Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nutr Cancer ; 69(6): 920-931, 2017.
Article in English | MEDLINE | ID: mdl-28718669

ABSTRACT

Polyphenols represent a large group of natural substances with different biological properties. Currently, polyphenols are well studied due to their free radicals' scavenging and antioxidant activities. However, some studies indicate that polyphenols also exhibit pro-oxidant properties. In this study, the possible involvement of the pro-oxidant activities of fruit polyphenols was investigated in relation to apoptosis induction. To determine the type of cell death induced by fruit polyphenols (Flavine; F7), we assessed a series of assays, including measurements of caspase-7 activation, membrane mitochondrial potential changes, reactive oxygen (ROS) and nitrogen species production, lipid peroxidation, antioxidant enzymes activities, and PARP cleavage. Moreover, the effect of F7 on selected pro- and antisurvival signaling pathways was determined. We demonstrated that fruit polyphenols induced caspase-dependent cell death associated with increased oxidative stress. We also showed fruit polyphenol-mediated release of mitochondrial pro- and antiapoptotic proteins of the Bcl-2 family and modulation activity of the Akt, p38 MAPK, and Erk 1/2 pathways as well as the signaling of ROS-mediated DNA damage. Our data demonstrated that fruit peel polyphenols suppressed breast cancer cell growth through increased intracellular oxidative stress and the activation of p38 MAPK and de-activation of the Erk 1/2 and Akt signaling pathways.


Subject(s)
MAP Kinase Signaling System , Plant Extracts/pharmacology , Polyphenols/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Caspase 7/genetics , Caspase 7/metabolism , Cell Proliferation/drug effects , DNA Damage/drug effects , Fruit/chemistry , Humans , Lipid Peroxidation/drug effects , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics
2.
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
3.
Int J Mol Sci ; 12(12): 8388-405, 2011.
Article in English | MEDLINE | ID: mdl-22272079

ABSTRACT

Several photodynamically-active substances and farnesyltransferase inhibitors are currently being investigated as promising anticancer drugs. In this study, the combined effect of hypericin (the photodynamically-active pigment from Hypericum perforatum) and selective farnesyltransferase inhibitor manumycin (manumycin A; the selective farnesyltransferase inhibitor from Streptomyces parvulus) on HT-29 adenocarcinoma cells was examined. We found that the combination treatment of cells with photoactivated hypericin and manumycin resulted in enhanced antiproliferative and apoptotic response compared to the effect of single treatments. This was associated with increased suppression of clonogenic growth, S phase cell cycle arrest, elevated caspase-3/7 activity and time-dependent total cleavage of procaspase-3 and lamin B, cleavage of p21Bax into p18Bax and massive PARP cleavage. Moreover, we found that the apoptosis-inducing factor is implicated in signaling events triggered by photoactivated hypericin. Our results showed the relocalization of apoptosis-inducing factor (AIF) to the nuclei after hypericin treatment. In addition, we discovered that not only manumycin but also photoactivated hypericin induced the reduction of total Ras protein level. Manumycin decreased the amount of farnesylated Ras, and the combination treatment decreased the amount of both farnesylated and non-farnesylated Ras protein more dramatically. The present findings indicate that the inhibition of Ras processing may be the determining factor for enhancing the antiproliferative and apoptotic effects of combination treatment on HT-29 cells.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Perylene/analogs & derivatives , Polyenes/pharmacology , Polyunsaturated Alkamides/pharmacology , Radiation-Sensitizing Agents/pharmacology , Anthracenes , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/genetics , Caspase 7/metabolism , Enzyme Inhibitors/pharmacology , HT29 Cells , Humans , Lamin Type B/genetics , Lamin Type B/metabolism , Perylene/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , bcl-2-Associated X Protein/metabolism , ras Proteins/genetics , ras Proteins/metabolism
4.
Photochem Photobiol ; 86(6): 1285-93, 2010.
Article in English | MEDLINE | ID: mdl-20718930

ABSTRACT

Our recent study follows up an earlier one which demonstrated hypericin-mediated photocytotoxic effects on HT-29 adenocarcinoma cells by light fractionation with a longer dark pause between two unequal light doses (Sackova, A. [2005] Photochem. Photobiol.81, 1411-1416). Here, we present closer study on events invoked by sublethal light dose (1 J cm(-2)) during the period of 6 h that is sufficient to invoke resistance to second lethal dose (11 J cm(-2)). First, we proved that the dark pause of 6 h, but not 1 h, resulted in better cell survival with suppressed phosphatidylserine externalization, decreased reactive oxygen species production and hypericin content as well as altered expression of HSP70, GRP94, clusterin, nuclear factor (NF)-κB, IκB-α or Mcl-1. NF-κB activity assay confirmed activation of this early-response pathway. However, inhibition of IκB (IKK) kinase by parthenolide by stopping NF-κB release from the complex with IκB did not prevent onset of resistance, but it invoked some resistance even in groups with shorter, 1 h dark pause. Therefore, we predict involvement of another signaling pathway, located upstream from NF-κB, responsible for onset of resistance to photodynamic therapy with hypericin in colon adenocarcinoma cells HT-29.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , NF-kappa B/metabolism , Photochemotherapy , Anthracenes , Cell Survival/drug effects , Drug Resistance, Neoplasm , HT29 Cells , Humans , Light , Perylene/analogs & derivatives , Perylene/metabolism , Perylene/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Stem Cell Assay
5.
Photochem Photobiol Sci ; 8(11): 1558-67, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19862414

ABSTRACT

Photodynamic therapy with hypericin (HY-PDT) is known as an efficient modality for treatment of various cancerous and non-cancerous diseases. Although the role of p53 protein in cell death signaling is well established, relatively little is known of its impact on the efficiency of HY-PDT. Comparison of sensitivity and long-term survival of p53-null versus wt-p53-expressing HCT-116 cells is reported here. The lack of p53 function did not affect cell proliferation or attenuate the initial phases of programmed cell death. However, analyses of apoptosis in the final stages revealed suppression of its incidence and delayed activation of caspase-3 in p53-null cells. Significantly higher clonogenic ability, especially in hypoxia, was identified in the case of p53-null cells. Induction of Mcl-1 and Bax levels were more prominent in wt-pt53 cells. Interestingly, the level of Bcl-2 did not react to HY-PDT at all, in both cell lines. Bringing the evidence together, we prove that despite insignificant impact on overall toxicity, expression of p53 affects the clonogenic efficiency of HCT-116 cells. Since destruction of tumor tissue and its vascular system by PDT tends to lead to hypoxia, superior survival of p53-deficient tumor cells under given conditions might result in recurrence of cancer diseases.


Subject(s)
Colonic Neoplasms/pathology , Perylene/analogs & derivatives , Photochemotherapy , Tumor Suppressor Protein p53/metabolism , Anthracenes , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Colony-Forming Units Assay , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/radiation effects , Perylene/pharmacology , Perylene/therapeutic use , Phosphatidylserines/metabolism , Time Factors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
6.
Photochem Photobiol ; 81(6): 1411-6, 2005.
Article in English | MEDLINE | ID: mdl-16354114

ABSTRACT

The present study demonstrates the in vitro effect of hypericin-mediated PDT with fractionated light delivery. Cells were photosensitized with unequal light fractions separated by dark intervals (1 or 6 h). We compared the changes in viability, cell number, survival, apoptosis and cell cycle on HT-29 cells irradiated with a single light dose (12 J/cm(2)) to the fractionated light delivery (1 + 11 J/cm(2)) 24 and 48 h after photodynamic treatment. We found that a fractionated light regime with a longer dark period resulted in a decrease of hypericin cytotoxicity. Both cell number and survival were higher after light sensitization with a 6-h dark interval. DNA fragmentation occurred after a single light-dose application, but in contrast no apoptotic DNA formation was detected with a 6-h dark pause. After fractionation the percentage of cells in the G1 phase of the cell cycle was increased, while the proportion of cells in the G2 phase decreased as compared to a single light-dose application, i.e. both percentage of cells in the G1 and G2 phase of the cell cycle were near control levels. We presume that the longer dark interval after the irradiation of cells by first light dose makes them resistant to the effect of the second illumination. These findings confirm that the light application scheme together with other photodynamic protocol components is crucial for the photocytotoxicity of hypericin.


Subject(s)
Adenocarcinoma/drug therapy , Cell Proliferation/drug effects , Dose Fractionation, Radiation , Light , Perylene/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Anthracenes , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Drug Screening Assays, Antitumor , HT29 Cells , Humans , Perylene/pharmacology , Photochemotherapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...