Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Environ Toxicol ; 36(2): 276-286, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33030807

ABSTRACT

Quinalizarin (Quina) is one of the main components of many herbal medicines and has good anti-tumor activity. However, the exact mode of cytotoxic action and signaling pathways on Quina in human esophageal cancer has not yet been confirmed. In this study, we explored the anticancer effect of Quina against human esophageal cancer HCE-4 cells and the underlying mechanisms. The results of the Cell Counting Kit-8 (CCK-8) assay showed that Quina inhibited the viability of human esophageal cancer HCE-4 cells in a dose-dependent and time-dependent manner. It also inhibited HCE-4 cells proliferation and induced apoptosis by increasing the levels of Bad, caspase-3, and PARP, decreasing the level of Bcl-2. The results of the cell cycle analysis suggested that Quina arrested HCE-4 cells in the G0/G1 cycle by downregulating cyclin-dependent (CDK) 2/4, cyclin D1/E and upregulating the levels of p21 and p27. We also found that Quina activated mitogen-activated protein kinase (MAPK) and inhibited the signal transducer and activator of transcription-3 (STAT3) and nuclear factor kappa B (NF-κB) signaling pathways. Furthermore, Quina significantly increased intracellular reactive oxygen species (ROS) level. The pretreatment of N-acetyl-L-cysteine (NAC) blocked the apoptosis induced by Quina and inhibited the activities of MAPK, STAT3, and NF-κB signaling pathways. These results indicate that Quina induces the apoptosis in HCE-4 cells, which is via accumulating ROS generation and regulating MAPK, STAT3, and NF-κB. In conclusion, this study demonstrated that Quina have good therapeutic effects on human esophageal cancer cells.


Subject(s)
Anthraquinones/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Esophageal Neoplasms/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Esophageal Neoplasms/metabolism , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
2.
Mol Med Rep ; 20(5): 4576-4586, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31702038

ABSTRACT

Quinalizarin has been demonstrated to exhibit potent antitumor activities in lung cancer and gastric cancer cells, but currently, little is known regarding its anticancer mechanisms in human breast cancer cells. The aim of the present study was to investigate the apoptotic effects of quinalizarin in MCF­7 cells and to analyze its molecular mechanisms. The MTT assay was used to evaluate the viability of human breast cancer cells that had been treated with quinalizarin and 5­fluorouracil. Flow cytometric analyses and western blotting were used to investigate the effects of quinalizarin on apoptosis and cycle arrest in MCF­7 cells with focus on reactive oxygen species (ROS) production. The results demonstrated that quinalizarin exhibited significant cytotoxic effects on human breast cancer cells in a dose­dependent manner. Accompanying ROS, quinalizarin induced MCF­7 cell mitochondrial­associated apoptosis by regulating mitochondrial­associated apoptosis, and caused cell cycle arrest at the G2/M phase in a time­dependent manner. Furthermore, quinalizarin can activate p38 kinase and JNK, and inhibit the extracellular signal­regulated kinase, signal transducer and activator of transcription 3 (STAT3) and NF­κB signaling pathways. These effects were blocked by mitogen­activated protein kinase (MAPK) inhibitor and N­acetyl­L­cysteine. The results from the present study suggested that quinalizarin induced G2/M phase cell cycle arrest and apoptosis in MCF­7 cells through ROS­mediated MAPK, STAT3 and NF­κB signaling pathways. Thus, quinalizarin may be useful for human breast cancer treatment, as well as the treatment of other cancer types.


Subject(s)
Anthraquinones/pharmacology , Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , MCF-7 Cells
3.
Drug Dev Res ; 80(5): 573-584, 2019 08.
Article in English | MEDLINE | ID: mdl-30916421

ABSTRACT

Glycitein is an isoflavone that reportedly inhibits the proliferation of human breast cancer and prostate cancer cells. However, its anti-cancer molecular mechanisms in human gastric cancer remain to be defined. This study evaluated the antitumor effects of glycitein on human gastric cancer cells and investigated the underlying mechanisms. We used MTT assay, flow cytometry and western blotting to investigate its molecular mechanisms with focus on reactive oxygen species (ROS) production. Our results showed that glycitein had significant cytotoxic effects on human gastric cancer cells. Glycitein markedly decreased mitochondrial transmembrane potential (ΔΨm) and increased AGS cells mitochondrial-related apoptosis, and caused G0/G1 cell cycle arrest by regulating cycle-related protein. Mechanistically, accompanying ROS, glycitein can activate mitogen-activated protein kinase (MAPK) and inhibited the signal transducer and activator of transcription 3 (STAT3) and nuclear factor-kappaB (NF-κB) signaling pathways. Furthermore, the MAPK signaling pathway regulated the expression levels of STAT3 and NF-κB upon treatment with MAPK inhibitor and N-acetyl-L-cysteine (NAC). These findings suggested that glycitein induced AGS cell apoptosis and G0/G1 phase cell cycle arrest via ROS-related MAPK/STAT3/NF-κB signaling pathways. Thus, glycitein has the potential to a novel targeted therapeutic agent for human gastric cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Isoflavones/pharmacology , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism , Stomach Neoplasms/metabolism , Acetylcysteine/pharmacology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/drug therapy
4.
Int J Mol Med ; 43(2): 1067-1075, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30535477

ABSTRACT

The present study investigated the mechanisms of apoptosis induced by cryptotanshinone (CT) in human rheumatoid arthritis fibroblast­like synoviocytes (RA­FLSs). Cell Counting kit­8 assay was performed to determine the cytotoxic effects of CT in human RA­FLSs, including primary RA­FLS, HFLS­RA and MH7A cells, and in HFLS cells derived from normal synovial tissue. Annexin V­FITC/PI staining was used to detect the apoptotic effects of CT in HFLS­RA and MH7A cells. Flow cytometry was performed to detect the apoptotic and reactive oxygen species (ROS) levels induced by CT in HFLS­RA cells. Western blotting was used to assess the expression levels of proteins associated with apoptosis and with the mitogen­activated protein kinase (MAPK), protein kinase B (Akt), and signal transducer and activator of transcription­3 (STAT3) signaling pathways. The results demonstrated that CT treatment significantly suppressed HFLS­RA and MH7A cell growth, whereas no clear inhibitory effect was observed in normal HFLS cells. CT exposure downregulated the expression levels of B­cell lymphoma 2 (Bcl­2), p­Akt, p­extracellular signal­related kinase and p­STAT3, while it upregulated the expression levels of Bcl­2­associated death promoter (Bad), caspase­3, poly (ADP­ribose) polymerase (PARP), p­p38 and p­c­Jun N­terminal kinase. Following ROS scavenging, the CT­induced apoptosis and altered expression levels of Bcl­2, Bad, cleaved caspase­3 and cleaved PARP were restored. Furthermore, the Akt, MAPK and STAT3 signaling pathways were regulated by intracellular ROS. These results suggest that ROS­mediated Akt, MAPK and STAT3 signaling pathways serve important roles in the CT­induced apoptosis of RA­FLSs.


Subject(s)
Apoptosis/drug effects , Arthritis, Rheumatoid/metabolism , Phenanthrenes/pharmacology , Reactive Oxygen Species/metabolism , Synoviocytes/drug effects , Synoviocytes/metabolism , Biomarkers , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Fibroblasts/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism
5.
Mol Med Rep ; 19(3): 1654-1664, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30592276

ABSTRACT

Derivatives of 1,4­naphthoquinone have excellent anti­cancer effects, but their use has been greatly limited due to their serious side effects. To develop compounds with decreased side effects and improved anti­cancer activity, two novel types of 1,4­naphthoquinone derivatives, 2,3­dihydro­2,3­epoxy­2­propylsulfonyl­5,8­dimethoxy­1,4­naphthoquinone (EPDMNQ) and 2,3­dihydro­2,3­epoxy­2­nonylsulfonyl­5,8­dimethoxy­1,4­naphthoquinone (ENDMNQ) were synthesized and their anti­tumor activities were investigated. The effects of EPDMNQ and ENDMNQ on cell viability, apoptosis and accumulation of reactive oxygen species (ROS) in liver cancer cells were determined by MTT cell viability assay and flow cytometry. The expression levels of mitochondrial, mitogen activated protein kinase (MAPK) and signal transducer and activator of transcription 3 (STAT3) signaling pathway­associated proteins in Hep3B liver cancer cells were analyzed by western blot analysis. The results demonstrated that EPDMNQ and ENDMNQ inhibited the proliferation of liver cancer Hep3B, HepG2, and Huh7 cell lines but not that of normal liver L­02, normal lung IMR­90 and stomach GES­1 cell lines. The number of apoptotic cells and ROS levels were significantly increased following treatment with EPDMNQ and ENDMNQ, and these effects were blocked by the ROS inhibitor N­acetyl­L­cysteine (NAC) in Hep3B cells. EPDMNQ and ENDMNQ induced apoptosis by upregulating the protein expression of p38 MAPK and c­Jun N­terminal kinase and downregulating extracellular signal­regulated kinase and STAT3; these effects were inhibited by NAC. The results of the present study demonstrated that EPDMNQ and ENDMNQ induced apoptosis through ROS­modulated MAPK and STAT3 signaling pathways in Hep3B cells. Therefore, these novel 1,4­naphthoquinone derivatives may be useful as anticancer agents for the treatment of liver cancer.


Subject(s)
Liver Neoplasms/drug therapy , Naphthoquinones/pharmacology , STAT3 Transcription Factor/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Mitochondria/genetics , Reactive Oxygen Species/metabolism
6.
Mol Med Rep ; 17(2): 2626-2634, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207064

ABSTRACT

Quinalizarin may be a potential chemical agent for cancer therapy, as it exerts anti­tumour effects against a variety of different types of cancer. However, the underlying regulatory mechanism and signalling pathways of quinalizarin in lung cancer cells remains unknown. The present study sought to investigate the effects of quinalizarin on proliferation, apoptosis and reactive oxygen species (ROS) generation in lung cancer. MTT assays were used to evaluate the effects of quinalizarin on the viability of lung cancer A549, NCI­H460 and NCI­H23 cells. Flow cytometry was employed to evaluate the effects of quinalizarin on the cell cycle, apoptosis and ROS generation in A549 cells. Western blotting was performed to detect cell cycle and apoptosis­associated protein expression levels in A549 cells. Quinalizarin inhibited A549, NCI­H460 and NCI­H23 cell proliferation and induced A549 cell cycle arrest at the G0/G1 phase. Quinalizarin induced apoptosis by upregulating the expression of B­cell lymphoma 2 (Bcl­2)­associated agonist of cell death, cleaved­caspase­3 and cleaved­poly (adenosine diphosphate­ribose) polymerase, and downregulating the expression of Bcl­2. Furthermore, quinalizarin activated mitogen­activated protein kinase (MAPK) and p53, and inhibited the protein kinase B and signal transducer and activator of transcription­3 (STAT3) signalling pathways. In addition, quinalizarin increased ROS generation. The ROS scavenger N­acetyl­L­cysteine restored quinalizarin­induced cell apoptosis, and inactivated the MAPK and STAT3 signalling pathways. The results of the present study demonstrated that quinalizarin induces G0/G1 phase cell cycle arrest and apoptosis via ROS mediated­MAPK and STAT3 signalling pathways.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , A549 Cells , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Humans , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
7.
Oncotarget ; 8(70): 115398-115412, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29383168

ABSTRACT

Cryptotanshinone (CT), isolated from the plant Salvia miltiorrhiza Bunge, has been reported to have potential anticancer effects on human prostate and breast cancer cells. However, the mechanisms of action of CT on gastric cancer (GC) cells are not well understood. Here we investigated the antitumor effects of CT on GC cells and its possible molecular mechanism. We found CT suppressed viability of twelve GC cell lines in a dose-dependent manner. CT induced cell cycle arrest at the G2/M phase and mitochondrial apoptosis accompanying the accumulation of reactive oxygen species (ROS). Pretreatment with ROS inhibitor N-acetyl-L-cysteine (NAC) blocked CT-induced apoptosis. CT increased p-JNK and p-p38, and decreased p-ERK and p-STAT3 protein expression, these effects were prevented by NAC. Furthermore, a xenograft assay showed that CT significantly inhibited MKN-45 cell-induced tumor growth in vivo by increasing expression of pro-apoptotic proteins (p-JNK, p-38 and cleaved-caspase-3) and reducing expression of anti-apoptotic proteins (p-ERK and p-STAT3) without adverse effects on nude mice weight. In conclusion, CT induced apoptosis and cell cycle arrest in GC cells via ROS-mediated MAPK and AKT signaling pathways, and this CT may be a useful compound for the developing anticancer agents for GC.

SELECTION OF CITATIONS
SEARCH DETAIL
...