ABSTRACT
Mulberry fruit water extract (MWE) has been reported to synergistically enhance the cytotoxic effect of paclitaxel by promoting mitotic catastrophe to induce apoptosis in bladder cancer cells in our previous work. The aim of this study was to evaluate and to mechanistically explore the effects of MWE on bladder cancer responses to ionizing radiation (IR) by treating TSGH 8301 bladder carcinoma cells with MWE after exposing to IR. The results of MTT assay showed a synergistic cytotoxicity of IR with the co-treatment of MWE (IR/MWE) by inducing G2/M phase arrest as demonstrated by flow cytometry analysis in TSGH 8301, HT1367 and HT1197 bladder carcinoma cells lines. The IR/MWE-treated cells expressed increased levels of the G2/M phase arrest-related proteins cdc2/cyclin B1 and displayed giant multinucleated morphology, a typical characteristic of mitotic catastrophe. Immunofluorescent confocal microscopy revealed that the combined strategy inhibited Aurora B phosphorylation through Ras/Raf/MEK/ERK signaling cascade as demonstrated by Western blotting analysis. IR/MWE also caused an inhibitory effect on Plk1 and the subsequent downstream regulator RhoA repression and Cep55 induction, which would influence cell cycle progression in the early steps of cytokinesis. A profound tumor growth suppression and inactivation of Aurora B activity in the tumor tissues by IR/MWE treatment were confirmed in the TSGH 8301 xenograft model in vivo. These data demonstrated that MWE could be an effective auxiliary to synergize with radiation on the anticancer efficacy by promoting mitotic catastrophe through inhibition of Aurora B, providing a novel and effective therapeutic option for bladder cancer management.
Subject(s)
Aurora Kinase B/antagonists & inhibitors , Fruit/chemistry , Mitosis/drug effects , Morus/chemistry , Plant Extracts/pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/radiotherapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G2 Phase/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Radiation, Ionizing , Signal Transduction/drug effects , Water/chemistryABSTRACT
Paclitaxel is a mitotic inhibitor used in cancer chemotherapy. Mulberry fruit is rich in phenolic compounds and flavonoids and exhibits chemopreventive activities. In this study, mulberry water extract (MWE) was used as a supplement to synergize with the effects of paclitaxel in the treatment of the TSGH 8301 human bladder cancer cell line. Treatment with paclitaxel combined with MWE (paclitaxel/MWE) enhanced the cytotoxicity of paclitaxel and induced severe G2/M arrest, mitotic catastrophe and subsequent apoptosis, as shown by MTT assay, HE staining and flow cytometry analyses. Differences in the expression and activation of Aurora A and Plk1 between cells treated with paclitaxel/MWE and paclitaxel alone suggested that the combined treatment caused a defect in the early steps of cytokinesis. Paclitaxel/MWE decreased EEA1 immunofluorescence staining and increased the expression of PTEN, indicating that the regimen inhibited the formation of the recycling endosome, which is required for cytokinesis. Paclitaxel/MWE also retarded tumor growth in a TSGH 8301 xenograft model via activation of PTEN and Caspase 3. These data demonstrated a synergistic effect on the anticancer efficacy of paclitaxel through MWE supplementation by promoting mitotic catastrophe through the activation of PTEN, providing a novel and effective therapeutic option for bladder cancer treatment strategies.
Subject(s)
Antineoplastic Agents/administration & dosage , Morus/chemistry , PTEN Phosphohydrolase/metabolism , Paclitaxel/administration & dosage , Plant Extracts/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mitosis/drug effects , Paclitaxel/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Urinary Bladder Neoplasms/metabolism , Xenograft Model Antitumor AssaysABSTRACT
PURPOSE: Wasabia japonica (wasabi) has been shown to exhibit properties of detoxification, anti-inflammation and the induction of apoptosis in cancer cells. This study aimed to investigate the molecular mechanism of the cytotoxicity of wasabi extract (WE) in colon cancer cells to evaluate the potential of wasabi as a functional food for chemoprevention. METHODS: Colo 205 cells were treated with different doses of WE, and the cytotoxicity was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide. Apoptosis and autophagy were detected by 4',6-diamidino-2-phenylindole, 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-imidacarbo-yanine iodide and staining for acidic vascular organelles (AVOs), along with Western blotting. RESULTS: The results demonstrated that WE induced the extrinsic pathway and mitochondrial death machinery through the activation of TNF-α, Fas-L, caspases, truncated Bid and cytochrome C. WE also induced autophagy by decreasing the phosphorylation of Akt and mTOR and promoting the expression of microtubule-associated protein 1 light chain 3-II and AVO formation. An in vivo xenograft model verified that tumor growth was delayed by WE treatment. CONCLUSION: Our studies revealed that WE exhibits anti-colon cancer properties through the induction of apoptosis and autophagy. These results provide support for the application of WE as a chemopreventive functional food and as a prospective treatment of colon cancer.
Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Colonic Neoplasms/drug therapy , Plant Extracts/pharmacology , Wasabia/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Colonic Neoplasms/pathology , Cytochromes c/genetics , Cytochromes c/metabolism , Humans , Indoles/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor AssaysABSTRACT
Hibiscus sabdariffa leaf has been previously shown to possess hypoglycemic, hypolipidemic, and antioxidant effects, and induce tumor cell apoptosis. However, the molecular mechanisms involved in the anticancer activity of H. sabdariffa leaf extract (HLE) are poorly understood. The object of the study was to examine the anti-invasive potential of HLE. First, HLE was demonstrated to be rich in polyphenols. The results of wound-healing assay and in vitro transwell assay revealed that HLE dose-dependently inhibited the migration and invasion of human prostate cancer LNCaP (lymph node carcinoma of the prostate) cells under non-cytotoxic concentrations. Our results further showed that HLE exerted an inhibitory effect on the activity and expressions of matrix metalloproteinase-9 (MMP-9). The HLE-inhibited MMP-9 expression appeared to be a consequence of nuclear factor-kappaB (NF-κB) inactivation because its DNA-binding activity was suppressed by HLE. Molecular data showed all these influences of HLE might be mediated via inhibition of protein kinase B (PKB, also known as Akt)/NF-kB/MMP-9 cascade pathway, as demonstrated by the transfection of Akt1 overexpression vector. Finally, the inhibitory effect of HLE was proven by its inhibition on the growth of LNCaP cells and the expressions of metastasis-related molecular proteins in vivo. These findings suggested that the inhibition of MMP-9 expression by HLE may act through the suppression of the Akt/NF-kB signaling pathway, which in turn led to the reduced invasiveness of the cancer cells.
Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Hibiscus/chemistry , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Plant Extracts/pharmacology , Plant Leaves/chemistry , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis/drug effects , Carcinoma/metabolism , Carcinoma/pathology , Cell Movement/drug effects , Down-Regulation , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Signal Transduction/drug effectsABSTRACT
Melanoma is the least common but most fatal form of skin cancer. Previous studies have indicated that an aqueous extract of Hibiscus sabdariffa leaves possess hypoglycemic, hypolipidemic, and antioxidant effects. In this study, we want to investigate the anticancer activity of Hibiscus leaf polyphenolic (HLP) extract in melanoma cells. First, HLP was exhibited to be rich in epicatechin gallate (ECG) and other polyphenols. Apoptotic and autophagic activities of HLP and ECG were further evaluated by DAPI stain, cell-cycle analysis, and acidic vascular organelle (AVO) stain. Our results revealed that both HLP and ECG induced the caspases cleavages, Bcl-2 family proteins regulation, and Fas/FasL activation in A375 cells. In addition, we also revealed that the cells presented AVO-positive after HLP treatments. HLP could increase the expressions of autophagy-related proteins autophagy-related gene 5 (ATG5), Beclin1, and light chain 3-II (LC3-II), and induce autophagic cell death in A375 cells. These data indicated that the anticancer effect of HLP, partly contributed by ECG, in A375 cells. HLP potentially could be developed as an antimelanoma agent.
Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Antioxidants/therapeutic use , Hibiscus/chemistry , Melanoma/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Polyphenols/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5 , Beclin-1 , Caspases/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/therapeutic use , Cell Line, Tumor , Humans , Melanoma/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Plant Extracts/pharmacology , Plant Leaves/chemistry , Polyphenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , fas Receptor/metabolismABSTRACT
Extracts of Piper betle leaf (PBLs) are rich in bioactive compounds with potential chemopreventive ability. In this study, Hep3B cells which are p53 null were used to investigate the anti-tumor effect of PBLs in the cell and in the xenograft model. The results revealed that PBLs (0.1 to 1 mg mL(-1)) induced a dose- and time-dependent increase of cell toxicity. The underlying mechanisms as evidenced by flow cytometry and western blot analysis showed that PBLs triggered ATM, cAbl, and p73 expressions and activated JNK and p38 pathways that subsequently led to cell cycle arrest and mitochondria-dependent apoptosis. PBLs also inhibited tumor growth in Hep3B-bearing mice via inducing the MAPK-p73 pathway. Our results demonstrated the in vitro and in vivo anti-tumor potential of PBLs, supporting their application as a novel chemopreventive agent for the treatment of human hepatocellular carcinoma (HCC) in the future via targeting the p73 pathway.
Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , DNA-Binding Proteins/metabolism , Drugs, Chinese Herbal/administration & dosage , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Nuclear Proteins/metabolism , Piper betle/chemistry , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/genetics , Tumor Protein p73 , Tumor Suppressor Proteins/geneticsABSTRACT
Piper betel leaf (PBL) has the biological capabilities of detoxification and can work as an anti-inflammatory agent and an anti-oxidant. In this study, we evaluated the anti-oxidative activity of the extract of Piper betel leaves (PBLs) on the basis of Cu(2+)-mediated oxidation, and its ability to prevent foam cell formation in a model for oxidised low density lipoprotein (oxLDL)-induced lipid accumulation in macrophages. Our data demonstrated that PBLs were able to inhibit LDL oxidation in vitro and are able to reduce the lipid accumulation in macrophages. We showed the underlying mechanisms to be the following: PBLs up-regulated the protein levels of the class A and class B scavenger receptors, the membrane lipid transporter ABCA1, and its upstream regulator Liver X receptor (LXR) in the macrophages exposed to oxLDL. The results suggested that PBLs activated the reverse cholesterol transport mechanism to enhance the metabolism of the oxLDL that could prevent both lipid accumulation and foam cell formation and further minimise the possible damage of vessels caused by the oxLDL.
Subject(s)
Cholesterol/metabolism , Lipid Metabolism/drug effects , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Piper/chemistry , Plant Extracts/pharmacology , Animals , Biological Transport/drug effects , Copper/metabolism , Foam Cells/drug effects , Foam Cells/metabolism , Macrophages/drug effects , Mice , Oxidation-Reduction , Plant Extracts/chemistry , Plant Leaves/chemistryABSTRACT
Many recent efforts have focused on targeting cell death pathways for discovering new cancer therapies. The relative resistance of liver cancer cells to ionizing radiation (IR) and chemotherapeutic agents due to autophagic response limits the available treatment options for this type of cancer. In this study, 3-methyladenine (3-MA), an autophagy inhibitor, was investigated for its potential to enhance radio-sensitivity under radio-resistant conditions both in vitro and in vivo. Hep3B and HepG2 cells were used to examine the radio-resistance of liver cancer cells. The results show that Hep3B cells respond to irradiation with increased apoptotic cell death and that HepG2 is radio-resistant due to the IR-induced autophagy, as verified by DNA fragmentation, electron microscopy, acidic vesicular organelle formation, and Western blot analysis. Application of IR with 3-MA to inhibit autophagy simultaneously suppressed the expression of LC3 and enhanced cell death. The tumor xenograft model in nude mice verified the synergistic cytotoxic effect of 3-MA and IR, which resulted in significant repression of tumor growth. The results demonstrate that IR-induced autophagy provides a self-protective mechanism against radiotherapy in HepG2 cells. In addition, 3-MA enhances the cytotoxicity of IR in cell models and suppresses tumor growth in animal models. Based on the results, application of 3-MA, or other autophagy inhibitors, could be used as an adjuvant for radiotherapy when radio-resistance develops as a result of autophagy response.
Subject(s)
Adenine/analogs & derivatives , Liver Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Adenine/pharmacology , Adenine/therapeutic use , Animals , Apoptosis/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/therapeutic use , Xenograft Model Antitumor AssaysABSTRACT
Andrographolide (Andro), a diterpenoid lactone isolated from a traditional herbal medicine Andrographis paniculata, is known to possess multiple pharmacological activities. In our previous study, Andro had been shown to inhibit non-small cell lung cancer (NSCLC) A549 cell migration and invasion via down-regulation of phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Here we demonstrated that Andro inhibited the expression of hypoxia-inducible factor-1α (HIF-1α) in A549 cells. HIF-1α plays an important role in tumor growth, angiogenesis and lymph node metastasis of NSCLC. The Andro-induced decrease of cellular protein level of HIF-1α was correlated with a rapid ubiquitin-dependent degradation of HIF-1α, and was accompanied by increased expressions of hydroxyl-HIF-1α and prolyl hydroxylase (PHD2), and a later decrease of vascular endothelial growth factor (VEGF) upon the treatment of Andro. The Andro-inhibited VEGF expression appeared to be a consequence of HIF-1α inactivation, because its DNA binding activity was suppressed by Andro. Molecular data showed that all these effects of Andro might be mediated via TGFß1/PHD2/HIF-1α pathway, as demonstrated by the transfection of TGFß1 overexpression vector and PHD2 siRNA, and the usage of a pharmacological MG132 inhibitor. Furthermore, we elucidated the involvement of Andro in HIF-1α transduced VEGF expression in A549 cells and other NSCLC cell lines. In conclusion, these results highlighted the potential effects of Andro, which may be developed as a chemotherapeutic or an anti-angiogenesis agent for NSCLC in the future.
Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Diterpenes/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Lung Neoplasms/metabolism , Cell Line, Tumor , Humans , Procollagen-Proline Dioxygenase/physiology , Signal Transduction , Transforming Growth Factor beta1/physiology , Vascular Endothelial Growth Factor A/biosynthesisABSTRACT
The objective of this study was to investigate the lipid-lowering effects of mulberry water extracts (MWEs). To evaluate the hypolipidemic effect of MWEs, hamsters were fed with either high fat/cholesterol diets (HFCD) or HFCD supplemented with 1 and 2% MWEs for 12 weeks. Plasma total cholesterol (TC) and triglyceride (TG) levels of hamsters fed HFCD with MWEs were significantly reduced by about 30-37% and 16-35%, respectively, as compared to those without MWEs. Similar results were also measured in hepatic TC and TG of hamsters fed HFCD with MWEs. Low-density lipoprotein receptor (LDLR) gene expression and the uptake ability of low-density lipoprotein (LDL) in HepG2 cells were also upregulated by additions of MWEs. MWEs also decreased the gene expressions of enzymes involved in the TG and TC biosyntheses. Results suggest that hypolipidemic effects of MWEs are via an enhancement of LDLR gene expression and the clearance ability of LDL and a decrease in the lipid biosynthesis. Therefore, MWEs could be used as a natural agent against hyperlipidemia.
Subject(s)
Lipid Metabolism/drug effects , Morus/chemistry , Plant Extracts/pharmacology , Animals , Cricetinae , Gene Expression/drug effects , Hep G2 Cells , Homeostasis/drug effects , Lipids/blood , Liver/drug effects , Liver/metabolism , Male , Mesocricetus , Receptors, LDL/genetics , Receptors, LDL/metabolismABSTRACT
PURPOSE: Radiotherapy is the most efficacious strategies for lung cancer. The radiation-enhancing effects and the underlying mechanisms of berberine were investigated both in vitro and in vivo. METHODS AND MATERIALS: Clonogenic survival assays were used to evaluate the radio-sensitivity of berberine on non-small-cell lung cancer. Electron microscopic observation of the features of cell death, flow cytometry of acidic vascular organelles formation, mitochondria membrane potential and cell-cycle progression, and Western blotting of caspase 3, PARP, and LC3 were performed to identify the mechanisms underlying the enhancing effects. Lewis lung carcinoma model in mice was conducted to evaluate the possible application of berberine in synergistic treatment with irradiation. RESULTS: Compared with radiation alone (SF2 = 0.423; D(0) = 5.29 Gy), berberine at 5 and 10 muM concentrations in combination with radiation showed significant enhancement on radiation-induced clonogenic inhibition (SF2 = 0.215: D(0) = 2.70 Gy and SF2 = 0.099: D(0) = 1.24 Gy) on A549 cells. The cellular ultrastructure showed the presence of autophagosome and an increased proportion of acridine orange stain-positive cells, demonstrating that berberine enhanced radiosensitivity via autophagy. The process involved LC3 modification and mitochondrial disruption. The animal model verified the synergistic cytotoxic effect of berberine and irradiation resulting in a substantial shrinkage of tumor volume. CONCLUSION: Supplement of berberine enhanced the cytotoxicity of radiation in both in vivo and in vitro models of lung cancer. The mechanisms underlying this synergistic effect involved the induction of autophagy. It suggests that berberine could be used as adjuvant therapy to treat lung cancer.
Subject(s)
Autophagy/drug effects , Berberine/therapeutic use , Carcinoma, Lewis Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Acridine Orange , Animals , Autophagy/physiology , Autophagy/radiation effects , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Collagen Type XI/metabolism , Combined Modality Therapy/methods , Drug Screening Assays, Antitumor , Flow Cytometry , Fluorescent Dyes , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mice , Mice, Inbred C57BL , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , Organelles/drug effects , Radiation Tolerance/drug effectsABSTRACT
Solanum nigrum L. (SN) is an herbal plant that has been used as hepatoprotective and anti-inflammation agent in Chinese medicine. In this study, the protective effects of water extract of SN (SNE) against liver damage were evaluated in carbon tetrachloride (CCl4)-induced chronic hepatotoxicity in rats. Sprague-Dawley (SD) rats were orally fed with SNE (0.2, 0.5, and 1.0 g kg(-1) bw) along with administration of CCl4 (20% CCl4/corn oil; 0.5 mL kg(-1) bw) for 6 weeks. The results showed that the treatment of SNE significantly lowered the CCl4-induced serum levels of hepatic enzyme markers (GOT, GPT, ALP, and total bilirubin), superoxide and hydroxyl radical. The hepatic content of GSH, and activities and expressions of SOD, GST Al, and GST Mu that were reduced by CCl4 were brought back to control levels by the supplement of SNE. Liver histopathology showed that SNE reduced the incidence of liver lesions including hepatic cells cloudy swelling, lymphocytes infiltration, hepatic necrosis, and fibrous connective tissue proliferation induced by CCl4 in rats. Therefore, the results of this study suggest that SNE could protect liver against the CCl4-induced oxidative damage in rats, and this hepatoprotective effect might be contributed to its modulation on detoxification enzymes and its antioxidant and free radical scavenger effects.
Subject(s)
Carbon Tetrachloride , Liver Cirrhosis/prevention & control , Liver/drug effects , Phytotherapy/methods , Solanum nigrum/chemistry , Administration, Oral , Animals , Antioxidants/administration & dosage , Antioxidants/isolation & purification , Biomarkers/blood , Body Weight/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Organ Size/drug effects , Oxidation-Reduction/drug effects , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/bloodABSTRACT
Solanum nigrum L. (SN) has been used in traditional folk medicine to treat different cancers. It is also used as a hepatoprotective and anti-inflammatory agent. In this study, we demonstrated that the extract of SN (SNE) induced a strong cytotoxic effect toward HepG2 cells but much less to Chang liver and WRL-68 cells. The mechanisms of the cytotoxic effect were concentration-dependent. High doses of SNE (2 and 5 mg/mL) induced apoptotic cell death in HepG2 cells, as evidenced by increases in the expressions of p-JNK and Bax, mitochodrial release of cytochrome c, and caspase activation. On the other hand, cells treated with low concentrations of SNE (50-1000 microg/mL) revealed morphological and ultrastructural changes of autophagocytic death under electron microscopic observation. Furthermore, these cells showed increased levels of autophagic vacuoles and LC3-I and LC3-II proteins, specific markers of autophagy. The levels of Bcl-2 and Akt that have been implicated in the down-regulation of autophagy were decreased upon SNE treatment. Taken together, these findings indicate that SNE induced cell death in hepatoma cells via two distinct antineoplastic activities of SNE, the ability to induce apoptosis and autophagocytosis, therefore suggesting that it may provide leverage to treat liver cancer.
Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Plant Extracts/pharmacology , Solanum nigrum/chemistry , Caspase 3/metabolism , Cell Line , Cell Line, Tumor , Cytochromes c/metabolism , Fetus , Humans , Liver , Liver Neoplasms , Stomach NeoplasmsABSTRACT
Magnolol, a compound extracted from the Chinese medicinal herb Magnolia officinalis, has several biological effects. However, its protective effects against endothelial injury remain unclear. In this study, we examined whether magnolol prevents oxidized low density lipoprotein (oxLDL)-induced vascular endothelial apoptosis. Incubation of oxLDL with magnolol (2.5-20 microM) inhibited copper-induced oxidative modification via diene formation, thiobarbituric acid reactive substances (TBARS) assay and electrophoretic mobility assay. Apoptotic cell death as characterized by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) stain. We measured the production of reactive oxygen species (ROS) by using the fluorescent probe 2',7'-dichlorofluorescein acetoxymethyl ester (DCF-AM), and observed the activity of antioxidant enzymes. Furthermore, several apoptotic signaling pathways which showed NF-kappaB activation, increased cytosolic calcium, alteration of mitochondrial membrane potential, cytochrome c release and activation of caspase 3 were also investigated. We demonstrated that magnolol prevented the copper-induced oxidative modification of LDL. Magnolol attenuated the oxLDL-induced ROS generation and subsequent NF-kappaB activation. Furthermore, intracellular calcium accumulation and subsequent mitochondrial membrane potential collapse, cytochome c release and activation of caspase 3 caused by oxLDL were also inhibited by magnolol. Our results suggest that magnolol may have clinical implications in the prevention of atherosclerotic vascular disease through decreasing the oxLDL-induced ROS production.
Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Endothelial Cells/drug effects , Lignans/pharmacology , Lipoproteins, LDL/metabolism , Calcium/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Copper Sulfate/chemistry , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Lipid Peroxidation/drug effects , Lipoproteins, LDL/chemistry , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , NF-kappa B/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Time FactorsABSTRACT
Piper betel leaves (PBL) are used in Chinese folk medicine for the treatment of various disorders. PBL has the biological capabilities of detoxication, antioxidation, and antimutation. In this study, we evaluated the antihepatotoxic effect of PBL extract on the carbon tetrachloride (CCl(4))-induced liver injury in a rat model. Fibrosis and hepatic damage, as reveled by histology and the activities of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were induced in rats by an administration of CCl(4) (8%, 1 ml/kg body weight) thrice a week for 4 weeks. PBL extract significantly inhibited the elevated AST and ALT activities caused by CCl(4) intoxication. It also attenuated total glutathione S-transferase (GST) activity and GST alpha isoform activity, and on the other hand, enhanced superoxide dismutase (SOD) and catalase (CAT) activities. The histological examination showed the PBL extract protected liver from the damage induced by CCl(4) by decreasing alpha-smooth muscle actin (alpha-sma) expression, inducing active matrix metalloproteinase-2 (MMP2) expression though Ras/Erk pathway, and inhibiting TIMP2 level that consequently attenuated the fibrosis of liver. The data of this study support a chemopreventive potential of PBL against liver fibrosis.
Subject(s)
Carbon Tetrachloride/toxicity , Drugs, Chinese Herbal/therapeutic use , Liver Cirrhosis, Experimental/prevention & control , Piper betle/chemistry , Plant Leaves/chemistry , Actins/metabolism , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Blotting, Western , Body Weight/drug effects , Carbon Tetrachloride/administration & dosage , Catalase/metabolism , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Glutathione Transferase/metabolism , Hydroxyl Radical/metabolism , Injections, Intraperitoneal , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/drug therapy , Male , Matrix Metalloproteinase 2/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Phytotherapy , Rats , Rats, Wistar , Superoxide Dismutase/metabolismABSTRACT
Eugenol, a natural constituent of a number of aromatic plants and their essential oil fractions, has several biological effects. However, its protective effects against endothelial injury remain unclarified. This study investigates how eugenol affects human umbilical vein endothelial cells (HUVECs) dysfunction mediated by oxidized low density lipoprotein (oxLDL). Our results showed that the suppression of endothelial NO synthase (eNOS) expression, enhancement of adhesion molecules (ICAM, VCAM, and E-selectin) expression, and adherence of monocytic THP1 cells caused by a non-cytotoxic concentration (100 microg/ml) of oxLDL were ameliorated following a eugenol treatment (12.5-100 microM) in HUVECs. Eugneol also inhibited the reactive oxygen species (ROS) generation, intracellular calcium accumulation, and the subsequent mitochondrial membrane potential collapse, cytochrome c release and caspase-3 activation induced by oxLDL. The cytotoxicity and apoptotic features induced by a cytotoxic concentration (200 microg/ml) of oxLDL was also attenuated by eugenol. Our results suggest that eugenol may protect against the oxLDL-induced dysfunction in endothelial cells.
Subject(s)
Antioxidants/pharmacology , Cell Adhesion Molecules/metabolism , Cell Survival/drug effects , Endothelium, Vascular/drug effects , Eugenol/pharmacology , Lipoproteins, LDL/pharmacology , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Nitric Oxide Synthase Type III/metabolism , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Umbilical Cord/cytologyABSTRACT
Honokiol, a compound extracted from Chinese medicinal herb Magnolia officinalis, has several biological effects. However, its protective effects against endothelial injury remain unclarified. In this study, we examined whether honokiol prevented oxidized low-density lipoprotein (oxLDL)-induced vascular endothelial dysfunction. Incubation of oxLDL with honokiol (2.5-20 microM) inhibited copper-induced oxidative modification as demonstrated by diene formation, thiobarbituric acid reactive substances (TBARS) assay and electrophoretic mobility assay. Expression of adhesion molecules (ICAM, VCAM and E-selectin) and endothelial NO synthase (eNOS) affected by oxLDL was investigated by flow cytometry and Western blot. We also measured the production of reactive oxygen species (ROS) using the fluorescent probe 2',7'-dichlorofluorescein acetoxymethyl ester (DCF-AM). Furthermore, several apoptotic phenomena including increased cytosolic calcium, alteration of mitochondrial membrane potential, cytochrome c release and activation of caspase-3 were also investigated. Apoptotic cell death was characterized by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) stain. The results showed that honokiol prevented the copper-induced oxidative modification of LDL. Honokiol also ameliorated the oxLDL-diminished eNOS protein expression and reduced the oxLDL-induced adhesion molecules and the adherence of THP-1 cells to HUVECs. Furthermore, honokiol attenuated the oxLDL-induced cytotoxicity, apoptotic features, ROS generation, intracellular calcium accumulation and the subsequent mitochondrial membrane potential collapse, cytochrome c release and activation of caspase-3. Our results suggest that honokiol may have clinical implications in the prevention of atherosclerotic vascular disease.
Subject(s)
Antioxidants/pharmacology , Biphenyl Compounds/pharmacology , Cell Adhesion/drug effects , Endothelial Cells/drug effects , Lignans/pharmacology , Lipoproteins, LDL/pharmacology , Apoptosis , Calcium/metabolism , Caspase 3 , Caspases/metabolism , Cell Line , Cytochromes c/metabolism , E-Selectin/metabolism , Endothelial Cells/physiology , Humans , Intercellular Adhesion Molecule-1/metabolism , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/metabolismABSTRACT
Berberine, a compound isolated from medicinal herbs, has been reported with many pharmacological effects related to anti-cancer and anti-inflammation capabilities. In this study, we observed that berberine exerted a dose- and time-dependent inhibitory effect on the motility and invasion ability of a highly metastatic A549 cells under non-cytotoxic concentrations. In cancer cell migration and invasion process, matrix-degrading proteinases are required. A549 cell treated with berberine at various concentrations showed reduced ECM proteinases including matrix metalloproteinase-2 (MMP2) and urokinase-plasminogen activator (u-PA) by gelatin and casein zymography analysis. The inhibitory effect is likely to be at the transcriptional level, since the reduction in the transcripts levels was corresponding to the proteins. Moreover, berberine also exerted its action via regulating tissue inhibitor of metalloproteinase-2 (TIMP-2) and urokinase-plasminogen activator inhibitor (PAI). The upstream mediators of the effect involved c-jun, c-fos and NF-kappaB, as evidenced by reduced phosphorylation of the proteins. These findings suggest that berberine possesses an anti-metastatic effect in non-small lung cancer cell and may, therefore, be helpful in clinical treatment.
Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Berberine/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Matrix Metalloproteinase Inhibitors , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Humans , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Urokinase-Type Plasminogen Activator/biosynthesisABSTRACT
Piper betel leaves (PBL) are used in Chinese folk medicine for the treatment of various disorders. PBL has the biological capabilities of de-toxication, anti-oxidation and anti-mutation. In this study we first examined the effect of PBL extract on the activity of Glutathione S-transferase (GST) isoforms, and found that it inhibited total GST and the alpha class of GST (GSTA), but not the pi class of GST (GSTP), and the mu class of GST (GSTM), activity in Hep G2 cells. RT-PCR results verified a reduction in the expression of GSTA1. Next, we examined whether PBL extract could increase the sensitivity of Hep G2 cells to anti-cancer drugs. The data showed that the cytotoxicity of cisplatin was significantly enhanced by the presence of PBL extract, accompanied by a reduction in the expression of multidrug resistance protein 2 (MRP2). These effects of PBL extract were compared to its major constitute, eugenol. Although eugenol decreased MRP2 level more effectively than PBL extract, it exhibited less sensitizing effect. In conclusion, we demonstrated that PBL extract was able to increase the sensitivity of Hep G2 cells to cisplatin via at least two mechanisms, reducing the expression of MRP2 and inhibiting the activity of total GST and the expression of GSTA. The data of this study support an application of PBL as an additive to reduce drug resistance.
Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , Hepatocytes/drug effects , Piper betle , Plant Extracts/pharmacology , Cell Fractionation , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Drugs, Chinese Herbal , Eugenol/pharmacology , G2 Phase , Glutathione Transferase/metabolism , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Isoenzymes , Liver Neoplasms/drug therapy , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Organoplatinum Compounds/pharmacology , Oxaliplatin , Plant Leaves/chemistryABSTRACT
Baicalin (BA) is a flavonoid compound purified from Scutellaria baicalensis Georgi that is used as a traditional Chinese herbal medicine. Baicalin was studied for the mechanism of its inhibitory effects on the tert-butyl hydroperoxide (t-BHP)-induced cytotoxicity and lipid peroxidation in rat liver system. Baicalin expressed an antioxidant property by its capacity for quenching the free radicals of 1,1-diphenyl-2-picrylhydrazyl (DPPH). Further investigations using the t-BHP-induced cytotoxicity in rat primary hepatocytes demonstrated that baicalin, at the concentrations of 2-220 microM, significantly decreased the leakages of lactate dehydrogenase (LDH) and alanine aminotransferase (ALT), and the formation of malondialdehyde (MDA) induced by 30 min treatment of t-BHP(1.5 mM). Baicalin also attenuated the t-BHP-induced depletion of glutathione (GSH) and high level of DNA repaired synthesis. An in vivo study in rats showed that pretreatment with baicalin (i.p.) at concentrations of 0.5 and 5 mg/kg for 5 days before a single i.p. dose of t-BHP (0.1 mmol/kg) significantly lowered the serum levels of hepatic enzyme markers (ALT and AST) and reduced oxidative stress in the liver. Histopathological evaluation of the rat livers revealed that baicalin reduced the incidence of liver lesions induced by t-BHP including hepatocyte swelling, leukocyte infiltration, and necrosis. Based on the results described above, we speculate that baicalin may play a chemopreventive role via reducing oxidative stress in living systems.