Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Molecules ; 21(12)2016 Nov 26.
Article in English | MEDLINE | ID: mdl-27898039

ABSTRACT

Indole phytoalexins from crucifers have been shown to exhibit significant anti-cancer, chemopreventive, and antiproliferative activity. Phytoalexins are natural low molecular antimicrobial compounds that are synthesized and accumulated in plants after their exposure to pathogenic microorganisms. Most interestingly, crucifers appear to be the only plant family producing sulfur-containing indole phytoalexins. The mechanisms underlying its anti-cancer properties are unknown. Isolation from cruciferous plants does not provide sufficient quantities of indole phytoalexins and, for biological screening, they are usually obtainable through synthesis. Understanding the molecular mechanism of the action of these substances and their structure-activity relationships is quite important in the development of new analogs with a more favorable profile of biological activities. In this review, we present the key features of indole phytoalexins, mainly their antiproliferative ativities.


Subject(s)
Brassicaceae/chemistry , Cell Proliferation/drug effects , Indoles/pharmacology , Sesquiterpenes/pharmacology , Humans , Indoles/chemistry , Molecular Structure , Sesquiterpenes/chemistry , Tumor Cells, Cultured , Phytoalexins
2.
Tumour Biol ; 35(10): 9967-75, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25008568

ABSTRACT

Colorectal cancer is the third most common cancer in the world, with 1.2 million new cancer cases annually. Chalcones are secondary metabolite precursors of flavonoids that exhibit diverse biological activities, including antioxidant and antitumor activities. The aim of this study was to investigate the antiproliferative effect of new synthetic chalcone derivatives on HCT116 cells. (E)-2-(2',4'-dimethoxybenzylidene)-1-tetralone (Q705) was found to be the most active (IC50 = 3.44 ± 0.25 µM). Based on these results, this compound was chosen for further analysis of its biochemical and molecular mechanisms. Our results showed that Q705 inhibited the growth and clonogenicity of HCT116 cells. The results of a flow cytometric analyses suggested that this compound caused a significant cell cycle arrest in G2/M phase and increased the proportion of cells in the subG0/G1 phase, marker of apoptosis. Q705-induced apoptosis was confirmed by TdT-mediated dUTP nick end labelling (TUNEL) assay. Treatment of HCT116 cells with this chalcone significantly increased the caspase-3,-7 activity and resulted in cleavage of poly-ADP-ribose polymerase (PARP). Changes in the nuclear morphology such as chromatin condensation were also observed. These effects were associated with a decreased expression of bcl-xL and increased overall ratio of bax/bcl-xL mRNA levels. Immunofluorescence and qRT-PCR analysis revealed that Q705 induced H2AX histone modifications characteristic of DNA damage, disruption of microtubule organization and downregulation of tubulins. In summary, these results suggest that the cyclic chalcone analogue Q705 has potential as a new compound for colorectal cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzylidene Compounds/pharmacology , Cell Cycle Checkpoints/drug effects , Chalcones/pharmacology , Colorectal Neoplasms/pathology , Blotting, Western , Cell Proliferation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , HCT116 Cells , Humans , In Situ Nick-End Labeling , Real-Time Polymerase Chain Reaction
3.
Molecules ; 19(8): 10877-97, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-25068784

ABSTRACT

This study was designed to examine the in vitro antiproliferative effect of brassinin and its derivatives on human cancer cell lines. Among seven tested compounds, homobrassinin (K1; N-[2-(indol-3-yl)ethyl]-S-methyldithiocarbamate) exhibited the most potent activity with IC50 = 8.0 µM in human colorectal Caco2 cells and was selected for further studies. The flow cytometric analysis revealed a K1-induced increase in the G2/M phase associated with dysregulation of α-tubulin, α1-tubulin and ß5-tubulin expression. These findings suggest that the inhibitory effect of K1 can be mediated via inhibition of microtubule formation. Furthermore, simultaneously with G2/M arrest, K1 also increased population of cells with sub-G1 DNA content which is considered to be a marker of apoptotic cell death. Apoptosis was also confirmed by annexin V/PI double staining, DNA fragmentation assay and chromatin condensation assay. The apoptosis was associated with the loss of mitochondrial membrane potential (MMP), caspase-3 activation as well as intracellular reactive oxygen species (ROS) production. Moreover, the antioxidant Trolox blocked ROS production, changes in MMP and decreased K1 cytotoxicity, which confirmed the important role of ROS in cell apoptosis. Taken together, our data demonstrate that K1 induces ROS-dependent apoptosis in Caco2 cells and provide the rationale for further in vivo anticancer investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Reactive Oxygen Species/metabolism , Thiocarbamates/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caco-2 Cells , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/chemistry , Inhibitory Concentration 50 , Mitochondria/drug effects , Mitochondria/metabolism , Thiocarbamates/chemistry , Tubulin/genetics , Tubulin/metabolism
4.
Nutrition ; 31(4): 560-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25770318

ABSTRACT

OBJECTIVES: There has been considerable interest in both clinical and preclinical research about the role of phytochemicals in the reduction of risk for cancer in humans. The aim of this study was to determine the antineoplastic effects of Chlorella pyrenoidosa in experimental breast cancer in vivo and in vitro. METHODS: In this experiment, the antineoplastic effects of C. pyrenoidosa in the chemoprevention of N-methyl-N-nitrosourea-induced mammary carcinogenesis in female rats were evaluated. Chlorella powder was administered through diet at concentrations of 0.3% and 3%. The experiment was terminated 14 wk after carcinogen administration. At autopsy, mammary tumors were removed and prepared for histopathological and immunohistochemical analysis. In vitro cytotoxicity assay, parameters of apoptosis, and proliferation after chlorella treatment in human breast adenocarcinoma (MCF-7) cells were carried out. RESULTS: Basic parameters of experimental carcinogenesis, mechanism of action (biomarkers of apoptosis, proliferation, and angiogenesis), chosen metabolic variables, and side effects after long-term chlorella treatment in animals were assessed. Chlorella at higher concentration suppressed tumor frequency by 61% (P < 0.02) and lengthened tumor latency by 12.5 d (P < 0.02) in comparison with the controls. Immunohistochemical analysis of rat tumor cells showed caspase-7 expression increase by 73.5% (P < 0.001) and vascular endothelial growth factor receptor-2 expression decrease by 19% (P = 0.07) after chlorella treatment. In a parallel in vitro study, chlorella significantly decreased survival of MCF-7 cells in a dose-dependent manner. In chlorella-treated MCF-7 cells, a significant increase in cells having sub-G0/G1 DNA content and significant increase of early apoptotic and late apoptotic/necrotic cells after annexin V/PI staining assay were found. Decreases in mitochondrial membrane potential and increasing reactive oxygen species generation were observed in the chlorella-treated MCF-7 cells. CONCLUSIONS: This study is the first report on the antineoplastic effects of C. pyrenoidosa in experimental breast cancer in vivo and in vitro.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Chlorella , Phytotherapy , Animals , Annexin A5/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Caspase 7/metabolism , Cell Proliferation , Diet , Female , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Microalgae , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Cell Cycle ; 13(14): 2296-305, 2014.
Article in English | MEDLINE | ID: mdl-24911186

ABSTRACT

Allosteric targeting of protein kinases via displacement of the structural αC helix with type III allosteric inhibitors is currently gaining a foothold in drug discovery. Recently, the first crystal structure of CDK2 with an open allosteric pocket adjacent to the αC helix has been described, prospecting new opportunities to design more selective inhibitors, but the structure has not yet been exploited for the structure-based design of type III allosteric inhibitors. In this work we report the results of a virtual screening campaign that resulted in the discovery of the first-in-class type III allosteric ligands of CDK2. Using a combination of docking and post-docking analyses made with our tool BEAR, 7 allosteric ligands (hit rate of 20%) with micromolar affinity for CDK2 were identified, some of them inhibiting the growth of breast cancer cell lines in the micromolar range. Competition experiments performed in the presence of the ATP-competitive inhibitor staurosporine confirmed that the 7 ligands are truly allosteric, in agreement with their design. Of these, compound 2 bound CDK2 with an EC50 value of 3 µM and inhibited the proliferation of MDA-MB231 and ZR-75-1 breast cancer cells with IC50 values of approximately 20 µM, while compound 4 had an EC50 value of 71 µM and IC50 values around 4 µM. Remarkably, the most potent compound 4 was able to selectively inhibit CDK2-mediated Retinoblastoma phosphorylation, confirming that its mechanism of action is fully compatible with a selective inhibition of CDK2 phosphorylation in cells. Finally, hit expansion through analog search of the most potent inhibitor 4 revealed an additional ligand 4g with similar in vitro potency on breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Computer-Aided Design , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Drug Discovery/methods , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Allosteric Regulation , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Ligands , Molecular Targeted Therapy , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Structure, Secondary , Retinoblastoma Protein/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
6.
Toxicol In Vitro ; 28(5): 909-15, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24747292

ABSTRACT

The aim of the study was to investigate the anti-proliferative activity of brassinin and its derivatives on human cancer cell lines. We found that among twenty-one tested compounds, 1- methoxybrassinin exerted the most potent anti-proliferative activity in Caco-2 cells with IC50 8.2 (±1.2)µmoll(-1). The flow cytometric analysis revealed a 1-methoxybrassinin-induced increase in the sub-G1 DNA content fraction which is considered to be a marker of apoptotic cell death. Apoptosis was also confirmed by DNA fragmentation assay. Moreover, quantitative real-time PCR showed that 1-methoxybrassinin upregulated the expression of pro-apoptotic Bax and downregulated the expression of anti-apoptotic genes Bcl-2 and Bcl-xL. The compound also increased activity of caspase-3, -7, cleaved PARP and decreased intracellular GSH content. The present study has assessed the in vitro anti-proliferative potential of 1-methoxybrassinin. The results generate a rationale for in vivo efficacy studies with this compound in preclinical cancer models.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Thiocarbamates/pharmacology , Caco-2 Cells , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Glutathione/metabolism , HCT116 Cells , Hep G2 Cells , Humans , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL