Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Enzyme Inhib Med Chem ; 34(1): 877-897, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30938202

ABSTRACT

A combination of biochemical, biophysical and biological techniques was used to study calf thymus DNA interaction with newly synthesized 7-MEOTA-tacrine thiourea 12-17 and urea heterodimers 18-22, and to measure interference with type I and II topoisomerases. Their biological profile was also inspected in vitro on the HL-60 cell line using different flow cytometric techniques (cell cycle distribution, detection of mitochondrial membrane potential dissipation, and analysis of metabolic activity/viability). The compounds exhibited a profound inhibitory effect on topoisomerase activity (e.g. compound 22 inhibited type I topoisomerase at 1 µM concentration). The treatment of HL-60 cells with the studied compounds showed inhibition of cell growth especially with hybrids containing thiourea (14-17) and urea moieties (21 and 22). Moreover, treatment of human dermal fibroblasts with the studied compounds did not indicate significant cytotoxicity. The observed results suggest beneficial selectivity of the heterodimers as potential drugs to target cancer cells.


Subject(s)
Acridines/pharmacology , Antineoplastic Agents/pharmacology , Tacrine/pharmacology , Thiourea/pharmacology , A549 Cells , Acridines/chemical synthesis , Acridines/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fibroblasts/drug effects , HL-60 Cells , Humans , Structure-Activity Relationship , Tacrine/chemistry , Thiourea/chemistry
2.
Clin Epigenetics ; 9: 62, 2017.
Article in English | MEDLINE | ID: mdl-28603560

ABSTRACT

BACKGROUND: Hypericin-mediated photodynamic therapy (HY-PDT) has recently captured increased attention as an alternative minimally invasive anticancer treatment, although cancer cells may acquire resistance. Therefore, combination treatments may be necessary to enhance HY-PDT efficacy. Histone deacetylase inhibitors (HDACis) are often used in combination treatments due to their non-genotoxic properties and epigenetic potential to sensitize cells to external stimuli. Therefore, this study attempts for the first time to investigate the therapeutic effects of HDACis in combination with visible light-mediated PDT against cancer. Specifically, the colorectal cancer cell model was used due to its known resistance to HY-PDT. RESULTS: Two chemical groups of HDACis were tested in combination with HY-PDT: the hydroxamic acids Saha and Trichostatin A, and the short-chain fatty acids valproic acid and sodium phenylbutyrate (NaPB), as inhibitors of all-class versus nuclear HDACs, respectively. The selected HDACis manifest a favorable clinical toxicity profile and showed similar potencies and mechanisms in intragroup comparisons but different biological effects in intergroup analyses. HDACi combination with HY-PDT significantly attenuated cancer cell resistance to treatment and caused the two HDACi groups to become similarly potent. However, the short-chain fatty acids, in combination with HY-PDT, showed increased selectivity towards inhibition of HDACs versus other key epigenetic enzymes, and NaPB induced the strongest expression of the otherwise silenced tumor suppressor CDKN1A, a hallmark gene for HDACi-mediated chromatin modulation. Epigenetic regulation of CDKN1A by NaPB was associated with histone acetylation at enhancer and promoter elements rather than histone or DNA methylation at those or other regulatory regions of this gene. Moreover, NaPB, compared to the other HDACis, caused milder effects on global histone acetylation, suggesting a more specific effect on CDKN1A chromatin architecture relative to global chromatin structure. The mechanism of NaPB + HY-PDT was P53-dependent and likely driven by the HY-PDT rather than the NaPB constituent. CONCLUSIONS: Our results show that HDACis potentiate the antitumor efficacy of HY-PDT in colorectal cancer cells, overcoming their resistance to this drug and epigenetically reactivating the expression of CDKN1A. Besides their therapeutic potential, hypericin and these HDACis are non-genotoxic constituents of dietary agents, hence, represent interesting targets for investigating mechanisms of dietary-based cancer prevention.


Subject(s)
Antineoplastic Agents/pharmacology , Chromatin/drug effects , Colonic Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Photochemotherapy/methods , Anthracenes , Antineoplastic Agents/chemistry , Cell Line, Tumor , Chromatin/genetics , Colonic Neoplasms/drug therapy , DNA Methylation/drug effects , Drug Synergism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/chemistry , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Perylene/analogs & derivatives , Perylene/pharmacology , Phenylbutyrates/chemistry , Phenylbutyrates/pharmacology , Valproic Acid/chemistry , Valproic Acid/pharmacology
3.
Toxicol Lett ; 243: 56-66, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26721606

ABSTRACT

Proadifen (SKF-525A) is a P450 monooxygenase inhibitor with potential anti-proliferative activity and the ability to potentiate the toxicity of hypericin-mediated photodynamic therapy and mitoxantrone via alteration of ABC transport proteins. Elevated expression of some ABC transporters may also determine the efficacy of cisplatin-based chemotherapy. Thus, the purpose of this study was to investigate the ability of proadifen to sensitize A2780 and A2780cis ovarian cancer cells to cisplatin (CDDP). Herein, we show for the first time that proadifen sensitized resistant ovarian cancer cells to CDDP-induced cell death. The chemosensitizing effect of proadifen on CDDP action was also confirmed by MTT assays in multicellular spheroids. The possible mechanisms responsible for the enhanced cytotoxicity of proadifen/CDDP combined treatment may be attributed to a decrease of reduced relative glutathione levels, downregulation of multidrug resistance-associated proteins 1 and 2 (MRP1, MRP2) and attenuation of survivin expression. Taken together, our results indicate that proadifen is a promising compound for further in vivo experiments related to overcoming multidrug resistance and sensitization of resistant ovarian carcinoma to CDDP.


Subject(s)
Cisplatin/pharmacology , Proadifen/pharmacology , Apoptosis/drug effects , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Down-Regulation , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mitochondrial Membranes/metabolism , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Survivin , Up-Regulation , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
4.
Int J Oncol ; 47(4): 1572-84, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26252082

ABSTRACT

Multidrug resistance caused by the overexpression of ABC transporter proteins in cancer cells remains a major obstacle limiting chemotherapy efficacy. Drugs inhibiting these transporters have been shown to increase the anti-proliferative properties of chemotherapeutics. As we previously described, proadifen, a P450 monooxygenase inhibitor, might also be able to inhibit some ABC transporters, including breast cancer resistance protein (BCRP). Because mitoxantrone (MTX) is a strong BCRP substrate and is often used in the treatment of leukemia, we investigated the effect of 24 h proadifen pre-treatment on the cytotoxicity of MTX in leukemic cell lines that are sensitive to MTX (HL-60) and MTX-resistant ABCG2-overexpressing subclone (cBCRP). We show for the first time that proadifen is able to enhance the cytotoxic properties of MTX in cBCRP cells, particularly through the inhibition of BCRP expression and activity. This proadifen-MTX synergism was also mediated by the inhibition of various cellular proteins engaged in apoptosis, including Mc-1, Bcl-xL, survivin and activation of procaspase-3. Proadifen also decreased the expression of γH2AX, which is involved in the recruitment of reparation proteins. Moreover, the inhibition of DNA damage repair proteins Ku86 and B23 after proadifen treatment indicate a possible role of proadifen in DNA repair blockage, thus suppressing the reparation rate of MTX-induced DSBs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Promyelocytic, Acute/pathology , Mitoxantrone/pharmacology , Proadifen/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Drug Synergism , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Membrane Potential, Mitochondrial/drug effects , Reverse Transcriptase Polymerase Chain Reaction
5.
Eur J Pharm Sci ; 76: 192-202, 2015 Aug 30.
Article in English | MEDLINE | ID: mdl-25960253

ABSTRACT

HL-60 cancer cells were treated with a series of novel acridine derivatives (derivatives 1-4) in order to test the compounds' ability to inhibit both cancer cell growth and topoisomerase I and II activity. Binding studies of derivatives 1-4 with calf thymus DNA were also performed using a number of techniques (UV-Vis and fluorescence spectroscopy, thermal denaturation, linear dichroism and viscometry) to determine the nature of the interaction between the compounds and ctDNA. The binding constants for the complexes of the studied acridine derivatives with DNA were calculated from UV-Vis spectroscopic titrations (K=3.1×10(4)-2.0×10(3)M(-1)). Some of the compounds showed a strong inhibitory effect against Topo II at the relatively low concentration of 5µM. Topo I/II inhibition mode assays were also performed and verified that the novel compounds are topoisomerase suppressors rather than poisons. The biological activities of derivatives were studied using MTT assay and flow cytometric methods (detection of mitochondrial membrane potential, measurement of cell viability) after 24 and 48h incubation. The ability of derivatives to impair cell proliferation was tested by an analysis of cell cycle distribution.


Subject(s)
Acridines/pharmacology , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type I/metabolism , Leukemia, Promyelocytic, Acute/drug therapy , Topoisomerase I Inhibitors/pharmacology , Topoisomerase II Inhibitors/pharmacology , Acridines/chemical synthesis , Acridines/metabolism , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Chemistry, Pharmaceutical , DNA/chemistry , DNA/metabolism , Dose-Response Relationship, Drug , HL-60 Cells , Hot Temperature , Humans , Leukemia, Promyelocytic, Acute/enzymology , Leukemia, Promyelocytic, Acute/pathology , Membrane Potential, Mitochondrial/drug effects , Nucleic Acid Conformation , Nucleic Acid Denaturation , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Technology, Pharmaceutical/methods , Time Factors , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/metabolism , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism , Viscosity
6.
Photodiagnosis Photodyn Ther ; 12(3): 490-503, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26003114

ABSTRACT

BACKGROUND: Pretreatment with 5-LOX pathway inhibitor MK-886 potentiates cytotoxic effects of photodynamic therapy mediated by natural photosensitizer, hypericin. In this study, we focused on elucidating mechanisms beyond the increased efficacy of combined treatment. METHODS: Metabolic activity/viability, caspase-3 activation/mitochondrial membrane potential dissipation, intracellular hypericin level, glutathione level and redox status (NAD(P)H/oxidized flavins ratio) analyses, as well as drug efflux assays, were performed by flow cytometry. Changes in protein expression of ATP-binding cassette transporters, GDF-15 and other selected proteins were evaluated by Western blotting. Silencing of gdf-15 was carried out to verify its role in response to treatment. RESULTS: MK-886 pretreatment led to a concentration-dependent increase in intracellular hypericin content, accompanied by changes in ATP-binding cassette transporters levels and efflux efficiency. Intracellular accumulation of cytokine GDF-15 correlated with increased cell death markers; however, the impact of gdf-15 silencing on the evaluated markers was negligible. A marked decrease in the glutathione level of a majority of cells was observed after more toxic combination treatment. CONCLUSION: The significant increase in cell death markers after combination treatment confirms the potentiating effect of MK-886 on hypericin-mediated photodynamic therapy in HT-29 and MCF-7 cells. Although BCRP downregulation was not confirmed as leading mechanism responsible for elevated levels of hypericin content, changes in expression and efflux activity of ABC transporters caused by MK-886 suggest its potential in combination treatment with drugs that are substrates of these transporters, predominantly MRP1. However, complex cellular response to MK-886 pretreatment needs to be considered and further elucidated.


Subject(s)
Indoles/pharmacology , Neoplasms/drug therapy , Perylene/analogs & derivatives , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , ATP-Binding Cassette Transporters/biosynthesis , Anthracenes , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Drug Synergism , Glutathione/biosynthesis , Growth Differentiation Factor 15/antagonists & inhibitors , Humans , Membrane Potential, Mitochondrial/drug effects , Oxidation-Reduction , Perylene/pharmacology , RNA, Small Interfering/metabolism
7.
Bioorg Chem ; 59: 168-76, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25827869

ABSTRACT

This study examines the binding properties of a series of newly synthetized tacrine derivatives 1-4 and their anticancer effects. Spectroscopic techniques (UV-Vis, fluorescence spectroscopy, thermal denaturation, and linear spectropolarimetry) and viscometry were used to study DNA binding properties and to determine the types of DNA interaction with the studied derivatives. The binding constants for the complexes with DNA were obtained using UV-Vis spectroscopic titrations (K = 1.6 × 10(4)-4.0 × 10(5) M(-1)) and electrophoretic methods were used to determine the effect of the derivatives on topoisomerase I and II activity. Monotacrine derivative 1 showed evidence of topoisomerase Irelaxation activity at a concentration of 30 × 10(-6) M, while bistacrine derivatives 2-4 produced a complete inhibition of topoisomerase Iat a concentration of 5 × 10(-6) M. The biological activities of the derivatives were studied using MTT-assay and flow cytometric methods (detection of mitochondrial membrane potential and measurement of cell viability) following incubation of 24 and 48 h with human leukemic cancer cell line HL60. The ability of the derivatives to impair cell proliferation was also tested through the analysis of cell cycle distribution.


Subject(s)
Antineoplastic Agents/pharmacology , Tacrine/pharmacology , Topoisomerase I Inhibitors/pharmacology , Topoisomerase II Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA/metabolism , DNA Topoisomerases/metabolism , HL-60 Cells , Humans , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Leukemia/drug therapy , Leukemia/metabolism , Tacrine/chemistry , Topoisomerase I Inhibitors/chemistry , Topoisomerase II Inhibitors/chemistry
8.
Bioorg Chem ; 57: 13-29, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25171773

ABSTRACT

A novel series of trisubstituted acridines were synthesized with the aim of mimicking the effects of BRACO19. These compounds were synthesized by modifying the molecular structure of BRACO19 at positions 3 and 6 with heteroacyclic moieties. All of the derivatives presented in the study exhibited stabilizing effects on the human telomeric DNA quadruplex. UV-vis spectroscopy, circular dichroism, linear dichroism and viscosimetry were used in order to study the nature of the DNA binding in more detail. The results show that all of the novel derivatives were able to fold the single-stranded DNA sequences into antiparallel G-quadruplex structures, with derivative 15 exhibiting the highest stabilizing capability. Cell cycle analysis revealed that a primary trend of the "braco"-like derivatives was to arrest the cells in the S- and G2M-phases of the cell cycle within the first 72h, with derivative 13 and BRACO19 proving particularly effective in suppressing cell proliferation. All studies derivatives were less toxic to human fibroblast cell line in comparison with HT 29 cancer cell line.


Subject(s)
Acridines/chemistry , Acridines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA/metabolism , G-Quadruplexes/drug effects , Animals , Cattle , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , Humans , Ligands , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/metabolism
9.
Toxicol In Vitro ; 28(7): 1259-73, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24994473

ABSTRACT

St. John's wort (SJW, Hypericum perforatum L.) is a commonly used natural antidepressant responsible for the altered toxicity of some anticancer agents. These interactions have been primarily attributed to the hyperforin-mediated induction of some pharmacokinetic mechanisms. However, as previously demonstrated by our group, hypericin induces the expression of two ABC transporters: multidrug resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP). Because cisplatin (CDDP) and mitoxantrone (MTX) are potential substrates of ABC transporters, we investigated the effect of 24h hypericin pre-treatment on the cytotoxicity of CDDP and MTX in human cancer cell lines. CDDP-sensitive and -resistant ovarian adenocarcinoma cell lines A2780/A2780cis, together with HL-60 promyelocytic leukemia cells and ABCG2-over-expressing cBCRP subclone, were used in our experiments. We present CDDP cytotoxicity attenuated by hypericin pre-treatment in both A2780 and A2780cis cells and MTX cytotoxicity in HL-60 cells. In contrast, hypericin potentiated MTX-induced death in cBCRP cells. Interestingly, hypericin did not restore cell proliferation in rescued cells. Nevertheless, hypericin did increase the expression of MRP1 transporter in A2780 and A2780cis cells indicating the impact of hypericin on certain resistance mechanisms. Additionally, our results indicate that hypericin may be the potential substrate of BCRP transporter. In conclusion, for the first time, we report the ability of hypericin to affect the onset and/or progress of CDDP- and MTX-induced cell death, despite strong cell cycle arrest. Thus, hypericin represents another SJW metabolite that might be able to affect the effectiveness of anti-cancer drugs and that could interact with ABC transporters, particularly with BCRP.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Mitoxantrone/pharmacology , Perylene/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Anthracenes , Cell Death/drug effects , Cell Line, Tumor , Drug Interactions , Humans , Hypericum , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Perylene/pharmacology , RNA, Messenger/metabolism
10.
Photodiagnosis Photodyn Ther ; 10(4): 470-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24284100

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) is a highly efficient approach for tumour therapy, though it also has its drawbacks, too. There are multiple mechanisms involved in cell death regulation that can be successfully targeted for improvement of PDT in particular cases. We assumed, however, that the potential to manage radical stress might be the primary factor responsible for resistance to hypericin-mediated PDT (HY-PDT). METHODS: We compared the sensitivity of six colon-derived cancer cell lines to HY-PDT at IC50 equitoxic doses acquired by formazan-based (MTT) assay. Intracellular hypericin content, cell survival/metabolic activity, caspase-3 activation/mitochondrial membrane potential dissipation, apoptosis, glutathione level, redox status (NAD(P)H/oxidized flavins ratio) and Western blot analyses of proteins relevant in apoptosis regulation were measured to demonstrate differences between tested cell lines. RESULTS: Analyses revealed a whole spectrum of responses from insignificant to high cytotoxicity, despite the MTT-based "equitoxicity". Further critical evaluation of multiple parameters linked to cell physiology and proteomics proved that intracellular hypericin content, glutathione level or redox status demonstrate partial but not direct correlation with resistance to HY-PDT, when considered separately. However, their logical conjunction did copy the trend of cellular resistance. CONCLUSIONS: We may conclude that intracellular level of hypericin and glutathione together with redox state of the target cell represent a potential combination of parameters responsible for the primary cytotoxicity of HY-PDT. We also present evidence that cytotoxic assays, such as the MTT, should be accompanied with other tests of cell survival/cytotoxicity in order to avoid incorrect conclusions.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Colonic Neoplasms/chemistry , Colonic Neoplasms/drug therapy , Glutathione/metabolism , Perylene/analogs & derivatives , Photochemotherapy , Anthracenes , Cell Line, Tumor , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/radiation effects , Humans , Oxidation-Reduction/drug effects , Oxidation-Reduction/radiation effects , Perylene/analysis , Perylene/therapeutic use , Photosensitizing Agents/analysis , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use
12.
Eur J Med Chem ; 57: 283-95, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23072739

ABSTRACT

A series of 3,6-bis(3-alkylguanidino) acridines was prepared and the interaction of these novel compounds with calf thymus DNA was investigated with UV-vis, fluorescence and circular dichroism spectroscopy, in addition to DNA melting techniques. The binding constants K were estimated to range from 1.25 to 5.26 × 10(5) M(-1), and the percentage of hypochromism was found to be 17-42% (from spectral titration). UV-vis, fluorescence and circular dichroism measurements indicated that the compounds act as effective DNA-intercalating agents. Electrophoretic separation proved that ligands 6a-e relaxed topoisomerase I at a concentration of 60 µM, although only those with longer alkyl chains were able to penetrate cell membranes and suppress cell proliferation effectively. The biological activity of novel compounds was assessed using different techniques (cell cycle distribution, phosphatidylserine externalization, caspase-3 activation, changes in mitochondrial membrane potential) and demonstrated mostly transient cytostatic action of the ethyl 6c and pentyl 6d derivatives. The hexyl derivative 6e proved to be the most cytotoxic. Different patterns of cell penetration were also observed for individual derivatives. Principles of molecular dynamics were applied to explore DNA-ligand interactions at the molecular level.


Subject(s)
Acridines/chemical synthesis , Antineoplastic Agents/chemical synthesis , DNA Topoisomerases, Type I/chemistry , DNA/chemistry , Guanidines/chemical synthesis , Intercalating Agents/chemical synthesis , Acridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Cattle , Cell Cycle/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability , Circular Dichroism , Guanidines/pharmacology , HL-60 Cells , Humans , Intercalating Agents/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Molecular Dynamics Simulation , Nucleic Acid Denaturation , Phosphatidylserines/metabolism , Spectrometry, Fluorescence , Structure-Activity Relationship
13.
Toxicol In Vitro ; 26(6): 775-82, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22683934

ABSTRACT

Proadifen (SKF-525A) is a well-known inhibitor of cytochrome P450 monooxygenases. Besides the prevention of drug metabolism it affects the proliferation of cancer cells, although the mechanisms of possible anti-cancer activity of proadifen have not been fully understood yet. The aim of this study therefore was to evaluate the potential anti-proliferative effect of proadifen on HT-29 colon cancer cells. Our results show that proadifen inhibited the growth of HT-29 cells by the accumulation of cells in the G1 phase of the cell cycle, reduction of metabolic activity and colony formation and by the induction of apoptosis. Analyses of Western blots and flow cytometry revealed time- and dose-dependent phosphatidylserine externalization, caspase-3 activation and PARP cleavage. Intense upregulation of NAG-1 and ATF3 and downregulation of Mcl-1 and Egr-1 were also observed. Further investigation showed that NAG-1 gene silencing by siRNA had no effect on the pro-apoptotic action of proadifen. In contrast, we found that AR-A014418, the specific inhibitor of glycogen synthase kinase-3 ß (GSK-3ß), significantly decreased proadifen-induced apoptosis. Inactivation of GSK-3ß (phosphorylation at serine 9) resulted in changes in phosphatidylserine externalization and caspase-3 activation. These data suggest that GSK-3ß is an important factor in the induction of apoptosis in HT-29 colon cancer cells treated with proadifen.


Subject(s)
Antineoplastic Agents/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Proadifen/pharmacology , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Urea/analogs & derivatives , Adenocarcinoma , Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms , Glycogen Synthase Kinase 3 beta , HT29 Cells , Humans , Urea/pharmacology
14.
Photochem Photobiol Sci ; 10(4): 626-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21298151

ABSTRACT

Preferential uptake of photosensitizer by tumour tissue is an elementary prerequisite of effective and successful photodynamic therapy (PDT). Therefore intracellular concentration of photosensitizer is one of the limiting factors affecting PDT efficiency. Hypericin (HY) has found applications in photodynamic diagnostics solely due to its high specificity for tumour cells and tissues. However, here we suggest that not only HY uptake, but importantly also the cell ability to manage oxidative stress induced by HY-PDT can be important decisive factors finally affecting the cell death response. We showed that despite the higher accumulation of HY in FHC human fetal colon epithelial cells compared to HT-29 colon adenocarcinoma cells, the cytotoxic effects of this photosensitizer were more pronounced in the latter cell line, and this was associated with enhanced accumulation of HY-PDT-induced reactive oxygen species (ROS).


Subject(s)
Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Epithelial Cells/drug effects , Perylene/analogs & derivatives , Photochemotherapy , Photosensitizing Agents/therapeutic use , Anthracenes , Cell Line , Colon/cytology , Epithelial Cells/radiation effects , Fetus , Humans , Microscopy, Fluorescence , Perylene/therapeutic use , Perylene/toxicity , Photosensitizing Agents/toxicity , Reactive Oxygen Species/metabolism
15.
Acta Histochem ; 113(2): 156-62, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20079525

ABSTRACT

Despite extensive investigations of gliogenesis, the time of origin of ependymal cells in the spinal cord has not yet been fully elucidated. Using a single dose of 5-bromo-2-deoxyuridine combined with various survival times we monitored: mitotic activity (short survival time), the presence of newly formed cells in the ventricular zone (intermediate survival time) and the formation of ependymal cells (long survival time) during the late embryonic and early postnatal development in the ventricular zone of the spinal cord of rats. In the period of study it was found that the ependymal cells populated this region in two waves. Most of the ependymal cells originated around embryonic day 18 and then between postnatal days 8 and 15. In addition, it was observed that in the ventricular zone of the spinal cord, proliferation and production of ependymal cells continues at the slower rate at least until day 36 of postnatal development. Elucidation of the relationship between progenitors in the embryonic ventricular zone and the relative quiescent ependymal lining of the central canal in adulthood could be important in the search for the adult neural stem cell niche.


Subject(s)
Ependyma/cytology , Spinal Cord/cytology , Animals , Bromodeoxyuridine/chemistry , Cell Proliferation , Embryo, Mammalian , Female , Immunohistochemistry , Male , Rats , Rats, Wistar
16.
Radiat Res ; 175(1): 51-6, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21175347

ABSTRACT

Photodynamic therapy is an alternative method for cancer treatment in which a photosensitizer exposed to a light source of suitable wavelength is excited and can subsequently react through free radical mechanisms. Recently, oxygen free radical-mediated changes in gene expression have been established. The present study shows the effect of photoactivated hypericin on the expression of the human epidermal growth factor receptor 2 (HER2) oncogene at both the mRNA and the protein level in SKBR-3 and MCF-7 breast adenocarcinoma cell lines. The photodynamic therapy-induced decrease in mRNA expression was reversed by the singlet oxygen scavenger trolox, which supports a role for singlet oxygen. In addition, prevention of the generation of reactive oxygen species by pretreatment with trolox effectively blocked the antiproliferation activity of photoactivated hypericin. These results may have important implications at least for recurrent breast cancer with HER2 expression alone or in combination with conventional therapies.


Subject(s)
Genes, erbB-2 , Perylene/analogs & derivatives , Photochemotherapy , Receptor, ErbB-2/antagonists & inhibitors , Anthracenes , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Female , Humans , Perylene/pharmacology , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/analysis , Receptor, ErbB-2/genetics , Singlet Oxygen/physiology
17.
Photochem Photobiol Sci ; 9(9): 1244-51, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20714672

ABSTRACT

Since many studies have suggested the impact of dietary polyunsaturated fatty acids on cancer progression and prognosis, there is an assumption of possible pre-sensitizing effects of their application in combined treatment. The present work evaluates modulation of photodynamic therapy (PDT) with hypericin by pre-treatment with n-3 and n-6 fatty acids in HT-29 and HeLa tumour cells. We observed stimulation of cytotoxic effects by docosahexaenoic acid (n-3) and arachidonic acid (n-6) in several stages of action in both cell lines. Treatment with either fatty acids or photodynamic therapy alone induced apoptosis in a dose- and time-dependent manner; however the effect was even more striking in mutual combination applied as pre-treatment with fatty acids prior to photodynamic therapy. Moreover, the combination also induced changes in membrane lipid composition leading to alteration in cell membrane fluidity. Increased toxicity of combined treatment was also confirmed by the presence of oxidative stress demonstrated by ROS production, RNS accumulation and increased presence of lipoperoxides. In conclusion, we suggest that pre-treatment with polyunsaturated fatty acids may contribute to cytotoxic effects induced by photodynamic therapy with hypericin.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis , Fatty Acids, Unsaturated/toxicity , Oxidative Stress , Perylene/analogs & derivatives , Anthracenes , Arachidonic Acid/toxicity , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Docosahexaenoic Acids/toxicity , HT29 Cells , Humans , Membrane Fluidity , Perylene/therapeutic use , Photochemotherapy
18.
Photochem Photobiol ; 86(1): 200-5, 2010.
Article in English | MEDLINE | ID: mdl-19912559

ABSTRACT

Current treatment of breast cancer is often affected by resistance to therapeutics, for which the human epidermal growth factor receptor 2 (HER2) may be responsible. Here, we report for the first time the use of hypericin-mediated photodynamic therapy (HY-PDT) in combination with a selective HER2 inhibitor (AG 825) on SKBR-3, a HER2 overexpressing human breast adenocarcinoma cell line. The results demonstrate that HY-PDT is able to degrade HER2 with an impact on its signaling cascade. Combination with AG 825 resulted in increased apoptosis induction, total degradation of HER2 and inhibition of colony formation. Downregulation of HSP90, Mcl-1, Bcl-xL and upregulation of Bax was also observed. This knowledge provides the basis for the possible application of HY-PDT in preclinical and clinical models of breast cancer treatment.


Subject(s)
Breast Neoplasms/therapy , Photochemotherapy/methods , Receptor, ErbB-2/metabolism , Anthracenes , Apoptosis/drug effects , Benzothiazoles/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Perylene/analogs & derivatives , Receptor, ErbB-2/antagonists & inhibitors , Signal Transduction/drug effects , Tyrphostins/pharmacology
19.
Photochem Photobiol Sci ; 8(12): 1716-23, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20024169

ABSTRACT

Photodynamic therapy (PDT) is a flexible multi-target therapeutic approach. One of the main requirements of successful PDT is sufficient intracellular concentration of an applicable photosensitizer. Mechanisms of anticancer drug elimination by tumour cells are mostly linked to the elevated expression and activity of P-glycoprotein (P-gp), multidrug resistance-associated protein 1 (MRP1), breast cancer resistance protein (BCRP) and P450 monooxygenases. The interaction of hypericin with this cell drug-defence system is still unclear. We report here for the first time increased activity of MRP1 and BCRP in HT-29 colon cancer cells treated with hypericin per se. On the contrary, pre-treatment with proadifen (SKF525A) affected the function of MRP1 and BCRP leading to increased hypericin content, which might indicate a possible link between proadifen and these ABC transporter proteins. Subsequent enhanced intracellular oxidative stress was accompanied by loss of mitochondrial membrane potential, activation of caspase-9 and -3, PARP cleavage and onset of apoptosis. In conclusion, our study suggests that drug efflux transporters MRP1 and BCRP affect the pharmacokinetics of hypericin in HT-29 colon adenocarcinoma cells, and the action of hypericin-mediated PDT (HY-PDT) should be modulated by pre-treatment with their specific inhibitors.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Perylene/analogs & derivatives , Photosensitizing Agents/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Adenocarcinoma/secondary , Anthracenes , Caspase 3/metabolism , Caspase 9/metabolism , Enzyme Inhibitors/pharmacology , HT29 Cells , Humans , Light , Membrane Potential, Mitochondrial/drug effects , Mixed Function Oxygenases/metabolism , Perylene/pharmacology , Photochemotherapy , Proadifen/pharmacology , Reactive Oxygen Species/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...