Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Radiat Oncol Biol Phys ; 89(5): 1106-1114, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25035215

ABSTRACT

PURPOSE: To test the hypothesis that small molecule targeting of nucleophosmin 1 (NPM1) represents a rational approach for radiosensitization. METHODS AND MATERIALS: Wilde-type and NPM1-deficient mouse embryo fibroblasts (MEFs) were used to determine whether radiosensitization produced by the small molecule YTR107 was NPM1 dependent. The stress response to ionizing radiation was assessed by quantifying pNPM1, γH2AX, and Rad51 foci, neutral comet tail moment, and colony formation. NPM1 levels in a human-derived non-small-cell lung cancer (NSCLC) tissue microarray (TMA) were determined by immunohistochemistry. YTR107-mediated radiosensitization was assessed in NSCLC cell lines and xenografts. RESULTS: Use of NPM1-null MEFs demonstrated that NPM1 is critical for DNA double- strand break (DSB) repair, that loss of NPM1 increases radiation sensitivity, and that YTR107-mediated radiosensitization is NPM1 dependent. YTR107 was shown to inhibit NPM1 oligomerization and impair formation of pNPM1 irradiation-induced foci that colocalized with γH2AX foci. Analysis of the TMA demonstrated that NPM1 is overexpressed in subsets of NSCLC. YTR107 inhibited DNA DSB repair and radiosensitized NSCLC lines and xenografts. CONCLUSIONS: These data demonstrate that YTR107-mediated targeting of NPM1 impairs DNA DSB repair, an event that increases radiation sensitivity.


Subject(s)
Barbiturates/pharmacology , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Indoles/pharmacology , Molecular Targeted Therapy/methods , Neoplasm Proteins/drug effects , Nuclear Proteins/drug effects , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/radiotherapy , Fibroblasts/chemistry , Fibroblasts/radiation effects , Histones/analysis , Humans , Lung Neoplasms/chemistry , Lung Neoplasms/radiotherapy , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Proteins/analysis , Neoplasm Proteins/deficiency , Neoplasm Proteins/physiology , Nuclear Proteins/analysis , Nuclear Proteins/deficiency , Nuclear Proteins/physiology , Nucleophosmin , Rad51 Recombinase/analysis , Tissue Array Analysis , Tumor Cells, Cultured
2.
Invest New Drugs ; 31(3): 535-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23054211

ABSTRACT

Targeting tumor vasculature represents a rational strategy for controlling cancer. (Z)-(+/-)-2-(1-benzylindol-3-ylmethylene)-1-azabicyclo[2.2.2]octan-3-ol (denoted VJ115) is a novel chemical entity that inhibits the enzyme ENOX1, a NADH oxidase. Genetic and small molecule inhibition of ENOX1 inhibits endothelial cell tubule formation and tumor-mediated neo-angiogenesis. Inhibition of ENOX1 radiosensitizes tumor vasculature, a consequence of enhanced apoptosis. However, the molecular mechanisms underlying these observations are not well understood. Herein, we mechanistically link ENOX1-mediated regulation of cellular NADH concentrations with proteomics profiling of endothelial cell protein expression following exposure to VJ115. Pathway Studios network analysis of potential effector molecules identified by the proteomics profiling indicated that a VJ115 exposure capable of altering intracellular NADH concentrations impacted proteins involved in cytoskeletal reorganization. The analysis was validated using RT-PCR and immunoblotting of selected proteins. RNAi knockdown of ENOX1 was shown to suppress expression of stathmin and lamin A/C, proteins identified by the proteomics analysis to be suppressed upon VJ115 exposure. These data support the hypothesis that VJ115 inhibition of ENOX1 can impact expression of proteins involved in cytoskeletal reorganization and support a hypothesis in which ENOX1 activity links elevated cellular NADH concentrations with cytoskeletal reorganization and angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cytoskeletal Proteins/metabolism , Indoles/pharmacology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Quinuclidines/pharmacology , Cells, Cultured , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , NAD/metabolism , Proteomics
3.
Clin Cancer Res ; 17(20): 6490-9, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21878537

ABSTRACT

PURPOSE: Radiation therapy continues to be an important therapeutic strategy for providing definitive local/regional control of human cancer. However, oncogenes that harbor driver mutations and/or amplifications can compromise therapeutic efficacy. Thus, there is a need for novel approaches that enhance the DNA damage produced by ionizing radiation. EXPERIMENTAL DESIGN: A forward chemical genetic approach coupled with cell-based phenotypic screening of several tumor cell lines was used to identify a novel chemical entity (NCE) that functioned as a radiation sensitizer. Proteomics, comet assays, confocal microscopy, and immunoblotting were used to identify the biological target. RESULTS: The screening process identified a 5-((N-benzyl-1H-indol-3-yl)-methylene)pyrimidine-2,4,6(1H,3H,5H)trione as an NCE that radiosensitized cancer cells expressing amplified and/or mutated RAS, ErbB, PIK3CA, and/or BRAF oncogenes. Affinity-based solid-phase resin capture followed by liquid chromatography/tandem mass spectrometry identified the chaperone nucleophosmin (NPM) as the NCE target. SiRNA suppression of NPM abrogated radiosensitization by the NCE. Confocal microscopy showed that the NCE inhibited NPM shuttling to radiation-induced DNA damage repair foci, and the analysis of comet assays indicated a diminished rate of DNA double-strand break repair. CONCLUSION: These data support the hypothesis that inhibition of DNA repair due to inhibition of NPM shuttling increases the efficacy of DNA-damaging therapeutic strategies.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Neoplasms/drug therapy , Nuclear Proteins/metabolism , Radiation-Sensitizing Agents/pharmacology , Animals , Barbiturates/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , Humans , Indoles/pharmacology , Mice , Mice, Nude , Molecular Chaperones/metabolism , Molecular Targeted Therapy , Neoplasms/metabolism , Nucleophosmin , Radiation Tolerance/drug effects , Random Allocation
4.
FASEB J ; 23(9): 2986-95, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19395476

ABSTRACT

There is a need for novel strategies that target tumor vasculature, specifically those that synergize with cytotoxic therapy, in order to overcome resistance that can develop with current therapeutics. A chemistry-driven drug discovery screen was employed to identify novel compounds that inhibit endothelial cell tubule formation. Cell-based phenotypic screening revealed that noncytotoxic concentrations of (Z)-(+/-)-2-(1-benzenesulfonylindol-3-ylmethylene)-1-azabicyclo[2. 2.2]octan-3-ol (analog I) and (Z)-(+/-)-2-(1-benzylindol-3-ylmethylene)-1-azabicyclo[2.2.2]octan-3-ol (analog II) inhibited endothelial cell migration and the ability to form capillary-like structures in Matrigel by > or =70%. The ability to undergo neoangiogenesis, as measured in a window-chamber model, was also inhibited by 70%. Screening of biochemical pathways revealed that analog II inhibited the enzyme ENOX1 (EC(50) = 10 microM). Retroviral-mediated shRNA suppression of endothelial ENOX1 expression inhibited cell migration and tubule formation, recapitulating the effects observed with the small-molecule analogs. Genetic or chemical suppression of ENOX1 significantly increased radiation-mediated Caspase3-activated apoptosis, coincident with suppression of p70S6K1 phosphorylation. Administration of analog II prior to fractionated X-irradiation significantly diminished the number and density of tumor microvessels, as well as delayed syngeneic and xenograft tumor growth compared to results obtained with radiation alone. Analysis of necropsies suggests that the analog was well tolerated. These results suggest that targeting ENOX1 activity represents a novel therapeutic strategy for enhancing the radiation response of tumors.


Subject(s)
Endothelium, Vascular/cytology , Neovascularization, Pathologic/drug therapy , Protein Disulfide Reductase (Glutathione)/antagonists & inhibitors , Quinuclidines/pharmacology , Transcription Factors/antagonists & inhibitors , Cell Movement/drug effects , Cell Shape/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Indoles , Membrane Proteins/antagonists & inhibitors , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic/radiotherapy , Quinuclidines/therapeutic use
5.
Cancer Res ; 68(2): 364-8, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18199529

ABSTRACT

The transcription factor nuclear factor erythroid-derived 2-related factor 2 (Nrf2) regulates induction of an extensive cellular stress response network when complexed with the cAMP-responsive element binding protein (CBP) at antioxidant response elements (ARE) located in the promoter region of target genes. Activating transcription factor 3 (ATF3) can repress Nrf2-mediated signaling in a manner that is not well understood. Here, we show that ATF3-mediated suppression is a consequence of direct ATF3-Nrf2 protein-protein interactions that result in displacement of CBP from the ARE. This work establishes ATF3 as a novel repressor of the Nrf2-directed stress response pathway.


Subject(s)
Activating Transcription Factor 3/physiology , Gene Expression Regulation , NF-E2-Related Factor 2/physiology , Stress, Physiological/genetics , Activating Transcription Factor 3/antagonists & inhibitors , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Antioxidants/pharmacology , Cells, Cultured , Mice , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Protein Binding , RNA, Small Interfering/pharmacology , Repressor Proteins/metabolism , Repressor Proteins/physiology , Response Elements/drug effects , Signal Transduction/genetics , Transfection
6.
Bioorg Med Chem Lett ; 17(24): 6821-4, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17980582

ABSTRACT

Use of ionizing radiation is essential for the management of many human cancers, and therapeutic hyperthermia has been identified as a potent radiosensitizer. Radiation therapy combined with adjuvant hyperthermia represents a potential tool to provide outstanding local-regional control for refractory disease. (Z)-(+/-)-2-(N-Benzylindol-3-ylmethylene)quinuclidin-3-ol (2) and (Z)-(+/-)-2-(N-benzenesulfonylindol-3-ylmethylene)quinuclidin-3-ol (4) were initially identified as potent thermal sensitizers that could lower the threshold needed for thermal sensitivity to radiation treatment. To define the structural requirements of the molecule that are essential for thermal sensitization, we have synthesized and evaluated a series of (Z)-2-(N-benzylindol-3-ylmethylene)quinuclidin-3-one (9), and (Z)-(+/-)-2-(N-benzylindol-3-ylmethylene)quinuclidin-3-ol (10) analogs that incorporate a variety of substituents in both the indole and N-benzyl moieties. These systematic structure-activity relationship (SAR) studies were designed to further the development and optimization of potential clinically useful thermal sensitizing agents. The most potent analog was compound 10 (R(1)=H, R(2)=4-Cl), which potently inhibited (93% inhibition at 50 microM) the growth of HT-29 cells after a 41 degrees C/2h exposure.


Subject(s)
Quinuclidines/chemical synthesis , Quinuclidines/pharmacology , Temperature , Cell Line, Tumor , Cell Survival/drug effects , Humans , Molecular Structure , Quinuclidines/chemistry , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Structure-Activity Relationship
7.
Chem Res Toxicol ; 20(10): 1528-35, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17854155

ABSTRACT

15-deoxy-Delta12,14-prostaglandin J2 (15-d-PGJ2) is a dehydration product of PGD2. This compound possesses a highly reactive polyunsaturated carbonyl moiety that is a substrate for Michael addition with thiol-containing biomolecules such as glutathione and cysteine residues on proteins. By reacting with glutathione and proteins, 15-d-PGJ2 is believed to exert potent biological activity. Despite the large number of publications that have ascribed bioactivity to this molecule, it is not known to what extent 15-d-PGJ2 is formed in vivo. Levels of free 15-d-PGJ2 measured in human biological fluids such as urine are low, and the biological importance of this compound has thus been questioned. Because of its reactivity, we hypothesized that 15-d-PGJ2 is present in vivo primarily as a Michael conjugate. Therefore, we undertook a detailed study of the metabolism of this compound in HepG2 cells that are known to metabolize other cyclopentenone eicosanoids. We report that HepG2 cells primarily convert 15-d-PGJ2 to a glutathione conjugate in which the carbonyl at C-11 is reduced to a hydroxyl. Subsequently, the glutathione portion of the molecule is hydrolyzed with loss of glutamic acid and glycine resulting in a cysteine conjugate. These findings confirm a general route for the metabolism of cyclopentenone eicosanoids in HepG2 cells and may pave the way for new insights regarding the formation of 15-d-PGJ2 in vivo.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glutathione/metabolism , Hepatocytes/metabolism , Metabolic Detoxication, Phase II , Prostaglandin D2/analogs & derivatives , Cell Line, Tumor , Chromatography, High Pressure Liquid , Glutathione/analysis , Hepatocytes/drug effects , Humans , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , Spectrometry, Mass, Electrospray Ionization
8.
Cancer Res ; 67(2): 695-701, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17234780

ABSTRACT

Radiation therapy combined with adjuvant hyperthermia has the potential to provide outstanding local-regional control for refractory disease. However, achieving therapeutic thermal dose can be problematic. In the current investigation, we used a chemistry-driven approach with the goal of designing and synthesizing novel small molecules that could function as thermal radiosensitizers. (Z)-(+/-)-2-(1-Benzenesulfonylindol-3-ylmethylene)-1-azabicyclo[2.2.2]octan-3-ol was identified as a compound that could lower the threshold for Hsf1 activation and thermal sensitivity. Enhanced thermal sensitivity was associated with significant thermal radiosensitization. We established the structural requirements for activity: the presence of an N-benzenesulfonylindole or N-benzylindole moiety linked at the indolic 3-position to a 2-(1-azabicyclo[2.2.2]octan-3-ol) or 2-(1-azabicyclo[2.2.2]octan-3-one) moiety. These small molecules functioned by exploiting the underlying biophysical events responsible for thermal sensitization. Thermal radiosensitization was characterized biochemically and found to include loss of mitochondrial membrane potential, followed by mitotic catastrophe. These studies identified a novel series of small molecules that represent a promising tool for the treatment of recurrent tumors by ionizing radiation.


Subject(s)
Colonic Neoplasms/therapy , Hyperthermia, Induced/methods , Indoles/chemistry , Indoles/pharmacology , Mitosis/physiology , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/radiotherapy , DNA-Binding Proteins/metabolism , HCT116 Cells , Heat Shock Transcription Factors , Humans , Indoles/chemical synthesis , Mitosis/drug effects , Protein Conformation/drug effects , Radiation-Sensitizing Agents/chemical synthesis , Structure-Activity Relationship , Transcription Factors/metabolism
9.
J Biol Chem ; 282(4): 2529-37, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17127771

ABSTRACT

Consumption of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) can mitigate the progression of diseases in which oxidative stress represents a common underlying biochemical process. Nrf2-regulated gene expression regulates detoxification of reactive oxygen species. EPA and DHA were subjected to an in vitro free radical oxidation process that models in vivo conditions. Oxidized n-3 fatty acids reacted directly with the negative regulator of Nrf2, Keap1, initiating Keap1 dissociation with Cullin3, thereby inducing Nrf2-directed gene expression. Liquid chromatography-tandem mass spectrometry analyses of oxidized EPA demonstrated the presence of novel cyclopentenone-containing molecules termed J3-isoprostanes in vitro and in vivo and were shown to induce Nrf2-directed gene expression. These experiments provide a biochemical basis for the hypothesis that formation of J-ring compounds generated from oxidation of EPA and DHA in vivo can reach concentrations high enough to induce Nrf2-based cellular defense systems.


Subject(s)
Cell Cycle Proteins/metabolism , Cullin Proteins/metabolism , Fatty Acids, Omega-3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Cell Cycle Proteins/chemistry , Cell Line , Cullin Proteins/chemistry , Fatty Acids, Omega-3/chemistry , Gene Expression Regulation , Genes, Reporter , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Kelch-Like ECH-Associated Protein 1 , Mice , Molecular Structure , Oxidation-Reduction , Oxidative Stress/genetics , Transcriptional Activation
10.
Chem Res Toxicol ; 17(1): 17-25, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14727915

ABSTRACT

Cyclopentenone isoprostanes (IsoPs), A(2)/J(2)-IsoPs, are one class of IsoPs formed via the free radical-initiated peroxidation of arachidonic acid. These compounds, which are structurally similar to cyclooxygenase-derived PGA(2) and PGJ(2), contain highly reactive alpha,beta-unsaturated carbonyl moieties. A(2)/J(2)-IsoPs are generated in vivo in humans esterified in glycerophospholipids. Unlike other classes of IsoPs, however, cyclopentenone IsoPs cannot be detected in the free form; we postulated that this might be due to their rapid adduction to various thiol-containing biomolecules via Michael addition. Recently, we reported that the A-ring IsoP, 15-A(2t)-IsoP, is efficiently conjugated with glutathione in vitro by certain human and rat glutathione transferases (GSTs), with the isozyme GSTA4-4 displaying the highest activity. Herein, we examined the metabolic disposition of 15-A(2t)-IsoP in HepG2 cells. We report that 15-A(2t)-IsoP is primarily metabolized by these cells via conjugation to glutathione. Within 6 h, approximately 60% of 15-A(2t)-IsoP added to HepG2 cells was present in the form of a water soluble conjugate(s). Structural characterization of the adduct(s) by liquid chromatography-tandem mass spectrometry revealed four major conjugates. These include the intact 15-A(2t)-IsoP-GSH conjugate, the GSH conjugate in which the carbonyl at C-9 of 15-A(2t)-IsoP is reduced, and the corresponding cysteine conjugates. These studies thus show that the primary pathway of metabolic disposition of endogenously derived cyclopentenone IsoPs occurs via conjugation with thiols.


Subject(s)
Cyclopentanes/metabolism , Glutathione/metabolism , Lipid Peroxidation , Prostaglandins A/metabolism , Cysteine/chemistry , Cysteine/metabolism , Glutathione/chemistry , Humans , Prostaglandins A/chemistry , Prostaglandins A/pharmacology , Spectrometry, Mass, Electrospray Ionization , Time Factors , Tritium , Tumor Cells, Cultured
11.
Cancer Res ; 63(17): 5636-45, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500406

ABSTRACT

Glutamate cysteine ligase, the rate-limiting enzyme for the synthesis of glutathione, represents an important component of chemoprevention paradigms. GCLC and GCLM, the genes encoding glutamate cysteine ligase subunits, are induced by indoles, such as indomethacin. Novel functionalized indole analogues and other structurally related compounds were synthesized and used for a comparative structure analysis of GCLC induction. Use of mouse embryo fibroblasts null for Nrf2 (nuclear factor-erythroid 2p45-related transcription factor) and HepG2 cells overexpressing Keap1 demonstrated that indole analogue-mediated GCLC expression was regulated by Nrf2-Keap1 interactions. Indole analogues capable of inducing GCLC were found to increase NADPH oxidase activity. Indole analogues unable to induce GCLC did not increase oxidase activity. HepG2 cells transfected with FLAG/Keap1 were exposed to indomethacin, and the redox state of Keap1 cysteine residues was assessed. The data indicated that Keap1 exhibited several oxidation states that were sensitive to indomethacin treatment. These indomethacin-mediated changes in thiol oxidation states were suppressed by diphenyleneiodonium, a NADPH oxidase inhibitor. Diphenyleneiodonium also suppressed indole analogue-mediated increases in GCLC mRNA. In summary, the use of the indole analogues identified NADPH oxidase activity as a novel upstream activity regulating Nrf2/Keap1 signaling of GCLC, provided data supporting the hypothesis that Keap1 is a downstream effector for oxidase activity, and afforded in vivo data to support the hypothesis that Keap1 thiols can act as molecular sensors of reactive oxygen species. Finally, the comparative structure analysis suggests that 2-indol-3-yl-methylenequinuclidin-3-ols may represent a prototype for the development of novel chemopreventative agents able to activate Keap1/Nrf2 signaling.


Subject(s)
Anticarcinogenic Agents/pharmacology , Glutamate-Cysteine Ligase/genetics , Indoles/pharmacology , NADPH Oxidases/metabolism , Proteins/physiology , Quinuclidines/pharmacology , Animals , Gene Expression Regulation, Enzymologic/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Humans , Intracellular Signaling Peptides and Proteins , Kelch-Like ECH-Associated Protein 1 , Mice , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...