Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add filters








Language
Year range
1.
Journal of Integrative Medicine ; (12): 143-153, 2016.
Article in English | WPRIM | ID: wpr-317038

ABSTRACT

<p><b>OBJECTIVE</b>To provide in vitro evidence of Psorinum treatment against cancer cells in a controlled study.</p><p><b>METHODS</b>Effects of homeopathic Psorinum 6× on cell viability were initially determined in several cancer cell lines, including A549, HepG2 and MCF-7, using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and an ethanol 6× control. The cell line that exhibited highest inhibition was selected and used in the following experiments. A range of Psorinum 6× doses was used to explore treatment effects on cell cycle arrest, cell death (apoptosis), generation of reactive oxygen species (ROS) and change in mitochondrial membrane potential (MMP) using flow cytometry and fluorescence microscopy, respectively. Expression of several signal proteins related to apoptosis and cell survival were quantified with Western blotting and confocal microscopy. Further, circular dichroism (CD) spectroscopy was used to determine possible drug-DNA interactions, as well as the induction of conformational changes.</p><p><b>RESULTS</b>Treatment of cancer cell lines with Psorinum showed greater anticancer effects in A549 cells than in others. In A549 cells Psorinum treatment inhibited cell proliferation at 24 h after treatment, and arrested cell cycle at sub-G1 stage. It also induced ROS generation, MMP depolarization, morphological changes and DNA damage, as well as externalization of phosphatidyl serine. Further, increases in p53 expression, Bax expression, cytochrome c release, along with reduction of Bcl-2 level and caspase-3 activation were observed after Psorinum 6× treatment, which eventually drove A549 cells towards the mitochondria-mediated caspase-3-dependent pathway. CD spectroscopy revealed direct interaction of Psorinum with DNA, using calf thymus-DNA as target.</p><p><b>CONCLUSION</b>Psorinum 6× triggered apoptosis in A549 cells via both up- and down-regulations of relevant signal proteins, including p53, caspase-3, Bax and Bcl-2.</p>


Subject(s)
Humans , Caspase 3 , Metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Homeopathy , Lung Neoplasms , Drug Therapy , Pathology , Proto-Oncogene Proteins c-bcl-2 , Reactive Oxygen Species , Metabolism , bcl-2-Associated X Protein
2.
Journal of Integrative Medicine ; (12): 209-218, 2016.
Article in English | WPRIM | ID: wpr-317030

ABSTRACT

<p><b>OBJECTIVE</b>Homeopathic nosodes have seldom been scientifically validated for their anticancer effects. This study was conducted to examine if a recently developed hepatitis C nosode has demonstrable anticancer potential in cancer cells in vitro.</p><p><b>METHODS</b>Anticancer effects of Hepatitis C 30C (Hep C 30), if any, were initially tested on three cancer cell lines, HepG2 (liver cancer), MCF-7 (breast cancer) and A549 (lung cancer) and one normal liver cell line WRL-68 cells and subsequently a more thorough study using further scientific protocols was undertaken on HepG2 cells (against WRL-68 cells as the normal control) as HepG2 cells showed better anticancer response than the other two. Three doses, one at 50% lethal dose (LD50) and the other two below LD50, were used on HepG2 cells subsequently. Protocols like apoptosis induction and its possible signaling mechanism were deployed using immunoblots of relevant signal proteins and confocal microscopy, with particular reference to telomerase and topoisomerase II (Top II) activities, two strong cancer biomarkers for their direct relationship with divisional activities of cells and DNAs.</p><p><b>RESULTS</b>Hep C 30 induced apoptosis, caused distorted cell morphology typical of apoptotic cells, increased reactive oxygen species generation and produced increased DNA nicks. Further it enhanced pro-apototic signal proteins like Bax, cytochrome c and inhibited anti-apoptotic signal proteins, Bcl-2, cytochrome c and caspase-3, changed mitochondrial membrane potential and caused externalization of phosphatidylserine. The drug also decreased expression of two cancer biomarkers, Top II and telomerase, consistent with its anticancer effect.</p><p><b>CONCLUSION</b>Hep C 30 has demonstrable anticancer effects against liver cancer cells in vitro.</p>


Subject(s)
Humans , Antineoplastic Agents , Pharmacology , Apoptosis , Cell Survival , DNA Topoisomerases, Type II , Metabolism , Hep G2 Cells , Hepacivirus , Liver Neoplasms , Drug Therapy , Pathology , Materia Medica , Mitochondria , Physiology , Telomerase , Metabolism
3.
Journal of Integrative Medicine ; (12): 425-438, 2014.
Article in English | WPRIM | ID: wpr-308184

ABSTRACT

<p><b>OBJECTIVE</b>Use of cisplatin, a conventional anticancer drug, is restricted because it generates strong hepatotoxicity by accumulating in liver. Therefore its anticancer potential can only be fully exploited if its own toxicity is considerably reduced. Towards this goal, ethanolic extract of the plant, Boldo (Peumus boldus), known for its antihepatotoxic effects, was used simultaneously with cisplatin, to test its ability to reduce cisplatin's cytotoxicity without affecting its anticancer potential.</p><p><b>METHODS</b>The cytotoxicity of Boldo extract (BE) and cisplatin, administered alone and in combination, was determined in three cancer cell lines (A549, HeLa, and HepG2) and in normal liver cells (WRL-68). Drug-DNA interaction, DNA damage, cell cycle, apoptosis, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP, ΔΨ) were also studied. Hepatotoxicity and antioxidant activity levels were determined by alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase and glutathione assays in mice. The cytotoxicity of related proteins was tested by Western blotting.</p><p><b>RESULTS</b>Co-administration of BE and cisplatin increased viability of normal cells, but had no effect on the viability of cancer cells. Boldo protected liver from damage and normalized different antioxidant enzyme levels in vivo and also reduced ROS and re-polarized MMP in vitro. Bax and cytochrome c translocation was reduced with caspase 3 down-regulation. Further, a drug-DNA interaction study revealed that BE reduced cisplatin's DNA-binding capacity, resulting in a reduction in DNA damage.</p><p><b>CONCLUSION</b>Results indicated that a low dose of BE could be used beneficially in combination with cisplatin to reduce its toxicity without hampering cisplatin's anticancer effect. These findings signify a potential future use of BE in cancer therapy.</p>


Subject(s)
Animals , Female , Humans , Male , Mice , Antineoplastic Agents , Toxicity , Cells, Cultured , Cisplatin , Toxicity , DNA Damage , Glutathione , Metabolism , Hepatocytes , Metabolism , Pathology , Neoplasms , Drug Therapy , Pathology , Peumus , Plant Extracts , Pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL