Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 39(6): 2797-2806, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29620286

ABSTRACT

Transforming growth factor­ß1 (TGF­ß1) has been demonstrated to promote epithelial­mesenchymal transition (EMT), invasion and proliferation in tumors via the activation of Rac1 and ß­catenin signaling pathways. The present study investigated the effects of diallyl disulfide (DADS) on TGF­ß1­induced EMT, invasion and growth of gastric cancer cells. TGF­ß1 treatment augmented EMT and invasion, concomitantly with increased expression of TGF­ß1, Rac1 and ß­catenin in gastric cancer cells. DADS downregulated the expression levels of TGF­ß1, Rac1 and ß­catenin. DADS, TGF­ß1 receptor inhibitor as well as Rac1 inhibitor antagonized the upregulation of the TGF­ß1­induced expression of these genes, abolishing the enhanced effects of TGF­ß1 on EMT and invasion. Blocking the TGF­ß1 receptor through inhibition resulted in the decreased expression of Rac1 and ß­catenin. Rac1 inhibitor reduced the TGF­ß1­induced ß­catenin expression. In addition, DADS and the aforementioned inhibitors attenuated the TGF­ß1­induced tumor growth and the expression changes of E­cadherin, vimentin, Ki­67 and CD34 in nude mice. These data indicated that the blockage of TGF­ß1/Rac1 signaling by DADS may be responsible for the suppression of EMT, invasion and tumor growth in gastric cancer.


Subject(s)
Allyl Compounds/administration & dosage , Antineoplastic Agents/administration & dosage , Disulfides/administration & dosage , Stomach Neoplasms/drug therapy , Transforming Growth Factor beta1/metabolism , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism , Allyl Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disulfides/pharmacology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Invasiveness , Stomach Neoplasms/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
2.
Oncotarget ; 7(9): 10498-512, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26871290

ABSTRACT

Diallyl disulfide (DADS) has been shown to have multi-targeted antitumor activities. We have previously discovered that it has a repressive effect on LIM kinase-1 (LIMK1) expression in gastric cancer MGC803 cells. This suggests that DADS may inhibit epithelial-mesenchymal transition (EMT) by downregulating LIMK1, resulting in the inhibition of invasion and growth in gastric cancer. In this study, we reveal that LIMK1 expression is correlated with tumor differentiation, invasion depth, clinical stage, lymph node metastasis, and poor prognosis. DADS downregulated the Rac1-Pak1/Rock1-LIMK1 pathway in MGC803 cells, as shown by decreased p-LIMK1 and p-cofilin1 levels, and suppressed cell migration and invasion. Knockdown and overexpression experiments performed in vitro demonstrated that downregulating LIMK1 with DADS resulted in restrained EMT that was coupled with decreased matrix metalloproteinase-9 (MMP-9) and increased tissue inhibitor of metalloproteinase-3 (TIMP-3) expression. In in vitro and in vivo experiments, the DADS-induced suppression of cell proliferation was enhanced and antagonized by the knockdown and overexpression of LIMK1, respectively. Similar results were observed for DADS-induced changes in the expression of vimentin, CD34, Ki-67, and E-cadherin in xenografted tumors. These results indicate that downregulation of LIMK1 by DADS could explain the inhibition of EMT, invasion and proliferation in gastric cancer cells.


Subject(s)
Allyl Compounds/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Disulfides/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Lim Kinases/metabolism , Stomach Neoplasms/pathology , Animals , Antigens, CD34/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cofilin 1/metabolism , Down-Regulation , Humans , Ki-67 Antigen/metabolism , Lim Kinases/biosynthesis , Lim Kinases/genetics , Lymphatic Metastasis , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/pathology , Neoplasm Transplantation , Stomach Neoplasms/mortality , Tissue Inhibitor of Metalloproteinase-3/metabolism , Transplantation, Heterologous , Vimentin/metabolism , p21-Activated Kinases/biosynthesis , rac1 GTP-Binding Protein/biosynthesis , rho-Associated Kinases/biosynthesis
3.
Oncol Lett ; 7(5): 1625-1631, 2014 May.
Article in English | MEDLINE | ID: mdl-24765190

ABSTRACT

The existence of cancer stem cells (CSCs) is central to the pathogenesis and therapeutic target of human hepatocellular carcinoma. The aim of this study was to investigate the effects of casticin on epithelial-mesenchymal transition (EMT) of liver cancer stem cells (LCSCs) derived from the SMMC-7721 cell line. Our results demonstrated that CD133+ sphere-forming cells (SFCs) sorted from the SMMC-7721 cell line not only possessed a higher capacity to form tumor spheroids in vitro, but also had a greater potential to form tumors when implanted in Balb/c-nu mice, indicating that CD133+ SFCs possessed similar traits to LCSCs. Casticin increased the expression levels of E-cadherin and decreased those of N-cadherin in LCSCs. Treatment of LCSCs with casticin for 48 h also decreased the levels of the EMT-associated transcription factor, Twist. Overexpression of Twist attenuated the casticin-induced regulation of E-cadherin and N-cadherin protein expression, as well as the EMT capacity of LCSCs. In conclusion, CD133+ SFCs of the SMMC-7721 cell line may represent a subpopulation of LCSCs with the characteristics of EMT. Furthermore, casticin targeted LCSCs through the inhibition of EMT by downregulating Twist.

4.
Food Chem Toxicol ; 68: 61-70, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24650757

ABSTRACT

Diallyl disulfide (DADS) has been shown to cause G2/M phase cell cycle arrest in several human cancers. Here we demonstrate a mechanism by which DADS induces G2/M phase arrest in BGC823 human gastric cancer cells via Chk1. From cell cycle gene array results, we next confirmed that cyclin B1 expression was decreased by DADS, while the expression of p21, GADD45α and p53 were increased. Despite the lack of change in Chk1 gene expression in response to DADS according to the array analysis, intriguingly overexpression of Chk1, but not Chk2, exhibited increased accumulation in G2/M phase. Moreover, overexpression of Chk1 promoted the effect of DADS-induced G2/M arrest. Augmented phosphorylation of Chk1 by DADS was observed in Chk1-transfected cells, followed by downregulation of Cdc25C and cyclin B1 proteins. In contrast, phosphorylated Chk2 showed no obvious change in Chk2-transfected cells after DADS treatment. Furthermore, knockdown of Chk1 by siRNA partially abrogated DADS-induced downregulation of Cdc25C and cyclin B1 proteins and G2/M arrest. In contrast, knockdown of Chk2 did not show these effects. Therefore, these data indicate that DADS may specifically modulate Chk1 phosphorylation, and DADS-induced G2/M phase arrest in BGC823 cells could result in part from Chk1-mediated inhibition of the Cdc25C/cyclin B1 pathway.


Subject(s)
Allyl Compounds/adverse effects , Disulfides/adverse effects , G2 Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/drug effects , Protein Kinases/metabolism , Cell Line, Tumor , Checkpoint Kinase 1 , Cyclin B1/genetics , Cyclin B1/metabolism , Down-Regulation , Humans , Phosphorylation , Protein Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
5.
Cytotechnology ; 64(1): 43-51, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21938469

ABSTRACT

Carvacrol is one of the members of monoterpene phenol and is present in the volatile oils of Thymus vulgaris, Carum copticum, origanum and oregano. It is a safe food additive commonly used in our daily life, and few studies have indicated that carvacrol has anti-hepatocarcinogenic activities. The rationale of the study was to examine whether carvacrol affects apoptosis of human hepatoma HepG2 cells. In this study, we showed that carvacrol inhibited HepG2 cell growth by inducing apoptosis as evidenced by Hoechst 33258 stain and Flow cytometric (FCM) analysis. Incubation of HepG2 cells with carvacrol for 24 h induced apoptosis by the activation of caspase-3, cleavage of PARP and decreased Bcl-2 gene expression. These results demonstrated that a significant fraction of carvacrol treated cells died by an apoptotic pathway in HepG2 cells. Moreover, carvacrol selectively altered the phosphorylation state of members of the MAPK superfamily, decreasing phosphorylation of ERK1/2 significantly in a dose-dependent manner, and activated phosphorylation of p38 but not affecting JNK MAPK phosphorylation. These results suggest that carvacrol may induce apoptosis by direct activation of the mitochondrial pathway, and the mitogen-activated protein kinase pathway may play an important role in the antitumor effect of carvacrol. These results have identified, for the first time, the biological activity of carvacrol in HepG2 cells and should lead to further development of carvacrol for liver disease therapy.

6.
World J Gastroenterol ; 15(14): 1744-50, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19360918

ABSTRACT

AIM: To investigate the anti-angiogenic and anti-tumor activities of recombinant vascular basement membrane-derived multifunctional peptide (rVBMDMP) in hepatocellular carcinoma (HCC). METHODS: HepG2, Bel-7402, Hep-3B, HUVE-12 and L-02 cell lines were cultured in vitro and the inhibitory effect of rVBMDMP on proliferation of cells was detected by MTT assay. The in vivo antitumor efficacy of rVBMDMP on HCC was assessed by HepG2 xenografts in nude mice. Distribution of rVBMDMP, mechanism by which the growth of HepG2 xenografts is inhibited, and microvessel area were observed by proliferating cell nuclear antigen (PCNA) and CD31 immunohistochemistry. RESULTS: MTT assay showed that rVBMDMP markedly inhibited the proliferation of human HCC (HepG2, Bel-7402, Hep-3B) cells and human umbilical vein endothelial (HUVE-12) cells in a dose-dependent manner, with little effect on the growth of L-02 cells. When the IC(50) was 4.68, 7.65, 8.96, 11.65 and 64.82 micromol/L, respectively, the potency of rVBMDMP to HepG2 cells was similar to 5-fluorouracil (5-FU) with an IC(50) of 4.59 micromol/L. The selective index of cytotoxicity to HepG2 cells of rVBMDMP was 13.8 (64.82/4.68), which was higher than that of 5-FU [SI was 1.9 (8.94/4.59)]. The VEGF-targeted recombinant humanized monoclonal antibody bevacizumab (100 mg/L) did not affect the proliferation of HepG2, Bel-7402, Hep-3B and L-02 cells, but the growth inhibitory rate of bevacizumab (100 mg/L) to HUVE-12 cells was 87.6% +/- 8.2%. Alternis diebus intraperitoneal injection of rVBMDMP suppressed the growth of HepG2 xenografts in a dose-dependent manner. rVBMDMP (1, 3, 10 mg/kg) decreased the tumor weight by 12.6%, 55.9% and 79.7%, respectively, compared with the vehicle control. Immunohistochemical staining of rVBMDMP showed that the positive area rates (2.2% +/- 0.73%, 4.5% +/- 1.3% and 11.5% +/- 3.8%) in rVBMDMP treated group (1, 3, 10 mg/kg) were significantly higher than that (0.13% +/- 0.04%) in the control group (P < 0.01). The positive area rates (19.0% +/- 5.7%, 12.2% +/- 3.5% and 5.2% +/- 1.6% ) of PCNA in rVBMDMP treated group (1, 3, 10 mg/kg) were significantly lower than that (29.5% +/- 9.4%) in the control group (P < 0.05). rVBMDMP at doses of 1, 3 and 10 mg/kg significantly reduced the tumor microvessel area levels (0.26% +/- 0.07%, 0.12% +/- 0.03% and 0.05% +/- 0.01% vs 0.45% +/- 0.15%) in HepG2 xenografts (P < 0.01), as assessed by CD31 staining. CONCLUSION: rVBMDMP has effective and unique anti-tumor properties, and is a promising candidate for the development of anti-tumor drugs.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Basement Membrane/chemistry , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/drug therapy , Liver Neoplasms/physiopathology , Peptides/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Recombinant Proteins/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Blood Vessels/anatomy & histology , Blood Vessels/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...