Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 49(2): 579-88, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27221634

ABSTRACT

4-Hydroxybutenolide (K87), a synthetic compound from furfuryl alcohol via photooxidation, was used to investigate whether it can inhibit mobility, migration and invasion of SCC-4 human oral cancer cells in vitro. Cell viability was measured by flow cytometric assay, the enzymatic activities of MMP-2/9 were assayed by gelatin zymography analysis, the protein levels were assayed by western blotting, confocal laser microscopy and EMSA assay, and the gene expression of MMP-2/-7, FAK and ROCK1 mRNA were assayed by PCR. K87 decreased the percentage of viable cells in dose-dependent manner. K87 suppressed cell mobility, migration and invasion of SCC-4 cells dose-dependently. K87 inhibited the enzymatic activities of MMP-2/9 of SCC-4 cells. Western blot analysis revealed that K87 decreased the protein levels in NF-κBp65, COX-2, ROCK1 and Rho A, MMP-1, -2,- 7, -9, VEGF, GRB2, SOS1, PI3K, PKC, PERK, p-PERK, FAK, MEKK3, MKK7, ERK1/2, JNK1/2, p-p38, p38, p-c-Jun, AKT, TIMP2, but increased the protein levels of iNOS, Ras, IRE-1α, p-c-JNK, p-AKT(308), p-AKT(473) and TIMP1. Results from PCR indicated that K87 inhibited the gene expression of MMP-2/-7, FAK and ROCK1 mRNA. Furthermore, confocal laser microscopy was used to confirm that K87 inhibited the translocation of RHOA and ROCK1 in SCC-4 cells. EMSA assay also show that K87 suppressed the nuclear activation of NF-κB and these effects are time-dependent. Western blotting assay indicated that expression of NF-κBp105, NF-κBp50 and NF-κBp65 proteins were decreased and these effects are time-dependent. Based on these observations, we suggest that K87 may be used as a potential agent for anticancer metastasis of human oral cancer in the future.


Subject(s)
4-Butyrolactone/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Movement/drug effects , Head and Neck Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Mouth Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , NF-kappa B/metabolism , Neoplasm Invasiveness , Squamous Cell Carcinoma of Head and Neck
2.
Environ Toxicol ; 31(8): 957-69, 2016 Aug.
Article in English | MEDLINE | ID: mdl-25689151

ABSTRACT

Cancer is the second cause of death in children. Osteosarcoma is the most common primary malignancy of solid bone cancer primarily affecting adolescents and young adults. In the Chinese population, the crude extract of Rheum palmatum L. (CERP) has been used for treating different diseases, including SARS, rheumatoid arthritis, coxsackievirus B3, and human colon cancer cell, pancreatic cancer. There are no reports on CERP and human osteosarcoma cells. The present study examined effects of CERP on cytotoxicity including cell cycle distribution and cell death (apoptosis) in U-2 OS human osteosarcoma cells. CERP significantly induced S phase arrest in U-2 OS cells in a dose-dependent. CERP produced DNA damage and DNA condensation. Other effects of CERP were stimulation of ROS and Ca(2+) , mitochondria impairment, and activation of caspase-3, -8, and -9. CERP increased the levels of Bax, Bak, Bad, cyclin B, Fas, PARP, GRP78, GADD153, AIF, Endo G, Calpain-2, p21, and p27, but decreased the levels of Bcl-2, BCL-X, XIAP, Akt, CDC25A, CDK2, Cyclin A, and Cyclin E of U-2 OS cells. It was also observed that CERP promoted the expression of AIF, Endo G, GADD153, and cytochrome c. These results indicate that CERP has anticancer effects in vitro and provide the foundation for in vivo studies of animal models of osteosarcoma. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 957-969, 2016.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , S Phase Cell Cycle Checkpoints/drug effects , Apoptosis Regulatory Proteins/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Calcium Signaling , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Chaperone BiP , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Protein Transport , Reactive Oxygen Species/metabolism , Rheum/chemistry
3.
Environ Toxicol ; 31(11): 1640-1651, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26174008

ABSTRACT

Although reports have shown that α-phellandrene (α-PA) is one of the monoterpenes and is often used in the food and perfume industry, our previous studies have indicated that α-PA promoted immune responses in normal mice in vivo. However, there is no available information to show that α-PA induced cell apoptosis in cancer cells, thus, we investigated the effects of α-PA on the cell morphology, viability, cell cycle distribution, and apoptosis in mice leukemia WEHI-3 cells in vitro. Results indicated that α-PA induced cell morphological changes and decreased viability, induced G0/G1 arrest and sub-G1 phase (apoptosis) in WEHI-3 cells. α-PA increased the productions of reactive oxygen species (ROS) and Ca2+ and decreased the levels of mitochondrial membrane potential (ΔΨm ) in dose- and time-dependent manners in WEHI-3 cells that were analyzed by flow cytometer. Results from confocal laser microscopic system examinations show that α-PA promoted the release of cytochrome c, AIF, and Endo G from mitochondria in WEHI-3 cells. These results are the first findings to provide new information for understanding the mechanisms by which α-PA induces cell cycle arrest and apoptosis in WEHI-3 cells in vitro. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1640-1651, 2016.


Subject(s)
Apoptosis/drug effects , Monoterpenes/pharmacology , Animals , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclohexane Monoterpenes , Leukemia/drug therapy , Leukemia/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Reactive Oxygen Species/metabolism
4.
Environ Toxicol ; 31(11): 1600-1611, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26122529

ABSTRACT

Prostate cancer is the most frequently diagnosed malignancy in men and the second highest contributor of male cancer mortality. The crude extract of Euphorbia formosana (CEEF) has been used for treatment of different diseases but the cytotoxic effects of CEEF on human cancer cells have not been reported. The purpose of the present experiments was to determine effects of CEEF on cell cycle distribution and induction of apoptosis in DU145 human prostate cancer cells in vitro. Contrast-phase microscope was used for examining cell morphological changes. Flow cytometric assays were used for cell viability, cell cycle, apoptosis, reactive oxygen species, and Ca2+ production and mitochondria membrane potential (ΔΨm ). Western blotting was used for examining protein expression of cell cycle and apoptosis associated proteins. Real-time PCR was used for examining mRNA levels of caspase-3, -8, and -9, AIF, and Endo G. Confocal laser microscope was used to examine the translocation of AIF, Endo G, and cytochrome in DU145 cells after CEEF exposure. CEEF-induced cell morphological changes, decreased the percentage of viable cells, and induced S phase arrest and apoptosis in DU145 cells. Furthermore, CEEF promoted RAS and Ca2+ production and reduced ΔΨm levels. Real-time QPCR confirmed that CEEF promoted the mRNA expression of caspase-3 and -9, AIF and Endo G and we found that AIF and Endo G and cytochrome c were released from mitochondria. Taken together, CEEF-induced cytotoxic effects via ROS production, induced S phase arrest and induction of apoptosis through caspase-dependent and independent and mitochondria-dependent pathways in DU245 cancer cells. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1600-1611, 2016.


Subject(s)
Apoptosis/drug effects , Caspases/physiology , Euphorbia , Plant Extracts/pharmacology , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Male , Mitochondria/physiology , Prostatic Neoplasms/pathology
5.
Am J Chin Med ; 43(6): 1247-64, 2015.
Article in English | MEDLINE | ID: mdl-26446205

ABSTRACT

Bufalin, a component of Chan Su (a traditional Chinese medicine), has been known to have antitumor effects for thousands of years. In this study, we investigated its anti-metastasis effects on NCI-H460 lung cancer cells. Under sub-lethal concentrations (from 25 up to 100 nM), bufalin significantly inhibits the invasion and migration nature of NCI-H460 cells that were measured by Matrigel Cell Migration Assay and Invasion System. Bufalin also suppressed the enzymatic activity of matrix metalloproteinase (MMP)-9, which was examined by gelatin zymography methods. Western blotting revealed that bufalin depressed several key metastasis-related proteins, such as NF-κB, MMP-2, MMP-9, protein kinase C (PKC), phosphatidylinositol 3-kinase (PI3-K), phosphorylated Akt, growth factor receptor-bound protein 2 (GRB2), phosphorylated extracellular signal-regulated kinase (ERK), phosphorylated p38, and phosphorylated c-Jun NH2-terminal kinase (JNK). As evidenced by immunostaining and the electrophoretic mobility shift assay (EMSA), bufalin induced not only a decreased cytoplasmic NF-κB production, but also decreased its nuclear translocation. Several metastasis-related genes, including Rho-associated (Rho A), coiled-coil-containing protein kinase 1 (ROCK1), and focal adhesion kinase (FAK), were down-regulated after bufalin treatment. In conclusion, bufalin is effective in inhibiting the metastatic nature of NCI-H460 cells in low, sub-lethal concentrations. Such an effect involves many mechanisms including MMPs, mitogen-activated protein kinases (MAPKs) and NF-κB systems. Bufalin has a potential to evolve into an anti-metastasis drug for human lung cancer in the future.


Subject(s)
Bufanolides/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , NF-kappa B/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction/drug effects
6.
Anticancer Res ; 35(5): 2691-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25964547

ABSTRACT

Demethoxycurcumin (DMC) is a key component of Chinese medicine (Turmeric) and has been proven effective in killing various cancer cells. Its role in inducing cytotoxic effects in many cancer cells has been reported, but its role regarding DNA damage on lung cancer cells has not been studied in detail. In the present study, we demonstrated DMC-induced DNA damage and condensation in NCI-H460 cells by using the Comet assay and DAPI staining examinations, respectively. Western blotting indicated that DMC suppressed the protein levels associated with DNA damage and repair, such as 14-3-3σ (an important checkpoint keeper of DNA damage response), DNA repair proteins breast cancer 1, early onset (BRCA1), O6-methylguanine-DNA methyltransferase (MGMT), mediator of DNA damage checkpoint 1 (MDC1), and p53 (tumor suppressor protein). DMC activated phosphorylated p53 and p-H2A.X (phospho Ser140) in NCI-H460 cells. Furthermore, we used confocal laser systems microscopy to examine the protein translocation. The results showed that DMC promotes the translocation of p-p53 and p-H2A.X from the cytosol to the nuclei in NCI-H460 cells. Taken together, DMC induced DNA damage and affected DNA repair proteins in NCI-H460 cells in vitro.


Subject(s)
Curcumin/analogs & derivatives , DNA Damage/drug effects , DNA Repair/drug effects , Lung Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Curcumin/administration & dosage , Diarylheptanoids , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Proteins/biosynthesis
7.
Anticancer Res ; 35(4): 2077-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25862863

ABSTRACT

Gallic acid (GA), a phenolic compound naturally present in plants, used as an antioxidant additive in food and in the pharmaceutical industry, may have cancer chemopreventive properties. In the present study, we investigated whether GA induced DNA damage and affected DNA repair-associated protein expression in human oral cancer SCC-4 cells. Flow cytometry assays were used to measure total viable cells and results indicated that GA decreased viable cells dose-dependently. The comet assay and 4',6-Diamidino-2-phenylindole dihydrochloride (DAPI) staining were used to measure DNA damage, as well as condensation and it was shown that GA induced DNA damage (comet tail) and DNA condensation in a dose-dependent manner. DNA gel electrophoresis was used to examine DNA fragmentation and we found that GA induced DNA ladder (fragmentation). Using western blotting it was shown that GA inhibited the protein expressions of MDC1, O(6)-methylguanine-DNA methyltransferase (MGMT), p-H2A.X, p53, DNA-dependent serine/threonine protein kinase (DNA-PK) and 14-3-3 proteins sigma (14-3-3σ) but increased p-p53, phosphate-ataxia-telangiectasia (p-H2A.X) and ataxia telangiectasia mutated and Rad3-related (p-ATR), phosphate-ataxia telangiectasia mutated (p-ATM) and breast cancer susceptibility protein 1 (BRCA1) in a 24-h treatment. The protein translocation was examined by confocal laser microscopy and results indicated that GA increased the levels of p-H2A.X, MDC1 and p-p53 in SCC-4 cells. In conclusion, we found that GA-induced cell death may proceed through the induced DNA damage and suppressed DNA repair-associated protein expression in SCC-4 cells.


Subject(s)
DNA Damage/drug effects , DNA Repair/genetics , Gallic Acid/administration & dosage , Mouth Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , DNA Repair/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics
8.
Oncol Rep ; 33(5): 2429-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25813094

ABSTRACT

Lung cancer is the most common cause of cancer-related mortality in the US as well as other regions of the world. Curcumin, demethoxycurcumin (DMC) and bisdemethoxycurcumin (BDMC) are the major components of Curcuma longa L. It has been reported that curcumin inhibits the growth of various types of cancer cells in vitro and in vivo. However, the mechanisms involved in the inhibition of cell growth and induced apoptosis by DMC in human lung cancer cells remain unclear. In the present study, we investigated the effect of DMC on cell death via the induction of apoptosis in NCI-H460 human lung cancer cells. Flow cytometric assay was used to examine the total percentage of viable cells, the population of cells in the sub-G1 phase of the cell cycle, the level of reactive oxygen species (ROS), Ca²âº production, mitochondrial membrane potential (ΔΨm) and caspase activity. Western blotting was used to examine the changes in the expression of cell cycle- and apoptosis-associated proteins. Confocal microscopy was used to examine the translocation of apoptosis-associated proteins. The results indicated that DMC significantly induced cell morphological changes and decreased the percentage of viable NCI-H460 cells and DMC induced apoptosis based on the cell distribution in the sub-G1 phase. Moreover, DMC promoted ROS and Ca²âº production and decreased the level of ΔΨm and promoted the activities of caspase-3, -8 and -9. The Western blotting results showed that DMC promoted the expression of AIF, Endo G and PARP. The levels of Fas ligand (Fas L) and Fas were also upregulated. Furthermore, DMC promoted expression of ER stress-associated proteins such as GRP78, GADD153, IRE1ß, ATF-6α, ATF-6ß and caspase-4. Based on the findings, we suggest that DMC may be used as a novel anticancer agent for the treatment of lung cancer in the future.


Subject(s)
Apoptosis/drug effects , Curcumin/analogs & derivatives , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mitochondria/drug effects , Signal Transduction/drug effects , Apoptosis Regulatory Proteins/metabolism , Calcium/metabolism , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Curcumin/pharmacology , Diarylheptanoids , Endoplasmic Reticulum Chaperone BiP , G1 Phase/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
9.
Mol Med Rep ; 12(1): 1030-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25815777

ABSTRACT

Cantharidin (CTD) induces cytotoxic effects in different types of human cancer cell; however, to date, there have been no studies on the effects of CTD on gene expression in human lung cancer cells and the potential associated signaling pathways. Therefore, the present study aimed to investigate how CTD affects the expression of key genes and functional pathways of human H460 lung cancer cells using complementary DNA microarray analysis. Human H460 lung cancer cells were cultured for 24 h in the presence or absence of 10 µM CTD; gene expression was then examined using microarray analysis. The results indicated that 8 genes were upregulated > 4-fold, 29 genes were upregulated >3-4-fold and 156 genes were upregulated >2-3-fold. In addition, 1 gene was downregulated >4 fold, 14 genes were downregulated >3-4-fold and 150 genes were downregulated >2-3 fold in H460 cells following exposure to CTD. It was found that CTD affected DNA damage genes, including DNIT3 and GADD45A, which were upregulated 2.26- and 2.60-fold, respectively, as well as DdiT4, which was downregulated 3.14-fold. In addition, the expression of genes associated with the cell cycle progression were altered, including CCND2, CDKL3 and RASA4, which were upregulated 2.72-, 2.19- and 2.72-fold, respectively; however, CDC42EP3 was downregulated 2.16-fold. Furthermore, apoptosis-associated genes were differentially expressed, including CARD6, which was upregulated 3.54-fold. In conclusion, the present study demonstrated that CTD affected the expression of genes associated with DNA damage, cell cycle progression and apoptotic cell death in human lung cancer H460 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cantharidin/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin D2/genetics , Cyclin D2/metabolism , DNA Damage , Gene Expression Profiling , Gene Ontology , Humans , Microarray Analysis , Molecular Sequence Annotation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
10.
Am J Chin Med ; 43(2): 365-82, 2015.
Article in English | MEDLINE | ID: mdl-25779644

ABSTRACT

Numerous evidences have shown that plant flavonoids (naturally occurring substances) have been reported to have chemopreventive activities and protect against experimental carcinogenesis. Kaempferol, one of the flavonoids, is widely distributed in fruits and vegetables, and may have cancer chemopreventive properties. However, the precise underlying mechanism regarding induced DNA damage and suppressed DNA repair system are poorly understood. In this study, we investigated whether kaempferol induced DNA damage and affected DNA repair associated protein expression in human leukemia HL-60 cells in vitro. Percentages of viable cells were measured via a flow cytometry assay. DNA damage was examined by Comet assay and DAPI staining. DNA fragmentation (ladder) was examined by DNA gel electrophoresis. The changes of protein levels associated with DNA repair were examined by Western blotting. Results showed that kaempferol dose-dependently decreased the viable cells. Comet assay indicated that kaempferol induced DNA damage (Comet tail) in a dose-dependent manner and DAPI staining also showed increased doses of kaempferol which led to increased DNA condensation, these effects are all of dose-dependent manners. Western blotting indicated that kaempferol-decreased protein expression associated with DNA repair system, such as phosphate-ataxia-telangiectasia mutated (p-ATM), phosphate-ataxia-telangiectasia and Rad3-related (p-ATR), 14-3-3 proteins sigma (14-3-3σ), DNA-dependent serine/threonine protein kinase (DNA-PK), O(6)-methylguanine-DNA methyltransferase (MGMT), p53 and MDC1 protein expressions, but increased the protein expression of p-p53 and p-H2AX. Protein translocation was examined by confocal laser microscopy, and we found that kaempferol increased the levels of p-H2AX and p-p53 in HL-60 cells. Taken together, in the present study, we found that kaempferol induced DNA damage and suppressed DNA repair and inhibited DNA repair associated protein expression in HL-60 cells, which may be the factors for kaempferol induced cell death in vitro.


Subject(s)
Cell Survival/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Flavonoids , Kaempferols/pharmacology , Leukemia, Prolymphocytic/genetics , Leukemia, Prolymphocytic/pathology , Chemoprevention , Gene Expression/drug effects , HL-60 Cells , Humans , Kaempferols/therapeutic use , Leukemia, Prolymphocytic/prevention & control , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phytotherapy , Stimulation, Chemical
11.
Anticancer Res ; 35(2): 729-38, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25667452

ABSTRACT

Cancer metastasis is the major cause of cancer patient death. Melanoma is a highly important metastasis in human cancer. Cantharidin (CTD), identified as an active component of natural mylabris (Mylabris phalerata Pallas), induces apoptosis in many human cancer cells. In the present study, we investigated the anti-metastasis effects of CTD in human melanoma cancer A375.S2 cells. Flow cytometry was used to measure CTD-induced cytotoxic effects in A375.S2 cells. Wound healing assay indicated that CTD suppressed the migration of A375.S2 cells in a dose-dependent manner. The Matrigel Transwell Assay was used for cell migration and invasion examination and the results showed that CTD inhibited both. Gelatin zymography was used to investigate the activities of MMP-2/9 and the results indicated that CTD inhibited the enzymatic activities of MMP-2/9 in A375.S2 cells. The protein expression of A375.S2 cells following incubation with CTD was examined by western blotting and the results showed that CTD decreased the expression of ERK1/2, PI3K, FAK, MMP-2, -9, COX-2, NF-κB p65, TIMP 1, TIMP 2, VEFG, uPA, Rho A, GRB2, ROCK-1 and Ras, but increased the expressions of p38, JNK, p-c-jun and PKC. Based on those observations, we suggest that CTD may be used as a novel anti-cancer metastasis agent of human melanoma cancer in the future.


Subject(s)
Cantharidin/pharmacology , Cell Movement/drug effects , Melanoma/pathology , Neoplasm Invasiveness/prevention & control , Signal Transduction/drug effects , Cell Line, Tumor , Humans , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 9/drug effects , Melanoma/enzymology , Melanoma/metabolism , NF-kappa B/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors
12.
In Vivo ; 29(1): 83-94, 2015.
Article in English | MEDLINE | ID: mdl-25600535

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths and new lung cancer cases are continuously emerging around the globe; however, treatment of lung cancer remains unsatisfactory. Demethoxycurcumin (DMC) has been shown to exert cytotoxic effects in human cancer cells via induction of apoptosis. However, the effects of DMC on genetic mechanisms associated with these actions have not been yet elucidated. Human lung cancer NCI-H460 cells were incubated with or without 35 µM of DMC for 24 h and total RNA was extracted for cDNA synthesis labeling and microarray hybridization, followed by fluor-labeled cDNA hybridization on chip. Expression Console software with default Robust Multichip Analysis (RMA) parameters were used for detecting and quantitating the localized concentrations of fluorescent molecules. The GeneGo software was used for investigating key genes involved and their possible interaction pathways. Genes associated with DNA damage and repair, cell-cycle check point and apoptosis could be altered by DMC; in particular, 144 genes were found up-regulated and 179 genes down-regulated in NCI-H460 cells after exposure to DMC. In general, DMC-altered genes may offer information to understand the cytotoxic mechanism of this agent at the genetic level since gene alterations can be useful biomarkers or targets for the diagnosis and treatment of human lung cancer in the future.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Curcumin/analogs & derivatives , DNA Damage/drug effects , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Curcumin/pharmacology , DNA Damage/genetics , Diarylheptanoids , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Signal Transduction
13.
J Cell Mol Med ; 19(2): 474-84, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25403643

ABSTRACT

Angiogenesis inhibitors are beneficial for the prevention and treatment of angiogenesis-dependent diseases including cancer. We examined the cytotoxic, anti-metastatic, anti-cancer and anti-angiogenic effects of diallyl trisulfide (DATS). In HT29 cells, DATS inhibited migration and invasion through the inhibition of focal adhesion kinase (FAK), extracellular signal-regulated kinase, c-Jun N-terminal kinase and p38 which was associated with inhibition of matrix metalloproteinases-2, -7 and -9 and VEGF. In human umbilical vein endothelial cells (HUVEC), DATS inhibited the migration and angiogenesis through FAK, Src and Ras. DATS also inhibited the secretion of VEGF. The capillary-like tube structure formation and migration by HUVEC was inhibited by DATS. The chicken egg chorioallantoic membrane (CAM) assay indicated that DATS treatment inhibited ex-vivo angiogenesis. We investigated the anti-tumour effects of DATS against human colon cancer xenografts in BALB/c(nu/nu) mice and its anti-angiogenic activity in vivo. In this in-vivo study, DATS also inhibited the tumour growth, tumour weight and angiogenesis (decreased the levels of haemoglobin) in HT29 cells. In conclusion, the present results suggest that the inhibition of angiogenesis may be an important mechanism in colon cancer chemotherapy by DATS.


Subject(s)
Allyl Compounds/pharmacology , Angiogenesis Inhibitors/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Neoplasm Invasiveness/prevention & control , Neovascularization, Pathologic/drug therapy , Sulfides/pharmacology , Animals , Cell Line , Cell Line, Tumor , HT29 Cells , Humans , Mice , Xenograft Model Antitumor Assays/methods
14.
Environ Toxicol ; 30(10): 1135-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24639390

ABSTRACT

Cantharidin is one of the major compounds from mylabris and it has cytotoxic effects in many different types of human cancer cells. Previously, we found that cantharidin induced cell death through cell cycle arrest and apoptosis induction in human lung cancer NCI-H460 cells. However, cantharidin-affected DNA damage, repair, and associated protein levels in NCI-H460 cells have not been examined. In this study, we determined whether cantharidin induced DNA damage and condensation and altered levels of proteins in NCI-H460 cells in vitro. Incubation of NCI-H460 cells with 0, 2.5, 5, 10, and 15 µM of cantharidin caused a longer DNA migration smear (comet tail). Cantharidin also increased DNA condensation. These effects were dose-dependent. Cantharidin (5, 10, and 15 µM) treatment of NCI-H460 cells reduced protein levels of ataxia telangiectasia mutated (ATM), breast cancer 1, early onset (BRCA-1), 14-3-3 proteins sigma (14-3-3σ), DNA-dependent serine/threonine protein kinase (DNA-PK), O(6) -methylguanine-DNA methyltransferase (MGMT), and mediator of DNA damage checkpoint protein 1 (MDC1). Protein translocation of p-p53, p-H2A.X (S140), and MDC1 from cytoplasm to nucleus was induced by cantharidin in NCI-H460 cells. Taken together, this study showed that cantharidin caused DNA damage and inhibited levels of DNA repair-associated proteins. These effects may contribute to cantharidin-induced cell death in vitro.


Subject(s)
Cantharidin/toxicity , DNA Damage/drug effects , DNA Repair Enzymes/metabolism , DNA Repair/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Comet Assay , DNA Repair Enzymes/antagonists & inhibitors , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microscopy, Confocal
15.
Environ Toxicol ; 30(7): 852-63, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24497447

ABSTRACT

Crude extract of Rheum palmatum L. (CERP) has been used to treat different diseases in the Chinese population for decades. In this study, we investigated the anti-metastasis effects of CERP on LS1034 human colorectal cancer cells in vitro and examined potential mechanisms of its effects. CERP significantly inhibited cell migration and invasion of LS1034 cells. We also found that CERP inhibited protein levels of matrix metalloproteinases-2 (MMP-2) and matrix metalloproteinases-9 (MMP-9), and cytosolic NF-kB p65, RHO A, ROCK 1. Furthermore, we found CERP inhibited protein levels of GRB2, SOS1, MKK7, FAK, Rho A, ROCK 1, VEGF, PKC, AKT, phosphor-AKT (Thr308), Cyclin D, iNOS, COX2, NF-kB p65, p-ERK1/2, p-JNK1/2, p-p38, p-c-jun, MMP-2, MMP-9, MMP-1, MMP-7, MMP-10, UPA and increased the protein level of Ras in LS1034 cells. In conclusion, our results suggest that CERP may be used as a novel anti-metastasis agent for the treatment of human colon cancer cells.


Subject(s)
MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Plant Extracts/pharmacology , Rheum/chemistry , Cell Culture Techniques , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Plant Extracts/chemistry , Rheum/metabolism , Transcription Factor RelA/metabolism , Wound Healing/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
16.
Environ Toxicol ; 30(1): 74-82, 2015 Jan.
Article in English | MEDLINE | ID: mdl-23949904

ABSTRACT

Metastasis plays an important role in mortality of cancer patients. Migration and invasion are the major characteristics of tumor metastasis. The induction of matrix metalloproteinases (MMPs) such as MMP-2 and -9 are particularly important for the invasiveness of various cancer cells. Bufalin, a class of toxic steroids, was purified from the skin glands of Bufo gargarizans or Bufo melanostictus; it is known to inhibit proliferation of human cancer cells. In this study, we investigated the antiinvasive mechanisms of bufalin in the human hepatocellular cancer cell line SK-Hep1. Bufalin significantly reduced serum-induced cell invasion and migration. Furthermore, bufalin markedly inhibited MMP-2 and -9 activity, mRNA expression and protein levels in SK-Hep1 cells. Bufalin attenuated phosphoinisitide-3-kinase (PI3K) and phosphorylation of AKT which was associated with reduced levels of nuclear factor kappa B (NF-κB). Bufalin also suppressed protein levels of FAK and Rho A, VEGF, MEKK3, MKK7, and uPA and it diminished NF-κB translocation. Based on these observations, we propose that bufalin is acts as an antiinvasive agent by inhibiting MMP-2 and -9 and involving PI3K/AKT and NF-κB pathways. Bufalin is a potential therapeutic agent that may have efficacy in preventing the invasion and metastasis of malignant liver tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Bufanolides/pharmacology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Humans , Neoplasm Invasiveness , Signal Transduction/drug effects
17.
Environ Toxicol ; 30(1): 53-63, 2015 Jan.
Article in English | MEDLINE | ID: mdl-23955962

ABSTRACT

Osteosarcoma is the most common primary malignancy of the bone cancers. In the Chinese population, the crude extract of Corni Fructus (CECF) has been used as Traditional Chinese medicine to treat several different diseases for hundreds of years. In the present study, effects of CECF on inhibition of migration and invasion in U-2 OS human osteosarcoma cells were examined. CECF significantly inhibited migration and invasion of U-2 OS human osteosarcoma cells. We also found that CECF inhibited activities of matrix metalloproteinases-2 (MMP-2) and matrix metalloproteinases-9 (MMP-9). CECF decreased protein levels of FAK, PKC, SOS1, MKK7, MEKK3, GRB2, NF-κB p65, COX-2, HIF-1α, PI3K, Rho A, ROCK-1, IRE-1α, p-JNK1/2, p-ERK1/2, p-p38, Ras, p-PERK, MMP-2, MMP-9, and VEGF in U-2 OS cells. Results of this study indicate that CECF may have potential as a novel anticancer agent for the treatment of osteosarcoma by inhibiting migration and invasion of cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cornus/chemistry , Drugs, Chinese Herbal/pharmacology , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Bone Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Drugs, Chinese Herbal/isolation & purification , Humans , Matrix Metalloproteinase Inhibitors/isolation & purification , NF-kappa B/metabolism , Neoplasm Invasiveness , Osteosarcoma/pathology
18.
Environ Toxicol ; 30(11): 1322-30, 2015 Nov.
Article in English | MEDLINE | ID: mdl-24861204

ABSTRACT

Although there are few reports regarding α-phellandrene (α-PA), a natural compound from Schinus molle L. essential oil, there is no report to show that α-PA induced DNA damage and affected DNA repair associated protein expression. Herein, we investigated the effects of α-PA on DNA damage and repair associated protein expression in murine leukemia cells. Flow cytometric assay was used to measure the effects of α-PA on total cell viability and the results indicated that α-PA induced cell death. Comet assay and 4,6-diamidino-2-phenylindole dihydrochloride staining were used for measuring DNA damage and condensation, respectively, and the results indicated that α-PA induced DNA damage and condensation in a concentration-dependent manner. DNA gel electrophoresis was used to examine the DNA damage and the results showed that α-PA induced DNA damage in WEHI-3 cells. Western blotting assay was used to measure the changes of DNA damage and repair associated protein expression and the results indicated that α-PA increased p-p53, p-H2A.X, 14-3-3-σ, and MDC1 protein expression but inhibited the protein of p53, MGMT, DNA-PK, and BRCA-1.


Subject(s)
DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Monoterpenes/pharmacology , Anacardiaceae/chemistry , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Comet Assay , Cyclohexane Monoterpenes , DNA-Activated Protein Kinase/genetics , Dose-Response Relationship, Drug , Gene Expression/drug effects , Mice , Microscopy, Confocal , Monoterpenes/isolation & purification , Protein Biosynthesis
19.
Int J Oncol ; 45(6): 2393-402, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25340978

ABSTRACT

Cantharidin (CTD), a component of natural mylabris (Mylabris phalerata Pallas) was reported to have high cytotoxicity in many human cancer cell lines. However, it was not reported to affect human melanoma A375.S2 cells. In the present study, we found that CTD induced cell morphological changes and decreased the percentage of viable cells and induced G2/M phase arrest and induction of apoptosis in A375.S2 cells. Results also showed that CTD induced the generation of reactive oxygen species (ROS) and Ca2+ and decreased mitochondria membrane potential and lead to the release of cytochrome c, AIF and Endo G. Further investigation revealed that CTD induced A375.S2 cells with an increase of caspase activation and caspase-dependent apoptotic proteins to trigger correlated pathway mechanisms according to western blotting results. Western blotting was used for examining the changes of G2/M phase arrest and apoptosis-associated protein expression and confocal laser microscopy was used to examine the translocation apoptosis-associated protein. Results showed that CTD increased the protein expression of caspase-3, -8 and -9, cytochrome c, Bax, Bid, Endo G and AIF but inhibited the levels of Bcl-2 and Bcl-x. CTD induced ER stress-associated protein expression such as GRP78, IRE1ß, ATF6α and caspase-12. Based on those observations, we suggest that CTD may have potential as a novel anti-cancer agent for the treatment of skin cancer.


Subject(s)
Cantharidin/administration & dosage , Cyclin A/biosynthesis , Melanoma/drug therapy , cdc25 Phosphatases/biosynthesis , Apoptosis/drug effects , Cell Line, Tumor , Cyclin A/antagonists & inhibitors , Endoplasmic Reticulum Chaperone BiP , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/genetics , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Neoplasm Proteins/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction , cdc25 Phosphatases/antagonists & inhibitors
20.
Molecules ; 19(10): 16588-608, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25322282

ABSTRACT

Osteosarcoma is the most common malignant primary bone tumor in children and young adults and lung metastasis is the main cause of death in those patients. Deguelin, a naturally occurring rotenoid, is known to be an Akt inhibitor and to exhibit cytotoxic effects, including antiproliferative and anticarcinogenic activities, in several cancers. In the present study, we determined if deguelin would inhibit migration and invasion in U-2 OS human osteosarcoma cells. Deguelin significantly inhibited migration and invasion of U-2 OS human osteosarcoma cells which was associated with a reduction of activities of matrix metalloproteinases-2 (MMP-2) and matrix metalloproteinases-9 (MMP-9). Furthermore, results from western blotting indicated that deguelin decreased the cell proliferation and cell growth-associated protein levels, such as SOS1, PKC, Ras, PI3K, p-AKT(Ser473), IRE-1α, MEKK3, iNOS, COX2, p-ERK1/2, p-JNK1/2, p-p38; the cell motility and focal adhesion-associated protein levels, such as Rho A, FAK, ROCK-1; the invasion-associated protein levels, such as TIMP1, uPA, MMP-2. MMP-9, MMP-13, MMP-1 and VEGF in U-2 OS cells. Confocal microscopy revealed that deguelin reduced NF-κB p65, Rho A and ROCK-1 protein levels in cytosol. MMP-7, MMP-9 and Rho A mRNA levels were suppressed by deguelin. These in vitro results provide evidence that deguelin may have potential as a novel anti-cancer agent for the treatment of osteosarcoma and provides the rationale for in vivo studies in animal models.


Subject(s)
Bone Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Osteosarcoma/metabolism , Rotenone/analogs & derivatives , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Vitro Techniques , Neoplasm Invasiveness , Osteosarcoma/pathology , Rotenone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...