Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
2.
Chin J Integr Med ; 25(11): 845-852, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31127506

ABSTRACT

OBJECTIVE: To characterize the molecular mechanism underlying the antineoplastic activity of Celastrus orbiculatus Thunb. extracts (COE). METHODS: The human hepatocellular carcinoma HepG2 cells with mammalian target of rapamycin (mTOR) knockdown expressed (HepG2/mTOR) were constructed using molecular biological technology. In vitro, the HepG2/mTOR- cells were treated with COE at various concentrations (10, 20, 40, 80, 160 and 320 µ g/mL). Cell viability was determined using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assays. According to the half-maximal inhibitory concentration (IC50) value (140 mg/L), the concentrations of COE in the subsequent experiment was set to alleviate cytotoxicity. The HepG2/mTOR- cells were divided into 5 groups: negative control (untreated), COE treatment groups (40, 80, 120 mg/L COE) and positive control group (cisplatin, DDP, 2 mg/L), respectively. Wild-type HepG2 cells were used as a blank control. The effects of COE on the cell apoptosis were analyzed by flow cytometry and transmission electronic microscopy (TEM), respectively. The protein expression levels of mTOR signal pathways were determined by Western blotting. In vivo, HepG2/mTOR- cells (2 × 106 cell/mice) were subcutaneously injected into the right flank of nude mice. Thirty-six female nude mice were randomly assigned to 6 groups according to body weight (6 mice per group) as follows: solvent vehicle control, Banmao Capsule treated group (BM, 195 mg/kg), Tegafur, Gimeracil and Oteracil Potassium Capsules (10 mg/kg) treated group, and different dosages of COE (10, 20, 40 mg/kg) groups. Tumor growth was monitored and immunohistochemical staining was used to examine the expression of apoptosis-related proteins in tumor tissues. RESULTS: COE inhibited the proliferation significantly in a concentration-dependent manner in HepG2/mTOR- cells (P<0.01). COE significantly induced the apoptosis of HepG2/mTOR- cells (P<0.01), and the apoptotic bodies can be observed under TEM. COE significantly inhibits the proteins expression of mTOR-related signal pathways. In vivo, COE significantly inhibited tumor growth in nude mice (P<0.01). Moreover, the results showed that COE down-regulated the expression of Bcl-2 and Bcl-xL, and up-regulated the levels of Bax and caspase-3 protein (P<0.01). CONCLUSION: COE was a potential chemotherapeutic drug in HCC treatments via targeting mTOR signal pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/pathology , Celastrus/chemistry , Cell Proliferation/drug effects , Liver Neoplasms/pathology , TOR Serine-Threonine Kinases/genetics , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Cell Survival/drug effects , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Mice , Mice, Nude , Plant Extracts/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
3.
Chin J Integr Med ; 25(5): 334-341, 2019 May.
Article in English | MEDLINE | ID: mdl-30046956

ABSTRACT

OBJECTIVE: To evaluate the effects of Celastrus Orbiculatus extracts (COE) on metastasis in hypoxia-induced hepatocellular carcinoma cells (HepG2) and to explore the underlying molecular mechanisms. METHODS: The effect of COE (160, 200 and 240 µ g/mL) on cell viability, scratch-wound, invasion and migration were studied by 3-4,5-dimethyl-2-thiazolyl-2,5-diphenyl-2-H-tetrazolium bromide (MTT), scratch-wound and transwell assays, respectively. CoCl2 was used to establish a hypoxia model in vitro. Effects of COE on the expressions of E-cadherin, vimentin and N-cadherin were investigated with Western blot and immunofluorescence analysis, respectively. RESULTS: COE inhibited proliferation and metastasis of hypoxia-induced hepatocellular carcinoma cells in a dose-dependent manner (P<0.01). Furthermore, the expression of epithelial-mesenchymal transition (EMT) related markers were also remarkably suppressed in a dose-dependent manner (P<0.01). In addition, the upstream signaling pathways, including the hypoxia-inducible factor 1 α (Hif-1 α) and Twist1 were suppressed by COE. Additionally, the Hif-1 α inhibitor 3-5'-hydroxymethyl-2'-furyl)-1-benzylindazole (YC-1), potently suppressed cell invasion and migration as well as expression of EMT in hypoxia-induced HepG2 cells. Similarly, the combined treatment with COE and YC-1 showed a synergistic effect (P<0.01) compared with the treatment with COE or YC-1 alone in hypoxia-induced HepG2 cells. CONCLUSIONS: COE significantly inhibited the tumor metastasis and EMT by suppressing Hif-1 α/Twist1 signaling pathway in hypoxia-induced HepG2 cell. Thus, COE might have potential effect to inhibit the progression of HepG2 in the context of tumor hypoxia.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Celastrus/chemistry , Down-Regulation , Epithelial-Mesenchymal Transition , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Plant Extracts/therapeutic use , Biomarkers, Tumor/metabolism , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cobalt , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hep G2 Cells , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects
4.
Chin J Integr Med ; 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25382615

ABSTRACT

OBJECTIVE: To assess the effect of Celastrus orbiculatus (COE) on growth, invasion and migration of human gastric cancer MGC-803 cells and to explore the possible mechanism. METHODS: The effect of COE on cell viability, apoptosis, adhesion, invasion and migration were studied by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, flow cytometric, cell adhesion and transwell assay, respectively. The activity and expression of matrix metalloproteinase-9 (MMP-9) were determined by gelatin zymography, Western blot and quantitative real-time polymerase chain reaction analysis. Meanwhile, effects of COE on the expression of mitogen-activated protein kinases (MAPKs), serine threonine kinase (Akt), nuclear factor κB (NF-κB) were investigated with Western blot analysis. RESULTS: COE inhibited proliferation and induced apoptosis of MGC-803 cells in a dose-dependent manner. When treated with low-toxic (below 80 µg/mL) doses of COE, cell adhesion, invasion and migration were markedly suppressed. Furthermore, the gelatinolytic activity and expression of MMP-9 were also remarkably suppressed in a dose-dependent manner. In addition, upstream signaling pathways, including the phosphatidylinositol-3 kinase (PI3K)/Akt and NF-κB, were suppressed by COE. Additionally, the PI3K/Akt inhibitor, LY294002, in treating MGC-803 cells potently suppressed cell invasion and migration as well as expression of MMP-9. Similarly, the combined treatment with COE and LY294002 showed a synergistic effect compared with the treatment with COE or LY294002 alone in MGC-803 cells. CONCLUSIONS: COE inhibits invasion and migration of MGC-803 cells by reducing MMP-9 expression. It also inhibit PI3K/Akt and NF-κB signaling pathways, which may offer a novel approach for the treatment of human gastric cancer.

5.
Int Immunopharmacol ; 17(4): 1028-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24144813

ABSTRACT

Rheumatoid arthritis (RA) joints are in a hypoxic condition. Hypoxia-induced migration and invasion of fibroblast-like synoviocytes (FLSs) are considered to play a critical role in the pathogenesis of RA. Among the key genes upregulated by hypoxia-inducible factor-1α (HIF-1α), CXC chemokine receptor 4 (CXCR4) plays an important role in FLS migration and invasion. Our previous studies have shown that celastrol exerts anti-arthritic effects by inhibiting FLS migration and invasion under normoxic conditions. However, the effect and molecular mechanisms underlying the effect of celastrol on hypoxia-induced FLS migration and invasion are poorly understood. In the present study, we assessed the effect of celastrol on hypoxia-induced FLS migration and invasion. Results showed that celastrol suppressed hypoxia-induced FLS migration and invasion. In addition, we also found that celastrol inhibited hypoxia-induced CXCR4 expression at both the mRNA and the protein levels in RA-FLSs. Meanwhile, it is revealed that celastrol inhibited the transcriptional activity of CXCR4 under hypoxic conditions by suppressing the binding activity of HIF-1α in the CXCR4 promoter, and blocked hypoxia-induced accumulation of nuclear HIF-1α. Furthermore, treatment with HIF-1α inhibitor reduced the hypoxia-induced expression and transcriptional activity of CXCR4. In conclusion, our results indicate that celastrol inhibits hypoxia-induced migration and invasion via suppression of HIF-1α mediated CXCR4 expression in FLSs under hypoxic conditions. These results provide a strong rationale for further testing and validation of the use of celastrol as a new alternative for using in the treatment of RA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia/metabolism , Receptors, CXCR4/antagonists & inhibitors , Triterpenes/pharmacology , Arthritis, Rheumatoid/metabolism , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pentacyclic Triterpenes , RNA, Small Interfering/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Synovial Membrane/cytology
6.
Mol Cell Biochem ; 372(1-2): 221-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23001847

ABSTRACT

Migration and invasion of fibroblast-like synoviocytes (FLSs) are critical in the pathogenesis of rheumatoid arthritis (RA). Hypoxic conditions are present in RA joints, and hypoxia has been extensively studied in angiogenesis and inflammation. However, its effect on the migration and invasion of RA-FLSs remains unknown. In this study, we observed that RA-FLSs exposed to hypoxic conditions experienced epithelial-mesenchymal transition (EMT), with increased cell migration and invasion. We demonstrated that hypoxia-induced EMT was accompanied by increased hypoxia-inducible factor (HIF)-1α expression and activation of Akt. After knockdown or inhibition of HIF-1α in hypoxia by small interfering RNA or genistein (Gen) treatment, the EMT transformation and invasion ability of FLSs were regained. HIF-1α could be blocked by phosphatidylinositol 3-kinase (PI3K) inhibitor, LY294002, indicating that HIF-1α activation was regulated by the PI3K/Akt pathway. Administration of LY294002 (20 mg/kg, intra-peritoneally) twice weekly and Gen (25 mg/kg, by gavage) daily for 3 weeks from day 20 after primary immunization in a collagen-induced arthritis rat model, markedly alleviated the clinical signs, radiology progression, synovial hyperplasia, and inflammatory cells infiltration of joints. Thus, results of this study suggest that activation of the PI3K/Akt/HIF-1α pathway plays a pivotal role in mediating hypoxia-induced EMT transformation and invasion of RA-FLSs under hypoxia.


Subject(s)
Arthritis, Rheumatoid/pathology , Epithelial-Mesenchymal Transition , Fibroblasts/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Cell Hypoxia , Cell Movement , Chromones/administration & dosage , Chromones/pharmacology , Enzyme Activation , Female , Fibroblasts/metabolism , Gene Knockdown Techniques , Genistein/administration & dosage , Genistein/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Knee Joint/pathology , Morpholines/administration & dosage , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Signal Transduction , Synovial Fluid/enzymology , Synovial Fluid/metabolism
7.
Int Immunopharmacol ; 14(4): 422-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22954486

ABSTRACT

Interleukin-17A (IL-17A)-induced migration and invasion of fibroblast-like synoviocytes (FLSs) is critical for the pathogenesis of rheumatoid arthritis (RA). More than 30% of RA patients are resistant to available therapies, despite the introduction of novel biologic agents. Therefore, it is necessary to develop new anti-arthritic agents. Recent studies have demonstrated that celastrol has anti-arthritic activity in an adjuvant-induced arthritis (AIA) model. However, the effect and molecular mechanisms of celastrol on the migration and invasion of RA-FLSs are not yet understood. Results showed that treatment of RA-FLSs with celastrol suppressed the IL-17A-induced migration and invasion abilities of the cells. In addition, celastrol inhibited IL-17A-induced matrix metalloproteinase (MMP)-9 mRNA and protein expression, and the proteolytic activity of MMP-9 in RA-FLSs. Furthermore, our results revealed that celastrol inhibited the transcriptional activity of MMP-9 by suppression of the binding activity of nuclear factor-κB (NF-κB) in the MMP-9 promoter, and inhibited IκBα phosphorylation and nuclear translocation of NF-κB. In conclusion, celastrol can inhibit IL-17A-induced migration and invasion by suppressing NF-κB-mediated MMP-9 expression in RA-FLSs. These results provide a strong rationale for further testing and validation of celastrol as an adjunct with conventional drugs for the treatment of RA in humans.


Subject(s)
Arthritis, Rheumatoid/immunology , Fibroblasts/drug effects , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Synovial Fluid/cytology , Triterpenes/pharmacology , Cell Movement/drug effects , Cells, Cultured , Fibroblasts/physiology , Gene Expression Regulation, Enzymologic , Humans , Interleukin-17 , Matrix Metalloproteinase 9/genetics , Molecular Structure , Pentacyclic Triterpenes , RNA Folding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Triterpenes/chemistry
8.
Chin J Integr Med ; 18(10): 752-60, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21805294

ABSTRACT

OBJECTIVE: Celastrus orbiculatus Thunb. has been used for thousands of years in China as a remedy against cancer and inflammatory diseases. This study aims to investigate whether C. orbiculatus extract (COE) could inhibit angiogenesis, which is the pivotal step in tumor growth, invasiveness, and metastasis. METHODS: In this study, the extract from the stem of C. orbiculatus was used. Mouse hepatic carcinoma cells (Hepa1-6) were treated with COE in different nontoxic concentrations (10, 20, 40, 80, and 160 µg/mL). The mRNA and protein expression levels of vascular endothelial growth factor (VEGF) were detected by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot, respectively; the active fractions were further tested on C57BL/6 mice and human umbilical vein endothelial cells (HUVEC) for any antiangiogenic effects. RESULTS: COE significantly inhibited proliferation and induced apoptosis in Hepa1-6 cells and inhibited VEGF expression at both mRNA and protein levels. Furthermore, this agent inhibited the formation of the capillary-like structure in primary cultured HUVEC in a dose-dependent manner. In vivo, COE significantly reduced the volume and weight of solid tumors with low adverse effects and decreased tumor angiogenesis. CONCLUSIONS: In summary, COE could be used to treat hepatic carcinoma. The mechanisms of the antitumor activity of COE may be due to its effects against tumor angiogenesis by targeting the VEGF protein.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Celastrus/chemistry , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Plant Extracts/therapeutic use , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Administration, Oral , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen/metabolism , Drug Combinations , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Laminin/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Plant Stems/chemistry , Proteoglycans/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/biosynthesis
9.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(12): 1161-3, 2009 Dec.
Article in Chinese | MEDLINE | ID: mdl-19961808

ABSTRACT

AIM: To study the immunogenicity of p210(bcr-abl);, the epitopes of HLA-A2 restricted T cells and the distribution of epitope-specific CTLs in chronic myeloid leukemia (CML) patients and in normal controls. METHODS: Two epitopes, BCR-ABL(642); and BCR-ABL(926m);, were selected using bioinfomatics software and further confirmed by T2 cell binding assay. The soluble HLA-A2 tetramers bound with each epitope were generated to detect the CD8(+); T cell frequencies in peripheral blood mononuclear cells. RESULTS: The epitope-specific CD8(+); T cell frequencies of both BCR-ABL(642); and BCR-ABL(926m); were significantly higher in CML patients, compared with those in healthy individuals(P<0.01), but no significant difference was observed in influenza epitope-specific CTLs between the patients and healthy individuals (P>0.05). The frequency of BCR-ABL(642); peptide-specific CTLs at chronic phase was significantly higher than that at blast phase in CML patients (P<0.05). CONCLUSION: The two candidate epitopes selected from p210(bcr-abl); are characterized by their immunogenicity and based on them, vaccines or adoptive CTL therapies can be developed.


Subject(s)
Epitopes, T-Lymphocyte , Leukocytes, Mononuclear , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukocytes, Mononuclear/metabolism , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...