Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
J Cancer ; 6(6): 538-41, 2015.
Article in English | MEDLINE | ID: mdl-26000045

ABSTRACT

ARF, NPM and FOXM1 proteins interact with each other in mammalian cells. We showed previously that proteasome inhibitors suppress not only FOXM1 expression, but also the expression of ARF and NPM proteins. Using RNA interference we found that the depletion of each of these proteins by RNAi in human cancer HeLa cells leads to down-regulation of the two other partners, suggesting that these proteins stabilize each other in human cancer cells. Since the suppression of FOXM1 is one of hallmarks of proteasome inhibition, suppression of ARF and NPM by proteasome inhibitors may be explained in part as a secondary effect of downregulation of FOXM1 that modulate stability of ARF and NPM1 proteins.

2.
Cell Cycle ; 13(20): 3202-6, 2014.
Article in English | MEDLINE | ID: mdl-25485499

ABSTRACT

Tumor suppressor p53 is one of the most frequently mutated genes in cancer, with almost 50% of all types of cancer expressing a mutant form of p53. p53 transactivates the expression of its primary negative regulator, HDM2. HDM2 is a ubiquitin ligase, which initiates the proteasomal degradation of p53 following ubiquitination. Proteasome inhibitors, by targeting the ubiquitin proteasome pathway inhibit the degradation of the majority of cellular proteins including wild-type p53. In contrast, in this study we found that the protein expression of mutant p53 was suppressed following treatment with established or novel proteasome inhibitors. Furthermore, for the first time we demonstrated that Arsenic trioxide, which was previously shown to suppress mutant p53 protein level, exhibits proteasome inhibitory activity. Proteasome inhibitor-mediated suppression of mutant p53 was partially rescued by the knockdown of HDM2, suggesting that the stabilization of HDM2 by proteasome inhibitors might be responsible for mutant p53 suppression to some extent. This study suggests that suppression of mutant p53 is a general property of proteasome inhibitors and it provides additional rationale to use proteasome inhibitors for the treatment of tumors with mutant p53.


Subject(s)
Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Arsenic Trioxide , Arsenicals/pharmacology , Cell Line, Tumor , Humans , Oxides/pharmacology , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Tumor Suppressor Protein p53/genetics
3.
Bioorg Med Chem Lett ; 23(24): 6902-4, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24169233

ABSTRACT

Anti-microtubule agents such as paclitaxel and docetaxel have played an important role in the treatment of cancer for many years. Recently, a small molecule that has a taxol-like mode of action (5HPP-33) was reported. Herein, the detailed structure-activity relationship (SAR) studies of 5HPP-33 analogs that are substituted at the isoindole and phenyl rings are described. Bulky substitutions (such as di-isopropyl groups) on the phenyl ring result in the isoindole and phenyl rings being perpendicular to each other. It was found that this conformation is critical for anti-microtubule activity. These studies have provided valuable information, which will be helpful in the design of more potent analogs.


Subject(s)
Isoindoles/chemistry , Microtubules/chemistry , Paclitaxel/chemistry , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Isoindoles/chemical synthesis , Isoindoles/pharmacology , Microtubules/metabolism , Structure-Activity Relationship , Thalidomide/chemical synthesis , Tubulin Modulators/chemical synthesis
4.
Bioorg Med Chem Lett ; 23(15): 4465-8, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23790539

ABSTRACT

A series of compounds originally derived from the vascular endothelial growth factor receptor tyrosine kinase inhibitor, SU5416, were synthesized and evaluated. The most potent compound in this series, compound 3, which structurally resembles the potent anti-microtubule agent combretastatin A-4, inhibited tubulin polymerization and showed potent growth inhibitory activities on both prostate and breast cancer lines with IC50 values in the low nanomolar range.


Subject(s)
Antineoplastic Agents/chemical synthesis , Drug Design , Tubulin Modulators/chemical synthesis , Tubulin/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Bibenzyls/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Polymerization/drug effects , Tubulin/chemistry , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
5.
Am J Pathol ; 183(1): 257-65, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23665201

ABSTRACT

Tumor cells accumulate high level of reactive oxygen species (ROS) because they are metabolically more active than normal cells. Elevated ROS levels increase tumorigenecity but also render cancer cells more vulnerable to oxidative stress than normal cells. The oncogenic transcription factor Forkhead Box M1 (FOXM1), which is overexpressed in a wide range of human cancers, was reported to protect cancer cells from the adverse effects of oxidative stress by up regulating the expression of scavenger enzymes. We therefore hypothesized that the combination of FOXM1 ablation and ROS inducers could selectively eradicate cancer cells. We show that RNA interference-mediated knockdown of FOXM1 further elevates intracellular ROS levels and increases sensitivity of cancer cells to ROS-mediated cell death after treatment with ROS inducers. We also demonstrate that the combination of ROS inducers with FOXM1/proteasome inhibitors induces robust apoptosis in different human cancer cells. In addition, we show evidence that FOXM1/proteasome inhibitor bortezomib in combination with the ROS inducer ß-phenylethyl isothiocyanate efficiently inhibits the growth of breast tumor xenografts in nude mice. We conclude that the combination of ROS inducers and FOXM1 inhibitors could be used as a therapeutic strategy to selectively eliminate cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Biomarkers, Tumor/antagonists & inhibitors , Forkhead Transcription Factors/antagonists & inhibitors , Mammary Neoplasms, Experimental/drug therapy , Oxidative Stress/drug effects , 2-Methoxyestradiol , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Drug Administration Schedule , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Isothiocyanates/administration & dosage , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Pyrazines/administration & dosage , RNA Interference , Random Allocation , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , Treatment Outcome
6.
Cell Cycle ; 10(22): 3827-9, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22071628

ABSTRACT

Proteasome inhibitors stabilize numerous proteins by inhibiting their degradation. Previously we have demonstrated that proteasome inhibitors thiostrepton, MG132 and bortezomib paradoxically inhibit transcriptional activity and mRNA/protein expression of FOXM1. Here we demonstrate that, in addition to FOXM1, the same proteasome inhibitors also decrease mRNA and protein expression of NPM and ARF genes. These data suggest that proteasome inhibitors may suppress gene expression by stabilizing their transcriptional inhibitors.


Subject(s)
ADP-Ribosylation Factors/genetics , Cysteine Proteinase Inhibitors/pharmacology , Gene Expression/drug effects , Nuclear Proteins/genetics , Proteasome Inhibitors , Boronic Acids/pharmacology , Bortezomib , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , HeLa Cells , Humans , Leupeptins/pharmacology , Nucleophosmin , Pyrazines/pharmacology , RNA, Messenger/metabolism , Thiostrepton/pharmacology
7.
Cell Cycle ; 10(19): 3269-73, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21941087

ABSTRACT

Apoptosis has been widely accepted as the primary mechanism of drug-induced cell death. Recently, a second type of cell death pathway has been demonstrated: autophagy, also called programmed type II cell death. Autophagy is a highly regulated process, by which selected components of a cell are degraded. It primarily functions as a cell survival mechanism under stress. However, persistent stress can also promote extensive autophagy leading to cell death. Forkhead box M1 (FoxM1), an oncogenic transcription factor that is abundantly expressed in a wide range of human cancers. Here we evaluated the role of FoxM1 in sensitivity of human cancer cells to proteasome inhibitor-induced apoptosis and autophagy. We found that FoxM1 knockdown sensitized the human cancer cells to apoptotic cell death induced by proteasome inhibitors, such as, MG132, bortezomib and thiostrepton, while it did not affect the levels of autophagy following treatment with these drugs.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Proteasome Inhibitors , Apoptosis/drug effects , Autophagy/drug effects , Boronic Acids/pharmacology , Bortezomib , Caspase 3/metabolism , Cell Line, Tumor , Forkhead Box Protein M1 , Forkhead Transcription Factors/antagonists & inhibitors , Humans , Leupeptins/pharmacology , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Thiostrepton/pharmacology
8.
PLoS One ; 6(2): e17110, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21365012

ABSTRACT

Thiazole antibiotic, thiostrepton was recently identified as proteasome inhibitor. We investigated the therapeutic potential of the combination of thiostrepton and proteasome inhibitor bortezomib (Velcade) on various human tumor cell lines. Combination of sub-lethal concentrations of thiostrepton and bortezomib induced potent apoptosis and inhibition of long-term colony formation in a wide variety of human cancer cell lines. The synergistic relationship between thiostrepton and bortezomib combination was also quantitatively demonstrated by calculating their combination index values that were much lower than 1 in all studied cell lines. The synergy between these drugs was based on their proteasome inhibitory activities, because thiostrepton modification, thiostrepton methyl ester, which did not have intact quinaldic acid ring and did not inhibit proteasome activity failed to demonstrate any synergy in combination with bortezomib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Boronic Acids/administration & dosage , Neoplasms/drug therapy , Pyrazines/administration & dosage , Thiostrepton/pharmacology , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Bortezomib , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Synergism , HCT116 Cells , Humans , Neoplasms/pathology , Thiazoles/administration & dosage , Thiazoles/pharmacology , Thiostrepton/administration & dosage
9.
Am J Pathol ; 178(1): 355-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224072

ABSTRACT

Proteasome inhibitors are used against human cancer, but their mechanisms of action are not entirely understood. For example, the role of the tumor suppressor p53 is controversial. We reevaluated the role of p53 in proteasome inhibitor-induced apoptosis by using isogenic human cancer cell lines with different p53 status. We found that well-known proteasome inhibitors such as MG132 and bortezomib, as well as the recently discovered proteasome inhibitor thiostrepton, induced p53-independent apoptosis in human cancer cell lines that correlated with p53-independent induction of proapoptotic Noxa but not Puma protein. In addition, these drugs inhibited growth of several cancer cell lines independently of p53 status. Notably, thiostrepton induced more potent apoptosis in HepG2 cells with p53 knockdown than in parental cells with wild-type p53. Our data confirm that proteasome inhibitors generally induce p53-independent apoptosis in human cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Cysteine Proteinase Inhibitors/pharmacology , Neoplasms/enzymology , Proteasome Inhibitors , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Thiostrepton/pharmacology , Tumor Suppressor Protein p53/genetics
10.
Cancer Biol Ther ; 11(1): 43-7, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21119308

ABSTRACT

Thiopeptides are sulfur containing highly modified macrocyclic antibiotics with a central pyridine/tetrapyridine/dehydropiperidine ring with up to three thiazole substituents on positions 2, 3 and 6. Thiazole antibiotics with central pyridine nucleus have a macrocyclic loop connecting thiazole rings at position 2 and 3 described as ring A. In addition antibiotics with central tetrahydropyridine nucleus have a quinaldic acid macrocycle also connected to thiazole on position 2 described as ring B. We have demonstrated before that thiazole antibiotics thiostrepton and Siomycin A act as proteasome inhibitors in mammalian tumor cells. Here we decided to test whether other known thiazole antibiotics such as berninamycin, micrococcin P1 and P2, thiocillin and YM-266183 (lacking the quinaldic acid ring B) demonstrate this activity. We found that none of them act as proteasome inhibitors. Moreover, structural modification of thiostrepton to thiostrepton methyl ester (with open B ring) also did not demonstrate this activity. These data suggest that B ring of thiostrepton and Siomycin A that is absent in other thiazole antibiotics determines the proteasome inhibitory activity of these drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Proteasome Inhibitors , Thiazoles/pharmacology , Anti-Bacterial Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cysteine Proteinase Inhibitors/chemistry , Drug Screening Assays, Antitumor , Humans , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Peptides/chemistry , Peptides/pharmacology , Thiazoles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL