Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
PLoS One ; 9(11): e112470, 2014.
Article in English | MEDLINE | ID: mdl-25383959

ABSTRACT

Celastrol, a tripterine derived from the traditional Chinese medicine plant Tripterygium wilfordii Hook F. ("Thunder of God Vine"), has been reported to have multiple effects, such as anti-inflammation, suppression of tumor angiogenesis, inhibition of tumor growth, induction of apoptosis and protection of cells against human neurodegenerative diseases. However, the mechanisms that underlie these functions are not well defined. In this study, we reported for the first time that Celastrol could induce HIF-1α protein accumulation in multiple cancer cell lines in an oxygen-independent manner and that the enhanced HIF-1α protein entered the nucleus and promoted the transcription of the HIF-1 target genes VEGF and Glut-1. Celastrol did not influence HIF-1α transcription. Instead, Celastrol induced the accumulation of the HIF-1α protein by inducing ROS and activating Akt/p70S6K signaling to promote HIF-1α translation. In addition, we found that the activation of Akt by Celastrol was transient. With increased exposure time, inhibition of Hsp90 chaperone function by Celastrol led to the subsequent depletion of the Akt protein and thus to the suppression of Akt activity. Moreover, in HepG2 cells, the accumulation of HIF-1α increased the expression of BNIP3, which induced autophagy. However, HIF-1α and BNIP3 did not influence the cytotoxicity of Celastrol because the main mechanism by which Celastrol kills cancer cells is through stimulating ROS-mediated JNK activation and inducing apoptosis. Furthermore, our data showed that the dose required for Celastrol to induce HIF-1α protein accumulation and enhance HIF-1α transcriptional activation was below its cytotoxic threshold. A cytotoxic dose of Celastrol for cancer cells did not display cytotoxicity in LO2 normal human liver cells, which indicated that the novel functions of Celastrol in regulating HIF-1 signaling and inducing autophagy might be used in new applications, such as in anti-inflammation and protection of cells against human neurodegenerative diseases. Future studies regarding these applications are required.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/metabolism , Signal Transduction/drug effects , Triterpenes/pharmacology , Cell Line , Cell Nucleus/metabolism , Glucose Transporter Type 1/genetics , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Neoplasms/genetics , Pentacyclic Triterpenes , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/genetics
2.
BMC Cancer ; 11: 170, 2011 May 14.
Article in English | MEDLINE | ID: mdl-21569548

ABSTRACT

BACKGROUND: Celastrol is an active ingredient of the traditional Chinese medicinal plant Tripterygium Wilfordii, which exhibits significant antitumor activity in different cancer models in vitro and in vivo; however, the lack of information on the target and mechanism of action of this compound have impeded its clinical application. In this study, we sought to determine the mode of action of celastrol by focusing on the processes that mediate its anticancer activity. METHODS: The downregulation of heat shock protein 90 (HSP90) client proteins, phosphorylation of c-Jun NH2-terminal kinase (JNK), and cleavage of PARP, caspase 9 and caspase 3 were detected by western blotting. The accumulation of reactive oxygen species (ROS) was analyzed by flow cytometry and fluorescence microscopy. Cell cycle progression, mitochondrial membrane potential (MMP) and apoptosis were determined by flow cytometry. Absorption spectroscopy was used to determine the activity of mitochondrial respiratory chain (MRC) complexes. RESULTS: Celastrol induced ROS accumulation, G2-M phase blockage, apoptosis and necrosis in H1299 and HepG2 cells in a dose-dependent manner. N-acetylcysteine (NAC), an antioxidative agent, inhibited celastrol-induced ROS accumulation and cytotoxicity. JNK phosphorylation induced by celastrol was suppressed by NAC and JNK inhibitor SP600125 (SP). Moreover, SP significantly inhibited celastrol-induced loss of MMP, cleavage of PARP, caspase 9 and caspase 3, mitochondrial translocation of Bad, cytoplasmic release of cytochrome c, and cell death. However, SP did not inhibit celastrol-induced ROS accumulation. Celastrol downregulated HSP90 client proteins but did not disrupt the interaction between HSP90 and cdc37. NAC completely inhibited celastrol-induced decrease of HSP90 client proteins, catalase and thioredoxin. The activity of MRC complex I was completely inhibited in H1299 cells treated with 6 µM celastrol in the absence and presence of NAC. Moreover, the inhibition of MRC complex I activity preceded ROS accumulation in H1299 cells after celastrol treatment. CONCLUSION: We identified ROS as the key intermediate for celastrol-induced cytotoxicity. JNK was activated by celastrol-induced ROS accumulation and then initiated mitochondrial-mediated apoptosis. Celastrol induced the downregulation of HSP90 client proteins through ROS accumulation and facilitated ROS accumulation by inhibiting MRC complex I activity. These results identify a novel target for celastrol-induced anticancer activity and define its mode of action.


Subject(s)
Antineoplastic Agents/pharmacology , Electron Transport Complex I/metabolism , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Triterpenes/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Electron Transport Complex I/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Hep G2 Cells , Humans , MAP Kinase Kinase 4/metabolism , Pentacyclic Triterpenes , Signal Transduction/drug effects
3.
Antimicrob Agents Chemother ; 47(11): 3485-93, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14576106

ABSTRACT

We show that an inducible rpoS antisense RNA complementary to the rpoS message can inhibit expression of RpoS in both exponential and stationary phases and can attenuate expression of the rpoS regulon in Escherichia coli. Plasmids containing rpoS antisense DNA expressed under the control of the T7lac promoter and T7 RNA polymerase were constructed, and expression of the rpoS antisense RNA was optimized in the pET expression system. rpoS antisense RNA levels could be manipulated to effectively control the expression of RpoS and RpoS-dependent genes. RpoS expression was inhibited by the expression of rpoS antisense RNA in both exponential and stationary phases in E. coli. RpoS-dependent catalase HPII was also downregulated, as determined by catalase activity assays and with native polyacrylamide gels stained for catalase. Induced RpoS antisense expression also reduced the level of RpoS-dependent glycogen synthesis. These results demonstrate that controlled expression of antisense RNA can be used to attenuate expression of a regulator required for the expression of host adaptation functions and may offer a basis for designing effective antimicrobial agents.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Escherichia coli/genetics , RNA, Antisense/biosynthesis , RNA, Antisense/genetics , Sigma Factor/antagonists & inhibitors , Sigma Factor/genetics , Bacterial Proteins/biosynthesis , Blotting, Northern , Blotting, Western , Catalase/metabolism , DNA Probes/chemical synthesis , DNA Probes/pharmacology , DNA, Bacterial/biosynthesis , DNA, Bacterial/genetics , DNA-Directed RNA Polymerases/genetics , Escherichia coli/drug effects , Escherichia coli/growth & development , Glycogen/metabolism , Plasmids/genetics , RNA, Bacterial/biosynthesis , RNA, Bacterial/genetics , Regulon , Reverse Transcriptase Polymerase Chain Reaction , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL