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










Database
Publication year range
1.
Int J Mol Sci ; 22(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34830011

ABSTRACT

The Chinese medicine monomer cynaroside (Cy) is a flavonoid glycoside compound that widely exists in plants and has a variety of pharmacological effects, such as its important role in the respiratory system, cardiovascular system and central nervous system. Studies have reported that Cy has varying degrees of anticancer activity in non-small cell lung cancer, cervical cancer, liver cancer, esophageal cancer and other cancers. However, there are no relevant reports about its role in gastric cancer. The MET/AKT/mTOR signaling pathway plays important roles in regulating various biological processes, including cell proliferation, apoptosis, autophagy, invasion and tumorigenesis. In this study, we confirmed that Cy can inhibit the cell growth, migration and invasion and tumorigenesis in gastric cancer. Our finding shows that Cy can block the MET/AKT/mTOR axis by decreasing the phosphorylation level of AKT, mTOR and P70S6K. Therefore, the MET/AKT/mTOR axis may be an important target for Cy. In summary, Cy has anti-cancer properties and is expected to be a potential drug for the treatment of gastric cancer.


Subject(s)
Glucosides/pharmacology , Luteolin/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/drug therapy , TOR Serine-Threonine Kinases/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medicine, Chinese Traditional , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
2.
Sheng Wu Gong Cheng Xue Bao ; 37(9): 3031-3041, 2021 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-34622615

ABSTRACT

Tigecycline is a novel glycylcycline antibacterial drug, which shows both antibiotic function and anti-tumor activity. This review summarizes the single and combined use of tigecycline for tumor treatment and the underpinning mechanisms. As an inhibitor for mitochondrial DNA translation, tigecycline affects the proliferation, migration, and invasion of tumor cells mainly through inhibiting mitochondrial protein synthesis and inducing mitochondrial dysfunction. Although the effect of tigecycline monotherapy is controversial, the efficacy of combined use of tigecycline is satisfactory. Therefore, it is important to explore the molecular mechanisms underpinning the anti-tumor activity of tigecycline, with the aim to use it as a cheap and effective new anti-tumor drug.


Subject(s)
Minocycline , Neoplasms , Anti-Bacterial Agents/pharmacology , Humans , Minocycline/pharmacology , Mitochondria , Neoplasms/drug therapy , Tigecycline/pharmacology
3.
Cells ; 8(8)2019 07 25.
Article in English | MEDLINE | ID: mdl-31349699

ABSTRACT

Tubeimoside-1 (TBMS1) is one of the extracts of rhizoma bolbostemmae, which has remarkable anti-cancer function in the treatment of esophagus and gastric cancer in traditional Chinese medicine. However the mechanisms of its anti-cancer function is remain unclear. In this study, we demonstrate that TBMS1 could inhibit cell growth and metastasis in glioblastoma. MET is a member of the receptor tyrosine kinase family, which amplifies frequently in various human cancers. As an important proto-oncogene, multiple inhibitors have been developed for the therapy of cancers. Here, we found TBMS1 could reduce/decrease the protein level of MET via increasing its Ubiquitination degradation. Therefore, TBMS1 is a promising compound for the treatment of glioblastoma and an inhibitor of MET.


Subject(s)
Glioblastoma/metabolism , Proto-Oncogene Proteins c-met/metabolism , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glioblastoma/pathology , Humans , Mice , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
4.
Cancer Biomark ; 21(2): 307-316, 2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29103022

ABSTRACT

BACKGROUND: The cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein which involves in the progression of several human malignancies. Development of cisplatin (DDP) resistance is the obstacle to an effective control of gastric cancer (GC) clinically. OBJECTIVE: We thus assessed whether CIP2A expression is associated with sensitivity of GC to DDP. METHODS: Real-time quantitative PCR, immunohistochemical analysis, or western blotting was performed to detect CIP2A expression in GC patients' tissues. SGC7901/DDP cells were transfected with CIP2A siRNA. MTT assay was used to determine the DDP-sensitivity of cells. Flow cytometry was used to measure cell apoptosis. RESULTS: CIP2A has higher expression in DDP-resistant GC patients. DDP-resistant GC patients with high CIP2A expression presented with poorer overall survival rates than those with low CIP2A expression. CIP2A knockdown in DDP-resistant GC cells resulted in attenuated proliferative abilities and increased apoptosis level. CIP2A depletion sensitizes DDP-resistant cells to DDP and CIP2A overexpression antagonizes DDP-sensitive cells to DDP. CIP2A influences the expression of multidrug resistance-related proteins in GC cells. CONCLUSIONS: Our results suggested that CIP2A oncoprotein plays an important role in DDP resistance of GC and could serve as a novel therapeutic target for the treatment of GC patients with DDP resistance.


Subject(s)
Autoantigens/biosynthesis , Cisplatin/pharmacology , Membrane Proteins/biosynthesis , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autoantigens/genetics , Autoantigens/metabolism , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tissue Array Analysis
5.
Oncol Rep ; 38(1): 598-606, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28560452

ABSTRACT

We have shown that a novel STAT3 inhibitor arctigenin (Atn) induces significant cytotoxicity in triple-negative breast cancer (TNBC) cells. This study further delineated molecular mechanisms where by Atn triggered cytotoxicity in TNBC cells. We found Atn can also inhibit metastasis in TNBC cells through cancerous inhibitor of protein phosphatase 2A (CIP2A) pathway. CIP2A is an endogenous inhibitor of protein phosphatase 2A (PP2A), which can increase the migration and invasion of various cancer cells. PP2A is a tumor suppressor, which is functionally defective in various cancers. Atn-induced metastasis inhibition was associated with reactivation of PP2A, downregulation of CIP2A and Akt phosphorylation. Silencing CIP2A enhanced Atn-induced metastasis inhibition and apoptosis in TNBCs. Furthermore, ectopic expression of CIP2A or inhibition of PP2A in TNBC cells abolished the effects of Atn. In conclusion, we found that enhancement of PP2A activity by inhibition of CIP2A, at least in part, promotes the anti-metastasis effect induced by Atn. Our findings disclose the novel therapeutic mechanism of this targeted agent, and suggest the therapeutic potential and feasibility of developing PP2A enhancers as a novel anticancer strategy.


Subject(s)
Antineoplastic Agents/pharmacology , Autoantigens/metabolism , Furans/pharmacology , Lignans/pharmacology , Membrane Proteins/metabolism , Protein Phosphatase 2/metabolism , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis , Autoantigens/genetics , Cell Line, Tumor , Down-Regulation , Furans/therapeutic use , Humans , Intracellular Signaling Peptides and Proteins , Lignans/therapeutic use , Membrane Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/pathology
6.
Oncol Rep ; 38(1): 271-278, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28534965

ABSTRACT

Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a human oncoprotein that is overexpressed in multiple kinds of tumors including gastric cancer (GC). Mammalian target of rapamycin complex 1 (mTORC1) over-activation is detected in GC and many other cancers. Previous study found that CIP2A/mTORC1 controls cell growth and autophagy through direct association. CIP2A plays an 'oncogenic nexus' in several cancer types to participate in the tumorigenic transformation and chemoresistance. In the present study, we investigated whether Cucurbitacin B (CuB), a natural compound found in Cucurbitaceae, can be used in cisplatin (DDP)-resistant human GC cell line SGC7901/DDP. Results demonstrated that CuB treatment significantly suppressed SGC7901/DDP cell proliferation, induced caspase-dependent apoptosis, and autophagy. The activation of autophagy was mediated through CuB-induced inhibition of mTORC1. Furthermore, CuB inhibited mTORC1 via the activation of protein phosphatase 2A (PP2A) which is mediated by CIP2A inhibition. These findings indicated that CuB can inhibit the proliferation, induce caspase-dependent apoptosis, and autophagy of SGC7901/DDP cells by suppressing CIP2A/PP2A/mTORC1 signaling axis. Thus, CuB may be a novel effective candidate to treat DDP-resistant human GC cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Stomach Neoplasms/pathology , Triterpenes/pharmacology , Antineoplastic Agents/pharmacology , Autoantigens/metabolism , Cell Proliferation/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Mechanistic Target of Rapamycin Complex 1/metabolism , Membrane Proteins/metabolism , Protein Phosphatase 2/metabolism , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...