Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Anticancer Res ; 41(4): 1859-1870, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33813391

ABSTRACT

BACKGROUND/AIM: Demethoxycurcumin (DMC), one of the derivatives of curcumin, has been shown to induce apoptotic cell death in many human cancer cell lines. However, there is no available information on whether DMC inhibits metastatic activity in human glioblastoma cancer cells in vitro. MATERIALS AND METHODS: DMC at 1.0-3.0 µM significantly decreased the proliferation of GBM 8401 cells; thus, we used 2.0 µM for further investigation regarding anti-metastatic activity in human glioblastoma GBM 8401 cells. RESULTS: The internalized amount of DMC has reached the highest level in GBM 8401 cells after 3 h treatment. Wound healing assay was used to determine cell mobility and results indicated that DMC suppressed cell movement of GBM 8401 cells. The transwell chamber assays were used for measuring cell migration and invasion and results indicated that DMC suppressed cell migration and invasion in GBM 8401 cells. Gelatin zymography assay was used to examine gelatinolytic activity (MMP-2) in conditioned media of GBM 8401 cells treated by DMC and results demonstrated that DMC significantly reduced MMP-2 activity. Western blotting showed that DMC reduced the levels of p-EGFR(Tyr1068), GRB2, Sos1, p-Raf, MEK, p-ERK1/2, PI3K, p-Akt/PKBα(Thr308), p-PDK1, NF-κB, TIMP-1, MMP-9, MMP-2, GSK3α/ß, ß-catenin, N-cadherin, and vimentin, but it elevated Ras and E-cadherin at 24 h treatment. CONCLUSION: DMC inhibited cancer cell migration and invasion through inhibition of PI3K/Akt and NF-κB signaling pathways in GBM 8401 cells. We suggest that DMC may be used as a novel anti-metastasis agent for the treatment of human glioblastoma cancer in the future.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/drug therapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Diarylheptanoids/pharmacology , Glioblastoma/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Signal Transduction , beta Catenin/metabolism
2.
Biosci Biotechnol Biochem ; 83(9): 1729-1739, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31010399

ABSTRACT

Lupeol, one of the common components from the fruits and natural foods, has been reported to exert antitumor activities in many human cancer cell lines; however, its effects on osteosarcoma cell metastasis were not elucidated. In the present study, lupeol at 10-25 µM induced cell morphological changes and decreased total viable cell number in U-2 OS cells. Lupeol (5-15 µM) suppressed cell mobility, migration, and invasion by wound healing and transwell chamber assays, respectively. Lupeol inhibited the activities of MMP-2 and -9 in U-2 OS cells by gelatin zymography assay. Lupeol significantly decreased PI3K, pAKT, ß-catenin, and increased GSK3ß. Furthermore, lupeol decreased the expressions of Ras, p-Raf-1, p-p38, and ß-catenin. Lupeol also decreased uPA, MMP-2, MMP-9, and N-cadherin but increased VE-cadherin in U-2 OS cells. Based on these observations, we suggest that lupeol can be used in anti-metastasis of human osteosarcoma cells in the future.


Subject(s)
Bone Neoplasms/pathology , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Osteosarcoma/pathology , Pentacyclic Triterpenes/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Bone Neoplasms/enzymology , Cell Line, Tumor , Cell Survival/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Matrix Metalloproteinases/metabolism , Osteosarcoma/enzymology
3.
Anticancer Res ; 38(7): 3989-3997, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29970522

ABSTRACT

BACKGROUND/AIM: Bisdemethoxycurcumin (BDMC) exhibits biological activities including anticancer and anti-metastasis in human cancer cell lines, but there is no available information to show whether BDMC suppresses cell migration and invasion of human cervical cancer cells. MATERIALS AND METHODS: Wound-healing, migration, invasion, zymography, and western blotting assays were used to investigate the effects of BDMC on HeLa cells in vitro. RESULTS: BDMC reduced the total viable cell number in a dose-dependent manner. The wound-healing assay show BDMC suppressed the movement of HeLa cells. Furthermore, the trans-well chamber assays showed that BDMC suppressed the cell migration and invasion. Gelatin zymograph assay showed that BDMC did not inhibit matrix metalloproteinase-2 (MMP-2) and -9 activities in vitro. However, western blotting assay showed that BDMC significantly reduced protein levels of growth factor receptor-bound protein 2 (GRB2), Ras homolog gene family, member A (Rho A), urokinase-type plasminogen activator (uPA), RAS, MMP-2, and N-cadherin but increased those of phosphor-extracellular-signal related kinase (p-ERK1/2), E-cadherin and nuclear factor-ĸB (NF-ĸB) in HeLa cells. Confocal laser microscopy assay was used to further confirm BDMC increased NF-ĸB when compared to controls. CONCLUSION: BDMC may have potential as a novel anti-metastasis agent for the treatment of human cervical cancer.


Subject(s)
Curcumin/analogs & derivatives , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/antagonists & inhibitors , Uterine Cervical Neoplasms/pathology , Blotting, Western , Curcumin/pharmacology , Diarylheptanoids , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Female , HeLa Cells , Humans , Microscopy, Confocal , Neoplasm Invasiveness , Neoplasm Metastasis
4.
Anticancer Res ; 38(5): 2761-2769, 2018 05.
Article in English | MEDLINE | ID: mdl-29715097

ABSTRACT

BACKGROUND/AIM: Demethoxycurcumin (DMC), one of the curcuminoids present in turmeric, has been shown to induce cell death in many human cancer cell lines, however, there has not been any investigation on whether DMC inhibits metastatic activity in human cervical cancer cells in vitro. In the present study, DMC at 2.5-15 µM decreased cell number, thus, we used IC20 (7.5 µM) for further investigation of its anti-metastatic activity in human cervical cancer HeLa cells. MATERIALS AND METHODS: The wound healing, migration, invasion, zymography, and western blotting assays were used to investigate the effects of DMC on HeLa cells. RESULTS: The wound healing assay was used to show that DMC suppressed cell movement of HeLa cells. Furthermore, the trans-well chamber assay was used to show that DMC suppressed HeLa cell migration and invasion. Gelatin zymography assay did not show any significant effects of DMC on the gelatinolytic activity (MMP-2 and -9) in conditioned media of HeLa cells treated by DMC. Western blotting showed that DMC significantly reduced protein levels of GRB2, MMP-2, ERK1/2, N-cadherin and Ras but increased the levels of E-cadherin and NF-κB in HeLa cells. Confocal laser microscopy indicated that DMC increased NF-κB in HeLa cells confirming the results from Western blotting. CONCLUSION: DMC may be used as a novel anti-metastatic agent for the treatment of human cervical cancer in the future.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/analogs & derivatives , Gene Expression Regulation, Neoplastic/drug effects , NF-kappa B/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Culture Media, Conditioned , Curcumin/pharmacology , Diarylheptanoids , Drug Screening Assays, Antitumor , Female , HeLa Cells , Humans , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...