Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 34(1): 495-503, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26063116

ABSTRACT

Indole-3-carbinol (I3C) and diindolylmethane (DIM), found in cruciferous vegetables, have chemopreventive and anticancer properties. In the present study, 14 substituted indoles were tested for activity against SW480 colon cancer cells. Among these, 3-(2-bromoethyl)-indole, named BEI-9, showed the greatest inhibition. The effects of BEI-9 on cancer cells were analyzed by MTS and CellTiter-Glo assays for effects on cell viability, by microscopy for phenotypic changes, by scratch wound assays for effects on migration, by flow cytometry for changes in the cell cycle, by immunoblotting for cyclin D and A to assess effects on cell cycle regulation, and by NF-κB reporter assays for effects on basal and drug-induced NF-κB activation. BEI-9 inhibited the growth of SW480 and HCT116 colon cancer cells at concentrations of 12.5 and 5 µM, respectively. BEI-9 also inhibited cell motility as determined with scratch wound assays, and reduced the levels of cyclin D1 and A. Furthermore, in reporter cells, BEI-9 (0.8 µM) inhibited basal and induced NF-κB activation and increased cell death when combined with the cytokine TNFα or the drug camptothecin (CPT), both of which activate NF-κB. Preliminary experiments to identify a safe dose range for immunodeficient mice showed that BEI-9, administered intraperitoneally, was tolerable at doses below 10 mg/kg. Thus, BEI-9 and other indole derivatives may be useful in chemoprevention or as chemosensitizers. Since NF-κB activation is implicated in carcinogenesis and in reducing sensitivity to anticancer drugs, BEI-9 should be investigated in combination with drugs such as CPT, which activate NF-κB.


Subject(s)
Antineoplastic Agents/administration & dosage , Colonic Neoplasms/drug therapy , Indoles/administration & dosage , NF-kappa B/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Hep G2 Cells , Humans , Indoles/pharmacology , Injections, Intraperitoneal , Mice , Xenograft Model Antitumor Assays
2.
BMC Cancer ; 14: 599, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25134433

ABSTRACT

BACKGROUND: The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway is activated in cells exposed to various stimuli, including those originating on the cell surface or in the nucleus. Activated NF-κB signaling is thought to enhance cell survival in response to these stimuli, which include chemotherapy and radiation. In the present effort, we determined which anticancer drugs preferentially activate NF-κB in colon cancer cells. METHODS: NF-κB reporter cells were established and treated with 5-fluorouracil (5-FU, DNA/RNA damaging), oxaliplatin (DNA damaging), camptothecin (CTP, topoisomerase inhibitor), phleomycin (radiomimetic), or erlotinib (EGFR inhibitor). The activation of NF-κB was assessed by immunofluorescence for p65 translocation, luciferase assays, and downstream targets of NF-κB activation (cIAP2, and Bcl-XL) were evaluated by immunoblotting, by ELISA (CXCL8 and IL-6 in culture supernatants), or by gene expression analysis. RESULTS: Colon cancer cells responded variably to different classes of therapeutic agents, and these agents initiated variable responses among different cell types. CPT activated NF-κB in SW480 colon cancer cells in a dose-dependent manner, but not in HCT116 cells that were either wild-type or deficient for p53. In SW480 colon cancer cells, NF-κB activation by CPT was accompanied by secretion of the cytokine CXCL8, but not by up-regulation of the anti-apoptotic genes, cIAP2 or Bcl-XL. On the contrary, treatment of HCT116 cells with CPT resulted in up-regulation of CXCR2, a receptor for CXCL8, without an increase in cytokine levels. In SW480 cells, NF-κB reporter activity, but not cytokine secretion, was inhibited by SM-7368, an NF-κB inhibitor. CONCLUSION: The results show that, in response to cancer therapeutic agents, NF-κB activation varies with the cellular make up and that drug-induced NF-κB activation may be functionally uncoupled from anti-apoptotic outcomes found for other stimuli. Some cancer cells in a heterogeneous tumor tissue may, under therapeutic pressure, release soluble factors that have paracrine activity on neighboring cells that express the cognate receptors.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , NF-kappa B/metabolism , Signal Transduction/drug effects , Antineoplastic Agents/therapeutic use , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/metabolism , Erlotinib Hydrochloride , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Phleomycins/pharmacology , Phleomycins/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use
3.
Anticancer Res ; 32(1): 61-71, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22213289

ABSTRACT

BACKGROUND: DNA-damaging drugs constitute standard chemotherapy regimen for advanced colorectal cancer. Here, the interactions between quercetin and 5-fluorouracil (5-FU), etoposide, and camptothecin were examined in cancer cells. MATERIALS AND METHODS: HCT116 colorectal or PPC1 prostate cancer cells were treated with quercetin and the drugs. Clonogenicity assays, cell cycle profiles, and expressions of p53, p21, BAX, survivin and cyclin B1 proteins were used to examine the effects of the treatments. RESULTS: Quercetin synergistically inhibited the clonogenicity of the wild-type cells, but inhibited the cell cycle effects of all the drugs tested. In p53-null cells, the combination of low dose 5-FU with up to 6 µM quercetin promoted clonogenic survival. Treatment of p53-wild-type cells with 50 µM quercetin reduced drug-induced up-regulation of p53, p21 and BAX. The combination of quercetin and the drugs also reduced the levels of cyclin B1 and survivin proteins. CONCLUSION: While high doses of quercetin synergize with DNA-damaging agents, the effect of drug combination with quercetin is influenced by the effective doses and the p53 status of the cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antioxidants/pharmacology , Colorectal Neoplasms/drug therapy , DNA Damage/drug effects , Prostatic Neoplasms/drug therapy , Quercetin/pharmacology , Apoptosis/drug effects , Blotting, Western , Camptothecin/administration & dosage , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Synergism , Etoposide/administration & dosage , Flow Cytometry , Fluorouracil/administration & dosage , Humans , Inhibitor of Apoptosis Proteins/metabolism , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Survivin , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
4.
Anticancer Res ; 31(1): 233-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21273604

ABSTRACT

BACKGROUND: Ruta graveolens is a medicinal herb that has been used for centuries against various ailments. This study examined the anticancer properties of the herb using cancer cell lines. MATERIALS AND METHODS: Methanolic extract of R. graveolens was tested on colon, breast and prostate cancer cells. Viability, cell cycle profiles, clonogenicity and capase activation were measured. Induction and subcellular localizations of p53, 53BP1 and γ-H2AX proteins were examined. RESULTS: The extract dose-dependently decreased the viability and the clonogenicity of treated cells and induced G2/M arrest, aberrant mitoses, and caspase-3 activation. It also induced the p53 pathway and focal concentration of the DNA damage response proteins 53BP1 and γ-H2AX. Moreover, the levels of phospho-Akt and cyclin B1 were reduced by treatment, whereas only cyclin B1 was reduced in normal dermal fibroblasts. CONCLUSION: R. graveolens extract contains bioactive compounds which, independently of known photoactivatable mechanisms, potently inhibit cancer cell proliferation and survival through multiple targets.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , DNA Damage/drug effects , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Ruta/chemistry , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA, Neoplasm/drug effects , Enzyme Activation/drug effects , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Plant Extracts/chemistry , Plants, Medicinal/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Tumor Stem Cell Assay
5.
Nutr Cancer ; 62(8): 1025-35, 2010.
Article in English | MEDLINE | ID: mdl-21058190

ABSTRACT

Quercetin is a flavonoid with anticancer properties. In this study, we examined the effects of quercetin on cell cycle, viability, and proliferation of cancer cells, either singly or in combination with the microtubule-targeting drugs taxol and nocodazole. Although quercetin induced cell death in a dose-dependent manner, 12.5-50 µM quercetin inhibited the activity of both taxol and nocodazole to induce G2/M arrest in various cell lines. Quercetin also partially restored drug-induced loss in viability of treated cells for up to 72 h. This antagonism of microtubule-targeting drugs was accompanied by a delay in cell cycle progression and inhibition of the buildup of cyclin-B1 at the microtubule organizing center of treated cells. However, quercetin did not inhibit the microtubule targeting of taxol or nocodazole. Despite the short-term protection of cells by quercetin, colony formation and clonogenicity of HCT116 cells were still suppressed by quercetin or quercetin-taxol combination. The status of cell adherence to growth matrix was critical in determining the sensitivity of HCT116 cells to quercetin. We conclude that although long-term exposure of cancer cells to quercetin may prevent cell proliferation and survival, the interference of quercetin with cell cycle progression diminishes the efficacy of microtubule-targeting drugs to arrest cells at G2/M.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Cell Cycle/drug effects , Neoplasms/drug therapy , Nocodazole/antagonists & inhibitors , Paclitaxel/antagonists & inhibitors , Quercetin/metabolism , Tubulin Modulators/antagonists & inhibitors , Cell Adhesion , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin B1/metabolism , Food-Drug Interactions , G2 Phase/drug effects , Humans , Microtubules/drug effects , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Nocodazole/pharmacology , Osmolar Concentration , Paclitaxel/pharmacology , Time Factors , Tubulin/metabolism , Tubulin Modulators/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...