Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Integr Cancer Ther ; 21: 15347354221124861, 2022.
Article in English | MEDLINE | ID: mdl-36154723

ABSTRACT

Owing to the crucial role of Src in cancer metastasis, interruption of Src and its signaling has been considered a promising strategy for cancer metastasis treatment. Cucurbitacin B, a dietary triterpenoid, has been shown to possess anti-proliferative and apoptosis-inducing activities in cholangiocarcinoma (CCA) cells via suppressing the activation of FAK which is a main downstream Src effector. We hypothesized that cucurbitacin B might act as a Src suppressant which conferring anti-metastasis effect against CCA cells. To investigate this, the role of Src in regulating metastasis behavior of CCA cells and the effect of cucurbitacin B on Src-mediated metastatic phenotype of these cells were determined. The results showed that activation of Src significantly enhanced the migratory and invasive abilities of CCA cells. Molecular analysis revealed that Src-facilitated metastasis behavior of CCA cells occurred by modifying expression of a wide range of metastasis-related genes in the cells. Consistent with gene expression results, activation of Src significantly induced the protein expression of 2 important metastasis-associated molecules, MMP-9 and VEGF. Cucurbitacin B markedly suppressed activation of Src and its key effector, FAK. As a consequence, the alteration of expression profiles of metastasis-associated genes induced by Src activator in CCA cells was diminished by cucurbitacin B treatment. The compound also down-regulated Src-induced expression of MMP-9 and VEGF proteins in the cells. Moreover, molecular docking analysis revealed that cucurbitacin B could interact with Src kinase domain and possibly restrain the kinase from being activated by hindering the ATP binding. In conclusion, cucurbitacin B exhibited anti-metastatic property in CCA cells via negatively influencing Src and Src-related oncogenic signaling. This compound may therefore be a potential therapeutic drug for further development as an anti-Src agent for treatment of metastatic CCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Triterpenes , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/therapeutic use , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Humans , Matrix Metalloproteinase 9 , Molecular Docking Simulation , Triterpenes/pharmacology , Triterpenes/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , src-Family Kinases/pharmacology , src-Family Kinases/therapeutic use
2.
J Pharm Pharmacol ; 74(7): 996-1005, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35640567

ABSTRACT

OBJECTIVES: Strategies that induce apoptosis of malignant cells are recognized as effective cancer treatments. This study evaluated the apoptosis-inducing ability of momordin Ic against cholangiocarcinoma (CCA) cells and the respective underlying mechanisms. METHODS: Quantification of apoptotic cells was performed using Annexin V/7-AAD double dye staining followed by flow cytometry. The effect of momordin Ic on the expression of epidermal growth factor receptor (EGFR) and its downstream signalling molecules was determined via Western blot analysis. The RT2 Profiler PCR Array was used to determine the expression of cell death-associated genes. Expression levels of apoptosis-related proteins were examined using an apoptosis antibody array. KEY FINDINGS: Momordin Ic potently limited the ability of CCA cells to thrive by promoting apoptotic cell death. This apoptosis-inducing activity was accompanied with suppression of expression of EGFR, p-EGFR, c-Myc and other downstream EGFR signalling-related molecules. Additional molecular analyses demonstrated that momordin Ic modified the expression profile of cell death-associated genes in CCA cells. Moreover, significant upregulation of apoptosis-activating proteins and downregulation of apoptosis-inhibiting protein were also observed after exposure to momordin Ic. CONCLUSIONS: These results suggest that momordin Ic has a potential therapeutic opportunity for CCA treatment by acting as an EGFR suppressant.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Apoptosis , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cell Line, Tumor , Cell Proliferation , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , ErbB Receptors , Humans , Plant Extracts
3.
Am J Chin Med ; 48(6): 1475-1489, 2020.
Article in English | MEDLINE | ID: mdl-32907364

ABSTRACT

Inadequate responses to traditional chemotherapeutic agents in cholangiocarcinoma (CCA) emphasize a requirement for new effective compounds for the treatment of this malignancy. This study aimed to investigate the antiproliferative property of cucurbitacin B on KKU-100 CCA cells. The determination of underlying molecular mechanisms was also carried out. The results revealed that cucurbitacin B suppressed growth and replicative ability to form colonies of CCA cells, suggesting the antiproliferative effect of this compound against the cells. Flow cytometry analysis demonstrated that the interfering effect of cucurbitacin B on the CCA cell cycle at the G2/M phase was accountable for its antiproliferation property. Accompanied with cell cycle disruption, cucurbitacin B altered the expression of proteins involved in the G2/M phase transition including downregulation of cyclin A, cyclin D1, and cdc25A, and upregulation of p21. Additional molecular studies demonstrated that cucurbitacin B suppressed the activation of focal adhesion kinase (FAK) which consequently resulted in inhibition of its kinase-dependent and kinase-independent downstream targets contributing to the regulation of cell proliferation including PI3K/PDK1/AKT and p53 proteins. In this study, the transient knockdown of FAK using siRNA was employed to ascertain the role of FAK in CCA cell proliferation. Finally, the effect of cucurbitacin B on upstream receptor tyrosine kinases regulating FAK activation was elucidated. The results showed that the inhibitory effect of cucurbitacin B on FAK activation in CCA cells is mediated via interference of EGFR and HER2 expression. Collectively, cucurbitacin B might be a promising drug for CCA treatment by targeting FAK protein.


Subject(s)
Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Triterpenes/pharmacology , Tumor Suppressor Protein p53/metabolism , Bile Duct Neoplasms/diet therapy , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/drug effects , Humans , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Triterpenes/therapeutic use
4.
Integr Cancer Ther ; 18: 1534735418820444, 2019.
Article in English | MEDLINE | ID: mdl-30584780

ABSTRACT

Altered expression of a cytosolic flavoenzyme NAD(P)H:quinone oxidoreductase-1 (NQO1) has been seen in many human tumors. Its remarkable overexpression in cholangiocarcinoma (CCA; an aggressive malignancy of the biliary duct system) was associated with poor prognosis and short survival of the patients. Inhibition of NQO1 has been proposed as a potential strategy to improve the efficacy of anticancer drugs in various cancers including CCA. This study investigated novel NQO1 inhibitors and verified the mechanisms of their enzyme inhibition. Among the different chemical classes of natural NQO1 inhibitors are coumarins, flavonoids, and triterpenoids. Coumarins are a group of particularly potent NQO1 inhibitors. The mechanisms and kinetics of enzyme inhibition of coumarin, aesculetin, umbelliferone, and scopoletin using the cell lysates as a source of NQO1 enzyme best fit with an uncompetitive inhibition model. Among the NOQ1 inhibitors tested in KKU-100 CCA cells, scopoletin and umbelliferone had the strongest inhibitory effect on this enzyme, while aesculetin and coumarin barely affected intracellular NQO1. All coumarins were further tested for cytotoxicity and anti-migration activity. At modest cytotoxic doses, scopoletin and umbelliferone greatly inhibited the migration of KKU-100 cells, whereas coumarin and aesculetin barely reduced cell migration. The anti-migration effect of scopoletin was associated with decreased ratio of matrix metalloproteinase 9/tissue inhibitors of metalloproteinases 1 ( MMP9/ TIMP1) mRNA. These findings suggest that natural compounds with potent inhibitory effect on intracellular NQO1 have useful anti-migration effects on CCA cells. In order to prove that the potent NQO1 inhibitor, scopoletin, is clinically useful in the enhancement of CCA treatment, additional in vivo studies to elucidate the mechanism of these effects are needed.


Subject(s)
Antineoplastic Agents/therapeutic use , Bile Duct Neoplasms/drug therapy , Cell Movement/drug effects , Cholangiocarcinoma/drug therapy , Coumarins/therapeutic use , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cholangiocarcinoma/pathology , Hep G2 Cells , Humans , Kinetics , Matrix Metalloproteinase 9/metabolism , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Umbelliferones/therapeutic use
5.
J Pharm Pharmacol ; 68(9): 1184-92, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27334841

ABSTRACT

OBJECTIVES: To investigate the apoptosis-inducing effect and underlying mechanisms of luteolin in cholangiocarcinoma (CCA) cells. METHODS: Cell viability was determined by sulphorhodamine B. Apoptosis was detected using acridine orange/ethidium bromide dye staining and annexin V/PI staining followed by flow cytometry. The effect of luteolin on the oxidative status of CCA cells was evaluated by measuring intracellular reactive oxygen species (ROS) levels using the dihydroethidium method and quantifying glutathione levels. The mitochondria transmembrane potential (ΔΨm) was examined through JC-1 staining. The protein levels were determined by Western blot. Caspase activity was determined using specific fluorogenic substrates. KEY FINDINGS: Luteolin decreased KKU-100 CCA cells' viability by induction of apoptosis. Luteolin treatment increased ROS production and decreased glutathione levels. These changes were associated with the decrease of Nrf2, γ-glutamylcysteine ligase and heme oxygenase-1 proteins. Moreover, luteolin induced mitochondrial depolarization, which was accompanied by the release of cytochrome c and a decrease of Bcl-2 and Bcl-XL proteins. Pretreatment with antioxidants, 4-hydroxy-TEMPO and N-acetyl-L-cysteine significantly prevented luteolin-induced CCA cell death and loss of ΔΨm. In addition, luteolin induced the activation of caspase-9 and caspase-3. CONCLUSIONS: Luteolin exerts its pro-apoptotic action partly through generating intracellular ROS that then contributes to the activation of mitochondria-mediated apoptotic cell death.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Luteolin/pharmacology , Mitochondria/drug effects , Phytotherapy , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Bile Duct Neoplasms/drug therapy , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Cell Survival , Cholangiocarcinoma/drug therapy , Cytochromes c/metabolism , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Heme Oxygenase-1/metabolism , Humans , Luteolin/therapeutic use , Membrane Potential, Mitochondrial/drug effects , Mitochondria/physiology , NF-E2-Related Factor 2/metabolism , Plant Extracts/therapeutic use
6.
Nutr Cancer ; 68(2): 328-41, 2016.
Article in English | MEDLINE | ID: mdl-26908056

ABSTRACT

Cratoxylum formosum Dyer has been used in Southeast Asian countries both for food and folk medicine. In this study, the leaf extracts of C. formosum were evaluated for anticancer effects on human cholangiocarcinoma (CCA) KKU-M156 cells. The results showed that the plant extracts possessed potent cytotoxicity against CCA cells. The cytotoxic activity was associated with an induction of cell apoptosis. Moreover, the colony forming ability of CCA cells was also inhibited by C. formosum extracts. Consistent with growth inhibitory effects, the plant extracts induced cell cycle arrest at the G2/M phase and downregulated cyclin A and Cdc25A protein expression. The extracts potently suppressed the migration and invasion properties of CCA cells. The effects were associated with the suppression of NF-κB and STAT3 nuclear translocation and transcriptional activity, and downregulation of genes involving in cancer progression and metastasis. Furthermore, the possible bioactive compounds in the extracts were analyzed by HPLC. Taken together, the potent anticancer activity of C. formosum against CCA indicates the plant promising use for CCA prevention and therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Clusiaceae/chemistry , Plant Extracts/pharmacology , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Cyclin A/metabolism , Drug Screening Assays, Antitumor/methods , Flavonoids/analysis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxybenzoates/analysis , NF-kappa B/metabolism , Plant Extracts/chemistry , STAT3 Transcription Factor/metabolism
7.
Phytother Res ; 28(6): 841-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24038588

ABSTRACT

Quercetin and epigallocatechin-3-gallate (EGCG) are dietary phytochemicals with antiinflammatory and antitumor effects. In the present study, we examined the effects of these two compounds on Janus-like kinase (JAK)/signal transduction and transcription (STAT) pathway of cholangiocarcinoma (CCA) cells, because CCA is one of the aggressive cancers with very poor prognosis and JAK/STAT pathway is critically important in inflammation and carcinogenesis. The results showed that the JAK/STAT pathway activation by proinflammatory cytokine interleukin-6 and interferon-γ in CCA cells was suppressed by pretreatment with quercetin and EGCG, evidently by a decrease of the elevated phosphorylated-STAT1 and STAT3 proteins in a dose-dependent manner. The cytokine-mediated up-regulation of inducible nitric oxide synthase (iNOS) and intercellular adhesion molecule-1 (ICAM-1) via JAK/STAT cascade was abolished by both quercetin and EGCG pretreatment. Moreover, these flavonoids also could inhibit growth and cytokine-induced migration of CCA cells. Pretreatment with specific JAK inhibitors, AG490 and piceatannol, abolished cytokine-induced iNOS and ICAM-1 expression. These results demonstrate beneficial effects of quercetin and EGCG in the suppression of JAK/STAT cascade of CCA cells. Quercetin and EGCG would be potentially useful as cancer chemopreventive agents against CCA.


Subject(s)
Catechin/analogs & derivatives , Cholangiocarcinoma/pathology , Quercetin/pharmacology , Signal Transduction/drug effects , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Catechin/pharmacology , Cell Line, Tumor , Cholangiocarcinoma/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interleukin-6/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor , Tumor Suppressor Proteins
8.
Am J Chin Med ; 40(1): 163-75, 2012.
Article in English | MEDLINE | ID: mdl-22298456

ABSTRACT

Mulberry (Morus alba L.) leaf tea is promoted for its health benefits and the control of diabetes in Asian nations. The blood glucose lowering activity of mulberry leaf extract (MA) has been proven; however, the molecular basis underlying this effect remains unclear. The aim of the present work is to elucidate its mechanism of the antihyperglycemic action, by examining the effect of MA on glucose uptake and the translocation of glucose transporter 4 protein (GLUT4) to the plasma membrane of adipocytes isolated from diabetic rats. The incubation of adipocytes with 5-45 µg/ml MA resulted in 31-54% increase of glucose uptake in a dose-dependent manner. This glucose uptake enhancing effect was inhibited by the phosphoinositol 3-kinase (PI3-K) inhibitor, wortmannin (100 nM). The GLUT4 protein on the plasma membrane fraction of adipocytes was markedly increased after treatment with 15 µg/ml MA extract. Interestingly, gallic acid, one of the phenolic compounds found in MA extract, increased glucose uptake and enhanced the translocation of GLUT4 at concentrations comparable to the amount of gallic acid in the effective concentration ranges of MA. Thus, it is likely that gallic acid contributes, at least in part, to its antihyperglycemic activity. The present results suggest that the antihyperglycemic action of MA is mediated by increasing glucose uptake via the activation of PI3-K signaling pathway and translocation of GLUT4 to the plasma membrane. These findings are the first molecular evidence supporting the mulberry tea as herbal medicine for diabetic patients.


Subject(s)
Adipocytes/drug effects , Diabetes Mellitus, Experimental/drug therapy , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Morus/chemistry , Phytotherapy , Adipocytes/metabolism , Androstadienes/pharmacology , Animals , Biological Transport/drug effects , Cell Membrane/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Dose-Response Relationship, Drug , Gallic Acid/pharmacology , Gallic Acid/therapeutic use , Hypoglycemic Agents/therapeutic use , Male , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Leaves , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Wortmannin
9.
Asian Pac J Cancer Prev ; 13 Suppl: 161-7, 2012.
Article in English | MEDLINE | ID: mdl-23480759

ABSTRACT

Antitumor activity (growth suppression) of vitamin D has been demonstrated using cholangiocarcinoma (CCA) cell lines, CCA cell-grafted animal models, and human CCA tissue cultures. The present study aimed to determine the toxicity and tolerability of intermittent-high dose calcitriol in advanced inoperable intrahepatic CCA patients and to evaluate the therapeutic efficacy of combinations of calcitriol and 5-fluorouracil-based chemotherapeutic drugs. The patients were divided into 3 groups: the first (n=2) received intermittent-high dose oral calcitriol 12 µg/day for 3 days, i.e. Monday-Wednesday, per week up to 3 months. The treatment did not cause any serious adverse events, except hypercalcemia grade I, once in 72 administrations. The second group (n=3) received chemotherapeutic drugs (5-fluorouracil, Mitomycin C and Leucovorin) for 3 cycles, one patient showing a partial response. The third group (n=4) received high dose calcitriol in combination with chemotherapeutic-drugs. All 4 patients encountered serious adverse events and two of them were withdrawn after the first drug cycle. This pilot study suggests that, although high dose-intermittent calcitriol appeared to be safe and tolerated well in advanced intrahepatic CCA patients, co-administration with 5-fluorouracil-based chemotherapeutic drugs caused unexpected potentiation of their toxicity. Adjustment of the doses of both drugs is required to avoid such toxicity and to optimize therapeutic efficacy of anticancer drugs when they were combined with high dose-intermittent calcitriol.


Subject(s)
Calcitriol , Cholangiocarcinoma , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bile Duct Neoplasms , Bile Ducts, Intrahepatic , Calcitriol/administration & dosage , Fluorouracil/administration & dosage , Humans , Pilot Projects
10.
Plant Foods Hum Nutr ; 65(4): 403-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21104319

ABSTRACT

Several vegetables have been shown to possess cytoprotective and antioxidant effects with various mechanisms of action. The aim of this study was to determine the antioxidant effects and mechanism underlying of Syzygium gratum, a dietary and herbal plant commonly found in the Southeast Asia. Additionally, its effects on the induction of endogenous antioxidant defensive system were also investigated. Results showed that the leaf extract possessed an exceptionally strong antioxidant and intracellular oxygen radical scavenging activity in both aqueous and ethanolic extracts. The plant aqueous extract was further studied in C57BL/6J mice to evaluate its effects in vivo. The extract was well tolerated by the animals throughout the 30 days of study. The cytoprotective enzyme, heme oxygenase (HO-1) activity was significantly increased in the high dose-treated animals (1 g/kg/day). Consistent with the enzymatic activity, the expression of HO-1 mRNA tended to increase in those mice. There was no significant increase in hepatic γ-glutamylcysteine ligase (γ-GCL) activity, glutathione levels and GCL mRNA expression. Taken together, this study provides evidence that S. gratum exhibits potent direct antioxidant properties and can induce cytoprotective enzyme in vivo. Consumption of S. gratum may provide a health benefit against oxidative stress and other related disorders.


Subject(s)
Antioxidants/pharmacology , Biological Products/pharmacology , Cytoprotection , Phytotherapy , Plant Extracts/pharmacology , Syzygium/chemistry , Animals , Asia, Southeastern , Dipeptides/metabolism , Heme Oxygenase-1/metabolism , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Plant Leaves/chemistry , RNA, Messenger , Rats , Rats, Sprague-Dawley
11.
Mol Carcinog ; 47(5): 321-5, 2008 May.
Article in English | MEDLINE | ID: mdl-17932952

ABSTRACT

Phenethyl isothiocyanate (PEITC) is an isothiocyanate which is a major constituent of watercress and other cruciferous vegetables. Its chemopreventive potential has been previously shown in various rodent models of cancer. In this study, we investigated the chemopreventive efficacy of PEITC in the Apc(Min/+) mouse model. Apc(Min/+) mice were fed with diet supplemented with 0.05% of PEITC for 3-wk. Our results clearly demonstrated that Apc(Min/+) mice fed with PEITC supplemented diet developed significantly less (31.7% reduction) and smaller polyps in comparison to mice fed with the standard AIN-76A diet. Subsequent mechanistic study using Western blotting shows that inhibition of growth of adenomas by PEITC is associated with increase of apoptosis (cleaved-caspase-3, -caspase-7, and PARP). Treatments also led to the inhibition of cell cycle-related biomarkers such as the cyclins (D1, A, and E) and activation of p21. However, PEITC has no effect on the expression of p-Erk, p-JNK or p-p38. In conclusion, our results demonstrate that PEITC is a potent natural dietary compound for chemoprevention of gastrointestinal cancers. Its mechanism of actions may include induction of apoptosis and cell cycle arrest.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Anticarcinogenic Agents/therapeutic use , Genes, APC/physiology , Intestinal Neoplasms/prevention & control , Isothiocyanates/therapeutic use , Adenomatous Polyposis Coli/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Chemoprevention , Dietary Supplements , Intestinal Neoplasms/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL