angiogenesis ability in vivo. The eggs and the nude mice were divided into five groups as before: Control group, conditioned medium without YM155 and rapamycin group, conditioned medium with YM155 single group, conditioned medium with rapamycin single group, conditioned medium with rapamycin and YM155 group. A volume of 100 nM of rapamycin and 60 nM of YM155 was used. (b) All data are presented as mean ± SD from three different experiments performed in duplicate and then analyzed by Graphpad (**p < 0.01 vs the control group (no drug added); † † p < 0.01 the drug combination group vs the YM155 single group). 

Source publication
Article
Full-text available
YM155, a small molecule inhibitor of survivin, has been studied in many tumors. It has been shown that YM155 inhibited oral squamous cell carcinoma through promoting apoptosis and autophagy and inhibiting proliferation. It was found that YM155 also inhibited the oral squamous cell carcinoma–mediated angiogenesis through the inactivation of the mamm...

Contexts in source publication

Context 1
... OSCC-induced angiogenesis ability in vivo was examined by the chick chorioallantoic membrane (CAM) assay and Matrigel plug assay. Rapamycin in combination with YM155 reversed the effect of OSCC conditioned medium on new microvessel formation in the the CAM assay (Figure 3(a), p < 0.01). In the Matrigel plug assay, the hemoglobin contents in the plugs were much less in the drug combination group (Figure 3(b), p < 0.01). ...
Context 2
... in combination with YM155 reversed the effect of OSCC conditioned medium on new microvessel formation in the the CAM assay (Figure 3(a), p < 0.01). In the Matrigel plug assay, the hemoglobin contents in the plugs were much less in the drug combination group (Figure 3(b), p < 0.01). It revealed that rapamycin promoted the anti-angiogenesis ability of YM155 in OSCC. ...

Citations

... Rapamycin, a known inducer of autophagy, is shown in clinical studies to be successful in treating a small cohort of patients with refractory IBD. [40][41][42][43][44] Some studies investigated the use of rapamycin in patients with IBD and reported rapamycin to be an effective pharmacologic agent for patients with refractory IBD or IBD-related gastrointestinal strictures. 45,46 What is yet to be determined from these studies is the reason why certain patients benefit from rapamycin while others do not. ...
Article
Full-text available
Inflammatory bowel disease (IBD) represents a set of idiopathic and chronic inflammatory diseases of the gastrointestinal tract. Central to the pathogenesis of IBD is a dysregulation of normal intestinal epithelial homeostasis. cGAS is a DNA‐sensing receptor demonstrated to promote autophagy, a mechanism that removes dysfunctional cellular components. Beclin‐1 is a crucial protein involved in the initiation of autophagy. We hypothesized that cGAS plays a key role in intestinal homeostasis by upregulating Beclin‐1‐mediated autophagy. We evaluated intestinal cGAS levels in humans with IBD and in murine colonic tissue after performing a 2% dextran sulfate sodium (DSS) colitis model. Autophagy and cell death mechanisms were studied in cGAS KO and WT mice via qPCR, WB analysis, H&E, IF, and TUNEL staining. Autophagy was measured in stimulated intestinal epithelial cells (IECs) via WB analysis. Our data demonstrates cGAS to be upregulated during human and murine colitis. Furthermore, cGAS deficiency leads to worsened colitis and decreased levels of autophagy proteins including Beclin‐1 and LC3‐II. Co‐IP demonstrates a direct binding between cGAS and Beclin‐1 in IECs. Transfection of cGAS in stimulated HCT‐116 cells leads to increased autophagy. IECs isolated from cGAS KO have diminished autophagic flux. cGAS KO mice subjected to DSS have increased cell death and cleaved caspase‐3. Lastly, treatment of cGAS KO mice with rapamycin decreased the severity of colitis. Our data suggest that cGAS maintains intestinal epithelial homeostasis during human IBD and murine colitis by upregulating Beclin‐1‐mediated autophagy and preventing IEC death. Rescue of autophagy can attenuate the severity of colitis associated with cGAS deficiency.
... Rapamycin, which can be used as immunosuppressant, is a new type macrocyclic lactone, isolated from Streptomyces hygroscopicus. 40 Preceding investigations showed that rapamycin down-regulated MYCN protein expression, which can prohibit neuroblastoma cells proliferation, and this process was intimately correlated with the PI3K/Akt/mTOR pathway. [41][42][43] Rapamycin has been generally utilized as an anti-proliferative drug and immunosuppressant agent in the clinics. ...
Article
Full-text available
Background Alterations in the cell metabolism, such as enhanced aerobic glycolysis, have been identified as a prominent hallmark of cancer cells. 3-Bromopyruvate (3-BrPA) is a proverbial hexokinase (HK)-II inhibitor, which can inhibit cancer cell energy metabolism. Rapamycin is a new type macrocyclic lactone, which can inhibit the serine/threonine protein kinase mTOR. In order to comprehend the influence of 3-BrPA on autophagy activity in vitro, we conducted a series of experiments using different human neuroblastoma (NB) cell lines. Materials and Methods The human NB cell lines were exposed to 3-BrPA and/or rapamycin, and the proliferation activity of the cells was detected by Cell Counting Kit-8 (CCK-8) assay. The mRNA expression of the cells treated with 3-BrPA and/or rapamycin was analyzed by quantitative real-time polymerase chain reaction (QPCR) assay. The protein expression of the cells was analyzed by Western Blotting (WB) assay. The effects of 3-BrPA and/or rapamycin treatment on cell cycle and cell apoptosis were analyzed by flow cytometry assay. Meanwhile, the cellular glucose absorption rate, lactate secretion rate and ATP content were also analyzed through the relevant metabolic analysis kits. Results Our results showed that 3-BrPA can induce growth inhibition in a dose-dependent pattern by cell apoptosis. 3-BrPA combined with rapamycin played a synergistic suppression role in NB cells, affected the cell apoptosis, cell cycle and the metabolic pathway. Up-regulated LC3-II accumulation was conscious in NB cells incubated with 3-BrPA and rapamycin. Rapamycin individually discourages the mTOR signaling pathway, while combined with 3-BrPA can enhance this phenomenon and influence cell metabolism of the NB cells. Conclusion The results suggested that 3-BrPA combined with rapamycin could induce cell apoptosis in NB cells by inhibiting mTOR activity. In conclusion, our research proposed that the dual inhibitory effect of the mTOR signaling pathway and the glycolytic activity may indicate a valid therapeutic tactic for NB chemoprevention.
... Rapamycin is a macrocyclic lactone isolated from Streptomyces hygroscopicus [17] . ...
Article
Full-text available
Objective: To investigate the effect of Rapamycin on proliferation and autophagy in human neuroblastoma (NB) cell lines and to elucidate the possible mechanism. Methods: NB cells were treated with different concentrations of Rapamycin. Cell counting kit-8 (CCK-8) was used to measure proliferation, and flow cytometry (FCM) was used to analyze the cell cycle. Electron microscopy was used to observe cell morphological changes. Western blotting (WB) was performed to detect the expression of Beclin-1, LC3-I/II, P62, mammalian target of Rapamycin (mTOR) and p-mTOR. Results: Rapamycin inhibited the spread of NB cells in a dose- and time-dependent manner and arrested the cell cycle at the G0/G1 phase. Electron microscopy showed autophagosomes in NB cells treated with Rapamycin. The WB results showed that the expression levels of Beclin-1 and LC3-II/LC3-I were significantly elevated in NB cells treated with Rapamycin, while the expression levels of P62, mTOR and p-mTOR proteins were significantly reduced compared to the control cells (P<0.05). Conclusion: Rapamycin inhibits cell proliferation and induces autophagy in human NB cell lines. The mechanism may be related to suppression of the mTOR signaling pathway.
Article
Full-text available
Sepantronium bromide (YM-155) is believed to elicit apoptosis and mitotic arrest in tumor cells by reducing (BIRC5, survivin) mRNA. In this study, we monitored changes in survivin mRNA and protein after treating MDA-MB-231 cells with YM-155 concurrent with evaluation of whole transcriptomic (WT) mRNA and long intergenic non-coding RNA at 2 time points: 8 h sub-lethal (83 ng/mL) and 20 h at the LC50 (14.6 ng/mL). The data show a tight association between cell death and the precipitating loss of survivin protein and mRNA (-2.67 fold-change (FC), p<0.001) at 20 h, questioning if the decline in survivin is attributed to cell death or drug impact. The meager loss of survivin mRNA was overshadowed by enormous differential change to the WT in both magnitude and significance for over 2000 differentially up/down-regulated transcripts: (+22 FC to -12 FC, p<0.001). The data show YM-155 to up-regulate transcripts in control of circadian rhythm (NOCT, PER, BHLHe40, NFIL3), tumor suppression (SIK1, FOSB), histone methylation (KDM6B) and negative feedback of NF-kappa B signaling (TNFAIP3). Down-regulated transcripts by YM-155 include glucuronidase (GUSBP3), numerous micro-RNAs, DNA damage repair elements (CENPI, POLQ, RAD54B) and the most affected system was the ataxia-telangiectasia mutated (ATM)/Fanconi anemia E3 monoubiquitin ligase core complexes (FANC transcripts - A/B/E/F/G/M), FANC2, FANCI, BRCA1, BRCA2, RAD51, PALB2 gene and ATR (ATM- and Rad3-Related) pathway. In conclusion, these findings suggest that a primary target of YM-155 is the loss of replicative DNA repair systems.