Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Environ Toxicol ; 37(11): 2718-2727, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35917206

ABSTRACT

Colorectal cancer is the third leading cause of cancer death in Taiwan. Current treatments involve combination of surgical resection, radiation, and chemotherapy. These treatments have demonstrated to increased five-year survival of a patient with colorectal cancer. However, metastasis is a major capability of cancer cells that causes poor prognosis, recurrence, and even death. Epidemiological and clinical studies have suggested the use of non-steroidal anti-inflammatory drugs (NSAIDs) as an effective class of compounds to prevent colon cancer. Parecoxib is an NSAID and the only parenterally administered selective cyclooxygenase (COX)-2 inhibitor. In this study, we evaluated whether parecoxib inhibits the metastasis of DLD-1 human colon cancer cells, a COX-2 null cell line, and the underlying mechanism. Cell migration of the DLD-1 cells was significantly inhibited by parecoxib treatment as shown by the Transwell migration assay. This enhanced anti-migration effect was correlated with the attenuated phosphorylation of Akt, expression of vimentin (a mesenchymal marker), and ß-catenin, and corresponded with the upregulated GSK3ß and E-cadherin (an epithelial marker). These findings suggested that parecoxib could inhibit the epithelial-mesenchymal transition (EMT) and metastasis in human colon cancer cells by downregulating ß-catenin. Thus, parecoxib could provide a novel prospective strategy for a combination treatment with chemotherapeutic drugs against metastasis of human colon cancer.


Subject(s)
Colonic Neoplasms , Epithelial-Mesenchymal Transition , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal , Cadherins/metabolism , Cell Line , Cell Line, Tumor , Cell Movement , Colonic Neoplasms/pathology , Cyclooxygenase 2/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Isoxazoles , Proto-Oncogene Proteins c-akt/metabolism , Vimentin/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
2.
Oncol Lett ; 15(4): 5915-5923, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29552223

ABSTRACT

Natural compounds have been candidates for anticancer medicine over the last 20 years. During the process of isolating seed oil from Calophyllum inophyllum L., yellow and green pigments containing multiple compounds with an aromatic structure were identified. High-performance liquid chromatography and nuclear magnetic resonance analysis of these pigments revealed that the compounds present were identical, but the concentration of the compounds was different. Treatment with the pigments was able to induce the death of DLD-1 human colon cancer cells and increase the percentage of the cells in the sub-G1 and sub-G2/M phases in a dose-dependent manner. Additionally, the pigments were able to exhibit cytotoxic activity on A549 and H1975 human non-small cell lung cancer (NSCLC) cell lines at 24 h, with half-maximal inhibitory concentrations (IC50) values of 0.1206 and 0.0676%, respectively for green pigments, and 0.0434 and 0.0501%, respectively for yellow pigments. Furthermore, a decrease in IC50 value was associated with an increase in the duration of treatment. However, a sharp decrease in IC50 value of the yellow pigment was observed for H1975 cells at 48 h and for A549 cells at 72 h compared with no change in IC50 value for the green pigment with time, suggesting that the pigments function and induce cell death differently in the two cell lines. An investigation was performed into the synergistic effect of the green pigment and gefitinib (Iressa®, ZD1839), which is a selective epidermal growth factor receptor-tyrosine kinase inhibitor to block growth factor-mediated cell proliferation. The combination of the green pigment and gefitinib resulted in an enhancement of the decrease in viability of A549 and H1975 cells compared with treatment with gefitinib alone, which suggested that treatment with the green pigments was able to enhance the sensitivity of NSCLC cells to gefitinib. In conclusion, these pigments may be considered for development as anti-colon cancer agents.

3.
Int Ophthalmol ; 38(2): 747-756, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28393322

ABSTRACT

AIM: This study was to investigate the anti-angiogenic effect of hexahydrocurcumin (HHC) to evaluate gene (p-basic fibroblast growth factor (bFGF)-SAINT-18 & p-vascular endothelial growth factor (VEGF)-SAINT-18 complex)-induced corneal neovascularization (CorNV) in rats. METHODS: CorNV was induced in 24 eyes of 24 rats. Four groups (Group A: 0 µg, B: 0.01 µg, C: 0.1 µg, and D: 1 µg) of HHC were prepared and implanted into the rat subconjunctival substantia propria 1.5 mm from the limbus at temporal side. The 1 µg of p-bFGF-SAINT-18 & p-VEGF-SAINT-18 complex were prepared and implanted into the rat corneal stroma 1.5 mm from the limbus at the same side. Inhibition of CorNV was observed and quantified from day 1 to day 60. bFGF and VEGF protein expression were analyzed by biomicroscopic examination, western blot analysis, and immunohistochemistry. RESULTS: Subconjunctival injection by 1 µg HHC successfully inhibited gene-induced CorNV in rats. bFGF and VEGF protein expression were reduced after 6 days. Meanwhile, the reduction of HLA-DR expression was detected. CONCLUSIONS: Our study showed that the HHC might provide an important anti-angiogenesis factor to inhibit CorNV development at the corneal experimental angiogenesis model.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Corneal Neovascularization/drug therapy , Curcumin/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Animals , Blotting, Western , Conjunctiva/drug effects , Conjunctiva/metabolism , Corneal Neovascularization/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Disease Models, Animal , Fibroblast Growth Factor 2/metabolism , HLA-DR Antigens/metabolism , Male , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
4.
J Immunol Res ; 2017: 9489383, 2017.
Article in English | MEDLINE | ID: mdl-29062841

ABSTRACT

In this study, we demonstrated that temozolomide (TMZ) and propyl gallate (PG) combination enhanced the inhibition of migration in human U87MG glioma cells. PG inhibited the TMZ-induced reactive oxygen species (ROS) generation. The mitochondrial complex III and NADPH oxidase are two critical sites that can be considered to regulate antimigration in TMZ-treated U87MG cells. PG can enhance the antimigration effect of TMZ through suppression of metalloproteinase-2 and metalloproteinase-9 activities, ROS generation, and the NF-κB pathway and possibly provide a novel prospective strategy for treating malignant glioma.


Subject(s)
Dacarbazine/analogs & derivatives , Glioma/drug therapy , Propyl Gallate/pharmacology , Apoptosis , Cell Line, Tumor , Cell Movement/drug effects , Dacarbazine/pharmacology , Drug Synergism , Drug Therapy, Combination , Glioma/pathology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Temozolomide
5.
PLoS One ; 12(10): e0186214, 2017.
Article in English | MEDLINE | ID: mdl-29016672

ABSTRACT

The bladder is an important organ for the storage of excreted water and metabolites. If metabolites with carcinogenic characteristics are present in urine, the urothelial lining of the bladder could be damaged and genetically altered. In this study, we analyzed the interaction of arsenic and N-butyl-N-(4-hydroxybutyl)nitrosamine (BBN) on mouse bladder carcinogenesis. Our previous study found that arsenic affects BBN-altered urothelial enzymatic activity, protein expression, DNA oxidation and global DNA CpG methylation levels. In this study, two mouse models were used. First, after administering a co-treatment of BBN and arsenic for 20 weeks, BBN alone led to a urothelial carcinoma formation of 20%, and arsenic promoted a BBN-induced urothelial carcinoma formation of 10%. The protein expression of GSTM1, GSTO1, NQO1, and p21 did not change by arsenic along with the BBN co-treatment, but the Sp1 expression increased. In the second mouse model, BBN was a pretreatment promoter; arsenic dose-dependently deteriorated BBN-promoted dysplasia by 10% and 40% at 10 ppm and 100 ppm, respectively. Conversely, BBN pretreatment also accelerated arsenic-induced dysplasia by 30%. The urothelial carcinogenic effect reversed after ceasing BBN for a period of 20 weeks. In summary, three conclusions were drawn from this study. The first is the mutual promotion of arsenic and BBN in bladder carcinogenesis. Second, arsenic dosages without bladder carcinogenicity (10 ppm) or with slight carcinogenicity (100 ppm) promote BBN-induced mice bladder cancer progression. Finally, the dysplastic urothelium had reverted to near-normal morphology after ceasing BBN intake for 20 weeks, providing a good suggestion for people who want to quit smoking.


Subject(s)
Arsenic/toxicity , Butylhydroxybutylnitrosamine/toxicity , Carcinogenesis/chemically induced , Carcinogens/toxicity , Carcinoma, Transitional Cell/chemically induced , Urinary Bladder Neoplasms/chemically induced , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Interactions , Female , Gene Expression Regulation, Neoplastic , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Male , Mice , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Sp1 Transcription Factor/agonists , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urothelium/drug effects , Urothelium/metabolism , Urothelium/pathology
6.
Bull Environ Contam Toxicol ; 98(5): 612-618, 2017 May.
Article in English | MEDLINE | ID: mdl-28315003

ABSTRACT

The aim of the present study was to identify whether the responses of oxidative stress in zebrafish liver are similar to those in mammalians upon low doses of Cd2+ exposure in short durations. Fish were exposed to 1.78 µM Cd2+ (treatment) and 0.0 µM Cd2+ (control) for 0, 1, 3, and 6 h. The reactive oxygen species (ROS) and lipid peroxidation (LPO) of hepatic tissues significantly increased after 3 and 6 h of Cd2+ exposure, respectively. Antioxidants glutathione peroxidase (gpx1a), superoxide dismutase (sod), and catalase (cat) were up regulated after 1-3 h, and metallothionein isoforms (smtB and mt2) increased after 3-6 h of Cd2+ exposure. The caspase-3 and p53 mRNA expressions significantly increased threefolds after 1 h of Cd2+ exposure. Results confirmed that oxidative stress in the hepatic tissue was induced by Cd2+ within 3 h. However, anti-oxidative functions immediately up regulated, causing cell apoptosis levels to decrease after 6 h of Cd2+ exposure.


Subject(s)
Antioxidants/metabolism , Cadmium/toxicity , Liver/metabolism , Zebrafish/metabolism , Animals , Apoptosis/drug effects , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
7.
Biomed Rep ; 4(3): 349-354, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26998274

ABSTRACT

Resveratrol (Re), a stilbenoid, is associated with a potential benefit in controlling certain biomarkers in type II diabetes. Genistein (Ge), a phytoestrogen, may act as an antioxidant and thus may diminish damaging effects of free radicals in tissues. In the present study, a potential synergistic antioxidant effect of an Re/Ge combination on high-glucose (HG) incubation in Madin-Darby canine kidney (MDCK) epithelial cells was evaluated. Compared with the treatment of Re or Ge alone, the Re/Ge combination synergistically decreased intracellular reactive oxygen species (ROS) and hydroxyl radicals in MDCK cells. This synergistic antioxidant effect correlated with the inhibition of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression and an increase in γ-glutamylcysteine synthetase expression. In addition, mitochondrial complex I, NADPH oxidase, xanthine oxidase and lipoxygenase contributed towards ROS overproduction when the MDCK cells were incubated with HG. In conclusion, the Re/Ge combination synergistically enhanced the antioxidant effect in HG-incubated kidney cells, possibly through an enhanced antioxidant regulation mechanism. The Re/Ge combination may be a potential benefit against oxidative stress in diabetes mellitus.

8.
PLoS One ; 11(1): e0146440, 2016.
Article in English | MEDLINE | ID: mdl-26771387

ABSTRACT

Despite a plethora of literature has documented that osteoarthritis (OA) is veritably associated with oxidative stress-mediated chondrocyte death and matrix degradation, yet the possible involvement of synoviocyte abnormality as causative factor of OA has not been thoroughly investigated. For this reason, we conduct the current studies to insight into how synoviocytes could respond to an episode of folate-deprived (FD) condition. First, when HIG-82 synoviocytes were cultivated under FD condition, a time-dependent growth impediment was observed and the demise of these cells was demonstrated to be apoptotic in nature mediated through FD-evoked overproduction of reactive oxygen species (ROS) and drastically released of cytosolic calcium (Ca2+) concentrations. Next, we uncovered that FD-evoked ROS overproduction could only be strongly suppressed by either mitochondrial complex II inhibitors (TTFA and carboxin) or NADPH oxidase (NOX) inhibitors (AEBSF and apocynin), but not by mitochondrial complex I inhibitor (rotenone) and mitochondrial complex III inhibitor (antimycin A). Interestingly, this selective inhibition of FD-evoked ROS by mitochondrial complex II and NOX inhibitors was found to correlate excellently with the suppression of cytosolic Ca2+ release and reduced the magnitude of the apoptotic TUNEL-positive cells. Taken together, we present the first evidence here that FD-triggered ROS overproduction in synoviocytes is originated from mitochondrial complex II and NOX. Both elevated ROS in tandem with cytosolic Ca2+ overload serve as final arbitrators for apoptotic lethality of synoviocytes cultivated under FD condition. Thus, folate supplementation may be beneficial to patients with OA.


Subject(s)
Calcium/metabolism , Electron Transport Complex II/metabolism , Folic Acid Deficiency/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Carboxin/pharmacology , Cell Line , Electron Transport Complex II/antagonists & inhibitors , Folic Acid/metabolism , HeLa Cells , Humans , NADPH Oxidases/antagonists & inhibitors , Oxidation-Reduction/drug effects , Oxidative Stress/physiology , Rabbits , Rotenone/pharmacology , Sulfones/pharmacology , Thenoyltrifluoroacetone/pharmacology
9.
Article in English | MEDLINE | ID: mdl-26025641

ABSTRACT

The present study seeks to detect oxidative damage and to compare anti-oxidative responses among liver, gills and brain of adult zebrafish that were cooled from 28 °C (control) to 12 °C (treatment) for 0-24 h. The lipid peroxidation of liver, gill and brain tissues significantly increased at 1h after transfer, but reactive oxygen species in the treatment group increased significantly after 24 h as compared to the control. The fish were found to develop a cascading anti-oxidative mechanism beginning with an increase in Cu/Zn-SOD levels, followed by increased CAT and GPx mRNA expressions in the three tissue types. Both smtB and mt2 mRNAs increased in the hepatic and brain tissues following 1h of cold stress, but only smtB exhibited a significant increase in the gills at 1 h and 6 h after transfer to 12 °C. Furthermore, cellular apoptosis in the brain was not evident after cold shock, but liver and gills showed cellular apoptosis at 1-3 h, with another peak in the liver at 6 h after cold shock. The results suggest that the cold shock induced oxidative stress, and the enzymatic (SOD, GPx and CAT) and non-enzymatic (mt-2 and smt-B) mRNA expressions all play a role in the resulting anti-oxidation within 1-6 h of cold shock. A functional comparison showed that the brain had the most powerful antioxidant defense system of the three tissue types since it had the highest smtB mRNA expression and a lower level of cell apoptosis than the liver and gills after exposure to cold stress.


Subject(s)
Antioxidants/metabolism , Brain/physiology , Cold Temperature , Gills/physiology , Liver/physiology , Stress, Physiological , Zebrafish/physiology , Animals , Apoptosis , Lipid Peroxidation , Reactive Oxygen Species/metabolism
10.
PLoS One ; 9(4): e94180, 2014.
Article in English | MEDLINE | ID: mdl-24714453

ABSTRACT

Shikonin is a quinone-containing natural product that induces the apoptotic death of some cancer cell lines in culture through increasing intracellular reactive oxygen species (ROS). Quinone-based drugs have shown potential in the clinic, making shikonin an interesting compound to study. Our previous study found that shikonin induces apoptosis in neuroblastoma by induction of ROS, but its mechanism of action and scope of activity are unknown. In this study, we investigated the mode of oxidative stress of shikonin in human glioma cells. ROS induction by shikonin was of mitochondrial origin, as demonstrated by detection of superoxide with MitoSOX Red. Pre-incubation of shikonin with inhibitors of different complexes of the respiratory chain suggested that shikonin-induced ROS production occurred via complex II. In addition, NADPH oxidase and lipooxygenase are two other main ROS-generated sites in shikonin treatment. ROS production by shikonin resulted in the inhibition of nuclear translocation of Nrf2. Stable overexpression of Nrf2 in glioma cells inhibited ROS generation by shikonin. ROS generation from mitochondrial complex II, NADPH oxidase and lipooxygenase is likely the primary mechanism by which shikonin induces apoptosis in glioma cells. These findings also have relevance to the development of certain ROS producers as anti-cancer agents. These, along with shikonin have potential as novel chemotherapeutic agents on human glioma.


Subject(s)
Glioma/metabolism , Naphthoquinones/pharmacology , Oxidative Stress/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle , Cell Line, Tumor , Cytochromes c/metabolism , Cytosol/metabolism , Glioma/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism
11.
PLoS One ; 8(11): e77931, 2013.
Article in English | MEDLINE | ID: mdl-24223745

ABSTRACT

It has been postulated that folic acid (folate) deficiency (FD) may be a risk factor for the pathogenesis of a variety of oxidative stress-triggered chronic degenerative diseases including diabetes, however, the direct evidence to lend support to this hypothesis is scanty. For this reason, we set out to study if FD can trigger the apoptotic events in an insulin-producing pancreatic RINm5F islet ß cells. When these cells were cultivated under FD condition, a time-dependent growth impediment was observed and the demise of these cells was demonstrated to be apoptotic in nature proceeding through a mitochondria-dependent pathway. In addition to evoke oxidative stress, FD condition could also trigger nitrosative stress through a NF-κB-dependent iNOS-mediated overproduction of nitric oxide (NO). The latter compound could then trigger depletion of endoplasmic reticulum (ER) calcium (Ca(2+)) store leading to cytosolic Ca(2+) overload and caused ER stress as evidence by the activation of CHOP expression. Furthermore, FD-induced apoptosis of RINm5F cells was found to be correlated with a time-dependent depletion of intracellular glutathione (GSH) and a severe down-regulation of Bcl-2 expression. Along the same vein, we also demonstrated that FD could severely impede RINm5F cells to synthesize insulin and their abilities to secret insulin in response to glucose stimulation were appreciably hampered. Even more importantly, we found that folate replenishment could not restore the ability of RINm5F cells to resynthesize insulin. Taken together, our data provide strong evidence to support the hypothesis that FD is a legitimate risk factor for the pathogenesis of diabetes.


Subject(s)
Apoptosis , Folic Acid Deficiency/metabolism , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Oxidative Stress , Animals , Calcium/metabolism , Diabetes Mellitus/metabolism , Feeder Cells , Folic Acid Deficiency/pathology , Glutathione/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/physiology , Membrane Potential, Mitochondrial , Nitric Oxide/metabolism , Rats , Reactive Oxygen Species/metabolism
12.
J Agric Food Chem ; 61(34): 8191-7, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23899086

ABSTRACT

Shikonin is a traditional Oriental medical herb extracted from Lithospermum erythrorhizon. Many studies have shown that shikonin possesses anticancer ability against many different cancers, including hepatocellular carcinoma (HCC). Recently, tumor metastasis has been become an important clinical obstacle. However, the effect of shikonin on metastasis by HCC is unknown. The 50% inhibitory concentration (IC50) of shikonin on HCC cells was determined by an MTT assay and the xCELLigence biosensor system. The migratory ability of HCC cells was detected by a transwell migration assay and the xCELLigence biosensor system. Matrix metalloproteinase-2 and -9 (MMP-2 and -9) expression levels were determined by Western blotting, and the activities of MMP-2 and -9 were determined by gelatin zymography. We found that IC50 values of HepJ5 and Mahlavu cells to shikonin treatment were around 2 µM. Exposure to a low dose of shikonin (0-0.4 µM) did not influence the survival of HCC cells. Interestingly, exposure to a low dose of shikonin inhibited the migratory ability on HepJ5 and Mahlavu cells. To further dissect the mechanism, we found that treatment with a low dose of shikonin reduced the activities and expression levels of MMP-2 and -9, which were correlated with the decreased cell migratory ability of HCC cells. In addition, we found a decrease of vimnetin expression, but no influence on the expression levels of N-cadherin, TWIST, or GRP78. In mechanism dissecting, we found that shikonin treatment may suppress the phosphorylation of AKT and then reduce the NF-κB (NF = nuclear factor) levels, but has no influence on the levels of c-Fos and c-Jun. Furthermore, we also found that shikonin may also reduce the phosphorylation of IκB. We concluded that a low dose of shikonin can suppress the migratory ability of HCC cells through downregulation of expression levels of vimentin and MMP-2 and -9. Our findings suggest that shikonin may be a new compound to prevent the migration of HCC cells.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cell Movement/drug effects , Drugs, Chinese Herbal/pharmacology , Lithospermum/chemistry , Liver Neoplasms/physiopathology , Naphthoquinones/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Neoplasm Metastasis
13.
Int J Mol Sci ; 14(4): 8291-305, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23591843

ABSTRACT

Bevacizumab, a 149-kDa protein, is a recombinant humanized monoclonal antibody to VEGF. PEDF, a 50-kDa glycoprotein, has demonstrated anti-vasopermeability properties. In this study, we demonstrated that the combination of bevacizumab and plasmid pigment epithelium-derived factor-synthetic amphiphile INTeraction-18(p-PEDF-SAINT-18) has a favorable antiangiogenic effect on corneal NV. Four groups(Group A: 0 µg + 0 µg, B: 0.1 µg + 0.1 µg, C: 1 µg + 1 µg, and D: 10 µg + 10 µg) of bevacizumab + p-PEDF-SAINT-18 were prepared and implanted into the rat subconjunctival substantia propria 1.5 mm from the limbus on the temporal side. Then, 1 µgof p-bFGF-SAINT-18 was prepared and implanted into the rat corneal stroma 1.5 mm from the limbus on the same side. The inhibition of NV was observed and quantified from days 1 to 60. Biomicroscopic examination, western blot analysis and immunohistochemistry were used to analyze the 18-kDa bFGF, 50-kDa PEDF and VEGF protein expression. No inhibition activity for normal limbal vessels was noted. Subconjunctival injection with the combination of bevacizumab and p-PEDF-SAINT-18 successfully inhibited corneal NV.The bFGF and PEDF genes were successfully expressed as shown by western blot analysis,and a mild immune response to HLA-DR was shown by immunohistochemistry. We concluded that the combination of bevacizumab and p-PEDF-SAINT-18 may have more potent and prolonged antiangiogenic effects, making it possible to reduce the frequency of subconjunctival bevacizumab administration combined with a relatively safe profile and low toxicity.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Corneal Neovascularization/prevention & control , Eye Proteins/genetics , Nerve Growth Factors/genetics , Pyridinium Compounds/administration & dosage , Serpins/genetics , Angiogenesis Inhibitors/administration & dosage , Animals , Bevacizumab , Corneal Neovascularization/genetics , Corneal Neovascularization/pathology , Disease Models, Animal , Eye Proteins/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression , Male , Nerve Growth Factors/metabolism , Plasmids/administration & dosage , Plasmids/genetics , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serpins/metabolism , Surface-Active Agents/administration & dosage , Transfection , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
14.
Exp Ther Med ; 5(3): 964-968, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23408313

ABSTRACT

Propyl gallate (PG) is an antioxidant that has been used as an additive in several foods to protect against oxidation. The present study examined the anti-inflammatory effect of PG on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced inflammation in human THP-1 monocytes. Pretreatment with PG markedly inhibited the TPA-induced expression levels of cyclooxygenase-2 and prostaglandin E2. The application of PG significantly inhibited the nuclear translocation of p65, a subunit of nuclear factor-κB (NF-κB) and phosphorylation of p65 (Ser536) in TPA-treated THP-1 cells. PG also inhibited the phosphorylation of IκB and IκB kinase. These results indicate that PG inhibits the inflammatory response by blocking the NF-κB signaling pathway in TPA-induced THP-1 monocytes. Therefore, PG may be useful as a therapeutic agent in inflammatory diseases.

15.
Mol Med Rep ; 6(4): 701-4, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22798045

ABSTRACT

Bevacizumab, a recombinant humanized monoclonal antibody, binds vascular endothelial growth factor (VEGF) and inhibits its interaction with receptors found on endothelial cells. Bevacizumab has been increasingly used as an off-label treatment for exudative age-related macular degeneration (AMD). Whether or not bevacizumab is capable of arresting the growth of human retinal pigment epithelial cells remains to be clarified. In this study, flow cytometry was used to evaluate whether bevacizumab markedly induced the G1/S phase arrest. The G1/S phase cycle-related protein analysis demonstrated that the expression of cyclin-dependent kinase (CDK)2, 4 and 6 and of cyclin D and E, as well as the phosphorylation of retinoblastoma tumor suppressor protein (ppRB) production were found to be markedly reduced by bevacizumab. By contrast, the protein levels of p53, p16, p21 and p27 were increased in bevacizumab-treated ARPE-19 cells (a human retinal pigment epithelial cell line). These events of G1/S arrest induced by bevacizumab in ARPE-19 cells suggest that a preventive effect of bevacizumab exists in AMD.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Retinal Pigment Epithelium/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Bevacizumab , Cell Survival/drug effects , Cells, Cultured , Cyclin D/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Humans , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology , Phosphorylation , Retinal Pigment Epithelium/cytology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
16.
PLoS One ; 7(4): e35123, 2012.
Article in English | MEDLINE | ID: mdl-22529978

ABSTRACT

INTRODUCTION: The 78-kDa glucose-regulated protein (GRP78) is induced in the cancer microenvironment and can be considered as a novel predictor of responsiveness to chemotherapy in many cancers. In this study, we found that intracellular reactive oxygen species (ROS) and nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation were higher in GRP78 knockdown DLD-1 colon cancer cells compared with scrambled control cells. METHODOLOGY/PRINCIPAL FINDINGS: Treatment with epirubicin in GRP78 knockdown DLD-1 cells enhanced apoptosis and was associated with decreased production of intracellular ROS. In addition, apoptosis was increased by the antioxidants propyl gallate (PG) and dithiothreitol (DTT) in epirubicin-treated scrambled control cells. Epirubicin-treated GRP78 knockdown cells resulted in more inactivated Akt pathway members, such as phosphorylated Akt and GSK-3ß, as well as downstream targets of ß-catenin expression. Knockdown of Nrf2 with small interfering RNA (siRNA) increased apoptosis in epirubicin-treated GRP78 knockdown cells, which suggested that Nrf2 may be a primary defense mechanism in GRP78 knockdown cells. We also demonstrated that epirubicin-treated GRP78 knockdown cells could decrease survival pathway signaling through the redox activation of protein phosphatase 2A (PP2A), which is a serine/threonine phosphatase that negatively regulates the Akt pathway. CONCLUSIONS: Our results indicate that epirubicin decreased the intracellular ROS in GRP78 knockdown cells, which decreased survival signaling through both the Akt pathway and the activation of PP2A. Together, these mechanisms contributed to the enhanced level of epirubicin-induced apoptosis that was observed in the GRP78 knockdown cells.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/genetics , Colonic Neoplasms/metabolism , Epirubicin/pharmacology , Heat-Shock Proteins/genetics , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Down-Regulation/drug effects , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/genetics , Gene Silencing , Humans , NF-E2-Related Factor 2/metabolism , Protein Phosphatase 2/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
17.
Ann Surg Oncol ; 19(9): 3097-106, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22446899

ABSTRACT

BACKGROUND: Shikonin is the main naphthoquinone compound of the root of Lithospermum erythrorhizon. Our previous study demonstrated that shikonin possesses anticancer activity in human hepatoma cells. However, the anticancer mechanism of shikonin in human glioma cells is unclear at present. In the present study, we demonstrated that shikonin induces apoptosis in three human glioma cell lines: U87MG, Hs683, and M059K cells. METHODS: Cell cycle, generation of reactive oxygen species (ROS), depletion of glutathione (GSH), and disruption of mitochondrial transmembrane potential in shikonin-treated cells were determined by flow cytometry. Apoptosis-related proteins, catalase, and superoxide dismutase-1 (SOD-1) were determined by Western blot testing. N-acetylcysteine (NAC), pifithrin-α (PFT-α), or cyclosporin A were applied to evaluate the molecular mechanism of shikonin in apoptosis. RESULTS: Shikonin induces the generation of ROS, depletion of GSH, disruption of mitochondrial transmembrane potential, upregulation of p53, and cleavage of PARP [poly(ADP-ribose) polymerase] in U87MG glioma cells. Moreover, shikonin causes catalase downregulation and SOD-1 upregulation as well as decreased Bcl-2 and increased Bax expression. Pretreatment with NAC, PFT-α, or cyclosporin A causes the recovery of shikonin-induced apoptosis. The ROS generation and GSH depletion induced by shikonin trigger mitochondrial transmembrane potential disruption. ROS production was partially dependent on the upregulation of p53 upon shikonin treatment. CONCLUSIONS: These studies are the first to show that shikonin-induced apoptosis occurs through multiple pathways in human glioma cells. We conclude that shikonin may be used as a potential chemotherapeutic agent against human glioma.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Glioma/metabolism , Naphthoquinones/pharmacology , Acetylcysteine/pharmacology , Benzothiazoles/pharmacology , Catalase/drug effects , Catalase/metabolism , Cell Line, Tumor , Cyclosporine/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Glutathione/drug effects , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Oxidative Stress/drug effects , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
18.
Article in English | MEDLINE | ID: mdl-21792367

ABSTRACT

Calvatia lilacina (CL), Pleurotus ostreatus (PO) and Volvariella volvacea (VV) are widely distributed worldwide and commonly eaten as mushrooms. In this study, cell viabilities were evaluated for a human colorectal adenocarcinoma cell line (SW480 cells) and a human monocytic leukemia cell line (THP-1 cells). Apoptotic mechanisms induced by the protein extracts of PO and VV were evaluated for SW480 cells. The viabilities of THP-1 and SW480 cells decreased in a concentration-dependent manner after 24 h of treatment with the protein extracts of CL, PO or VV. Apoptosis analysis revealed that the percentage of SW480 cells in the SubG(1) phase (a marker of apoptosis) was increased upon PO and VV protein-extract treatments, indicating that oligonucleosomal DNA fragmentation existed concomitantly with cellular death. The PO and VV protein extracts induced reactive oxygen species (ROS) production, glutathione (GSH) depletion and mitochondrial transmembrane potential (ΔΨ(m)) loss in SW480 cells. Pretreatment with N-acetylcysteine, GSH or cyclosporine A partially prevented the apoptosis induced by PO protein extracts, but not that induced by VV extracts, in SW480 cells. The protein extracts of CL, PO and VV exhibited therapeutic efficacy against human colorectal adenocarcinoma cells and human monocytic leukemia cells. The PO protein extracts induced apoptosis in SW480 cells partially through ROS production, GSH depletion and mitochondrial dysfunction. Therefore, the protein extracts of these mushrooms could be considered an important source of new anti-cancer drugs.

19.
J Mol Med (Berl) ; 89(3): 303-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21340685

ABSTRACT

Temozolomide (TMZ) is an oral alkylating agent that has been widely used in the treatment of refractory glioma, although inherent and acquired resistance to this drug is common. The clinical use of valproic acid (VPA) as an anticonvulsant and mood-stabilizing drug has been reported primarily for the treatment of epilepsy and bipolar disorder and less commonly for major depression. VPA is also used in the treatment of glioma-associated seizures with or without intracranial operation. In this study, we evaluated the potential synergistic effect of TMZ and VPA in human glioma cell lines. Compared with the use of TMZ or VPA alone, concurrent treatment with both drugs synergistically induced apoptosis in U87MG cells as evidenced by p53 and Bax expression, mitochondrial transmembrane potential loss, reactive oxygen species production, and glutathione depletion. This synergistic effect correlated with a decrease in nuclear translocation of the nuclear factor-erythroid 2 p45-related factor and corresponded with reduced heme oxygenase-1 and γ-glutamylcysteine synthetase expression. Pretreatment with N-acetylcysteine partially recovered the apoptotic effect of the TMZ/VPA combination treatment. The same degree of synergism is also seen in p53-mutant Hs683 cells, which indicates that p53 may not play a major role in the increased proapoptotic effect of the TMZ/VPA combination. In conclusion, VPA enhanced the apoptotic effect of TMZ, possibly through a redox regulation mechanism. The TMZ/VPA combination may be effective for treating glioma cancer and may be a powerful agent against malignant glioma. This drug combination should be further explored in the clinical setting.


Subject(s)
Apoptosis/drug effects , Dacarbazine/analogs & derivatives , Glioma/metabolism , Valproic Acid/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Dacarbazine/pharmacology , Drug Synergism , Humans , Oxidation-Reduction/drug effects , Temozolomide
20.
Food Chem Toxicol ; 49(2): 494-501, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21112369

ABSTRACT

Propyl gallate (PG) is a synthetic antioxidant that has been used in processed food and medicinal preparations. The anti-cancer effect of PG in leukemia is unclear. In the present study, we demonstrate that PG reduced cell viability in THP-1, Jurkat, and HL-60 leukemia cells and induced apoptosis in THP-1 cells. PG activated caspases 3, 8, and 9 and increased the levels of p53, Bax, Fas, and Fas ligand. PG activated mitogen-activated protein kinases (MAPKs), inhibited nuclear translocation of the nuclear factor erythroid 2-related factor 2 (Nrf-2) and induced intracellular glutathione (GSH) depletion. In addition, PG increased superoxide dismutase-1 expression and decreased intracellular levels of reactive oxygen species. Our data show for the first time that an early event of PG-induced apoptosis is MAPKs/Nrf-2-mediated GSH depletion and that PG induced apoptosis via multiple pathways in human leukemia. PG might serve as a potential chemotherapeutic agent or food supplement for human leukemia patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Propyl Gallate/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...