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










Database
Language
Publication year range
1.
Mol Cancer Ther ; 15(7): 1495-503, 2016 07.
Article in English | MEDLINE | ID: mdl-27197304

ABSTRACT

Multidrug resistance (MDR) is a major cause of tumor treatment failure; therefore, drugs that can avoid this outcome are urgently needed. We studied triptolide, which directly kills MDR tumor cells with a high potency and a broad spectrum of cell death. Triptolide did not inhibit P-glycoprotein (P-gp) drug efflux and reduced P-gp and MDR1 mRNA resulting from transcription inhibition. Transcription factors including c-MYC, SOX-2, OCT-4, and NANOG were not correlated with triptolide-induced cell killing, but RPB1, the largest subunit of RNA polymerase II, was critical in mediating triptolide's inhibition of MDR cells. Triptolide elicited antitumor and anti-MDR activity through a universal mechanism: by activating CDK7 by phosphorylating Thr170 in both parental and MDR cell lines and in SK-OV-3 cells. The CDK7-selective inhibitor BS-181 partially rescued cell killing induced by 72-hour treatment of triptolide, which may be due to partial rescue of RPB1 degradation. We suggest that a precise phosphorylation site on RPB1 (Ser1878) was phosphorylated by CDK7 in response to triptolide. In addition, XPB and p44, two transcription factor TFIIH subunits, did not contribute to triptolide-driven RPB1 degradation and cell killing, although XPB was reported to covalently bind to triptolide. Several clinical trials are underway to test triptolide and its analogues for treating cancer and other diseases, so our data may help expand potential clinical uses of triptolide, as well as offer a compound that overcomes tumor MDR. Future investigations into the primary molecular target(s) of triptolide responsible for RPB1 degradation may suggest novel anti-MDR target(s) for therapeutic development. Mol Cancer Ther; 15(7); 1495-503. ©2016 AACR.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cyclin-Dependent Kinases/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Diterpenes/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Phenanthrenes/pharmacology , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Epoxy Compounds/pharmacology , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phosphorylation , Proteolysis , Signal Transduction/drug effects , Tumor Stem Cell Assay , Cyclin-Dependent Kinase-Activating Kinase
2.
Oncotarget ; 6(11): 8960-73, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25840421

ABSTRACT

Both microtubule and topoisomerase II (Top2) are important anticancer targets and their respective inhibitors are widely used in combination for cancer therapy. However, some combinations could be mutually antagonistic and drug resistance further limits their therapeutic efficacy. Here we report YCH337, a novel α-carboline derivative that targets both microtubule and Top2, eliciting tumor proliferation and growth inhibition and overcoming drug resistance. YCH337 inhibited microtubule polymerization by binding to the colchicine site and subsequently led to mitotic arrest. It also suppressed Top2 and caused DNA double-strand breaks. It disrupted microtubule more potently than Top2. YCH337 induced reversible mitotic arrest at low concentrations but persistent DNA damage. YCH337 caused intrinsic and extrinsic apoptosis and decreased MCL-1, cIAP1 and XIAP proteins. In this aspect, YCH337 behaved differently from the combination of vincristine and etoposide. YCH337 inhibited proliferation of tumor cells with an averaged IC50 of 0.3 µM. It significantly suppressed the growth of HT-29 xenografts in nude mice too. Importantly, YCH337 nearly equally killed different-mechanism-mediated resistant tumor cells and corresponding parent cells. Together with the novelty of its chemical structure, YCH337 could serve as a promising lead for drug development and a prototype for a dual microtubule/Top2 targeting strategy for cancer therapy.


Subject(s)
Carbolines/therapeutic use , DNA Breaks, Double-Stranded/drug effects , DNA, Neoplasm/drug effects , Neoplasm Proteins/antagonists & inhibitors , Topoisomerase II Inhibitors/therapeutic use , Tubulin Modulators/therapeutic use , Animals , Apoptosis/drug effects , Binding Sites/drug effects , Binding, Competitive , Carbolines/pharmacology , Cell Line, Tumor , Colchicine/metabolism , Colonic Neoplasms/drug therapy , DNA Topoisomerases, Type II/physiology , DNA, Superhelical/drug effects , Drug Interactions , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Humans , Inhibitory Concentration 50 , Metaphase/drug effects , Mice , Mice, Nude , Microtubules/drug effects , Microtubules/metabolism , Molecular Structure , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/physiology , Paclitaxel/pharmacology , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/pharmacology , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/pharmacology , Vincristine/pharmacology
3.
Mol Cancer Ther ; 13(6): 1480-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24688049

ABSTRACT

Colchicine site-targeted tubulin inhibitors are a promising type of anticancer drugs. MT189 is a new derivative of MT119, a previously reported colchicine site-binding antitubulin agent. In this study, MT189 was demonstrated to retain the property of MT119 in disrupting microtubulin via binding to the colchicine site, causing mitotic arrest and inducing apoptosis, and to display 8.7-fold enhanced proliferative inhibition in a panel of cancer cells. MT189 was shown to elicit in vivo anticancer effects on MDA-MB-231 xenografts in nude mice, and the tumor growth was suppressed by 35.9% over 14 days. MT189 led to degradation of MCL-1, a member of the antiapoptotic BCL-2 protein family. Its overexpression reduced but its silenced expression increased the apoptotic induction followed by the treatment with MT189. Moreover, the treatment with MT189 caused activation of the MEKK1/TAK1-MKK4-JNK signaling pathway. The activated JNK resulted in phosphorylation of MCL-1, which facilitated its ubiquitination-mediated degradation. Our results show that MT189 inhibits microtubulin polymerization by binding to the colchicine site. Relief of apoptotic suppression by MCL-1 degradation together with mitotic arrest contributes to the anticancer activity of MT189.


Subject(s)
Antineoplastic Agents/administration & dosage , Imidazoles/administration & dosage , MAP Kinase Kinase 4/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/drug therapy , Pyridines/administration & dosage , Tubulin Modulators/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , HT29 Cells , Humans , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Neoplasms/metabolism , Phosphorylation , Proteolysis/drug effects , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...