Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Front Pharmacol ; 15: 1405521, 2024.
Article in English | MEDLINE | ID: mdl-39144617

ABSTRACT

Introduction: Almonertinib is an important third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) exhibiting high selectivity to EGFR-sensitizing and T790M-resistant mutations. Almonertinib resistance is a major obstacle in clinical use. Baicalein possesses antitumor properties, but its mechanism of antitumor action against almonertinib-resistant non-small cell lung cancer (NSCLC) remains unelucidated. Methods: CCK-8 assay was used to examine the survival rate of H1975/AR and HCC827/AR cells following treatment for 24 h with different concentrations of baicalein, almonertinib or their combination. The changes in colony formation ability, apoptosis, and intracellular reactive oxygen species (ROS) levels of the treated cells were analyzed using colony formation assay and flow cytometry. Western blotting was performed to detect the changes in protein expressions in the cells. The effects of pre-treatment with NAC on proliferation, apoptosis, and PI3K/Akt signaling pathway were observed in baicalein- and/or almonertinib-treated cells. A nude mouse model bearing subcutaneous HCC827/AR cell xenograft were treated with baicalein (20 mg/kg) or almonertinib (15 mg/kg), and the tumor volume and body mass changes was measured. Results: Both baicalein and almonertinib represses the viability of HCC827/AR and H1975/AR cells in a concentration-dependent manner. Compared with baicalein or almonertinib alone, the combined application of the two drugs dramatically attenuates cell proliferation; triggers apoptosis; causes cleavage of Caspase-3, PARP, and Caspase-9; downregulates the protein expressions of p-PI3K and p-Akt; and significantly inhibits tumor growth in nude mice. Furthermore, baicalein combined with almonertinib results in massive accumulation of reactive oxygen species (ROS) and preincubation with N-acetyl-L-cysteine (ROS remover) prevents proliferation as well as inhibits apoptosis induction, with partial recovery of the decline of p-PI3K and p-Akt. Discussion: The combination of baicalein and almonertinib can improve the antitumor activity in almonertinib-resistant NSCLC through the ROS-mediated PI3K/Akt pathway.

2.
Zhongguo Zhong Yao Za Zhi ; 42(12): 2361-2365, 2017 Jun.
Article in Chinese | MEDLINE | ID: mdl-28822194

ABSTRACT

Oridonin, which is an ent-kaurene diterpenoid isolated from traditional Chinese medicine Rabdosia rubescens, displays various bioactivities, including anti-inflammation, anti-bacteria and anti-tumor. This study aimed to investigate the effect of oridonin on apoptosis of triple-negative breast cancer MDA-MB-231 cells and its underlying mechanisms. The inhibitory effect of oridonin on proliferation of MDA-MB-231 cells was measured by MTT assay; Apoptosis was analyzed by flow cytometry with PI staining and Annexin V-FITC/PI staining; Intracellular reactive oxygen species (ROS) level was determined by ROS detection kit, and expressions of PARP, Bcl-2, caspase-3 were analyzed by Western blot. The results showed that oridonin exhibited a significant effect in inducing apoptosis of MDA-MB-231 cells, enhancing intracellular ROS level, down-regulating expression of Bcl-2 protein, and promoting cleavage of caspase-3 and its substrate PARP. These results indicated that the apoptosis-inducing effect of oridonin on MDA-MB-231 cells might be correlated with increase of intracellular ROS level, down-regulation of Bcl-2 protein and activation of caspase-3.


Subject(s)
Apoptosis/drug effects , Diterpenes, Kaurane/pharmacology , Reactive Oxygen Species/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Triple Negative Breast Neoplasms
3.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(7): 883-888, 2017 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-28736362

ABSTRACT

OBJECTIVE: To investigate the effect of small interfering RNA (siRNA)-mediated silencing of monocarboxylate transporter 1 (MCT1) on the sensitivity of drug-resistant nasopharyngeal carcinoma HNE1/DDP cells to cisplatin (DDP)-induced apoptosis and explore the possible mechanism. METHODS: The expression of MCT1 was analyzed in HNE1 and HNE1/DDP cells and in HNE1/DDP cells transfected with siRNA using Western blot. MTT assay was used to assess the inhibitory effect of different concentrations of DDP alone or in combination with MCT1 siRNA on the proliferation of HNE1/DDP cells. The apoptosis of cells treated with MCT1 siRNA or/and DDP (8 µmol/L) was assessed using flow cytometry with PI staining, and the mitochondrial membrane potential was detected using JC-1 staining assay; the expressions of Mcl-1, Bak, Bcl-2, and Bax were analyzed using Western blotting. RESULTS: HNE1/DDP cells showed a high expression of MCT1, and MCT1 silencing using siRNA significantly increased the sensitivity of HNE1/DDP cells to DDP (P<0.05) and partly reversed DDP resistance of the cells. MCT1 silencing enhanced the sensitivity of HNE1/DDP cells to DDP-induced apoptosis. Treatment of HNE1/DDP cells with MCT1 siRNA combined with 8 µmol/L DDP for 24 h resulted in an apoptotic rate of (51.23∓2.86)%, significantly higher than that in cells treated with MCT1 siRNA or DDP alone (P<0.05). The combined treatment also reduced the mitochondrial membrane potential, down-regulated the expression of Mcl-1 and Bcl-2, and up-regulated the expression of Bax in the DDP-resistant cells. CONCLUSION: MCT1 siRNA can enhance the sensitivity of HNE1/DDP cells to DDP-induced apoptosis, the mechanism of which may involve the down-regulation of Mcl-1 and Bcl-2 and up-regulation of Bax expression.

4.
Nan Fang Yi Ke Da Xue Xue Bao ; 35(1): 40-6, 2015 Jan.
Article in Chinese | MEDLINE | ID: mdl-25613607

ABSTRACT

OBJECTIVE: To evaluate the anticoagulant and antineoplastic activities of chemically modified low-molecular-weight heparin (LMWH). METHODS: LMWH obtained by splitting unfractionated heparin (UFH) with sodium periodate oxidation and sodium borohydride reduction was subjected to acetylation catalyzed by DCC and DMAP to produce acetylated LMWH (ALMWH). The anticoagulant activity of ALMWH was determined in mice, and its antiproliferative and anti-invasion activities was assessed in human breast cancer cells MDA-MB-231 and MFC-7. RESULTS: The anticoagulant activity of LMWH was decreased significantly after acetylation. The concentrations of commercial LMWH* and ALMWH for doubling the coagulation time (CT) were 33.04 µmol/L and 223.56 µmol/L, respectively, and the IC(50) of ALMWH for doubling CT was 6 times of that of LMWH*. ALMWH and LMWH at 0.1, 0.3, 0.9, 2.7 and 8.1 mmol/L both significantly inhibited the proliferation of MDA-MB-231 and MCF-7 cells in a concentration-dependent manner, but ALMWH produced stronger inhibitory effects. The IC(50) of LMWH and ALMWH for inhibiting cell proliferation was 3168.4 µmol/L and 152.6 µmol/L in MCF-7 cells, and 12299.6 µmol/L and 22.2 µmol/L in MDA-MB-231 cells, respectively. ALMWH and LMWH all markedly suppressed the invasion of MDA-MB-231 cells with comparable effects. CONCLUSION: Chemical modification of structure can endow LMWH with a low anticoagulant and high antiproliferative activities.


Subject(s)
Anticoagulants/chemistry , Antineoplastic Agents/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Heparin/chemistry , Animals , Anticoagulants/pharmacology , Antineoplastic Agents/chemistry , Blood Coagulation , Blood Coagulation Tests , Cell Line, Tumor , Heparin, Low-Molecular-Weight/chemistry , Humans , Mice
5.
Arch Pharm Res ; 38(6): 1066-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25193624

ABSTRACT

Bioassay-guided fractionation of the MeOH extract of Magnolia grandiflora seeds resulted in the isolation of a new dimeric neolignan, named bishonokiol A (1), as well as two known neolignans magnolol (2) and honokiol (3). The structures of the compounds were determined on the basis of data obtained using NMR and MS. Bishonokiol A (1) showed potent anti-proliferative activities in four human cancer cell lines, with IC50 values ranging from 5.1 to 7.5 µM. Additionally, bishonokiol A (1) induced apoptosis, as well as down-regulated the expression of the anti-apoptotic protein Bcl-2 and caspase-3 cleavage in HepG2 cell line.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Lignans/chemistry , Magnolia/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Biphenyl Compounds/chemistry , Biphenyl Compounds/isolation & purification , Caspase 3/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Humans , Lignans/isolation & purification , Lignans/pharmacology , Molecular Conformation , Plant Extracts/chemistry , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Seeds/chemistry
6.
J Microbiol Biotechnol ; 24(7): 914-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24705874

ABSTRACT

Triple-negative breast cancer (TNBC) possesses a higher rate of distant recurrence and a poorer prognosis than other breast cancer subtypes. Interestingly, most of the heat shock protein 90 (Hsp90) client proteins are oncoproteins, and some are closely related to unfavorable factors of TNBC patients. 17-Demethoxy-reblastatin (17-DR), a novel nonbenzoquinone- type geldanamycin analog, exhibited potent Hsp90 ATPase inhibition activity. In this study, the anticancer effects of 17-DR on TNBC MDA-MB-231 cells were investigated. These results showed that 17-DR inhibited cell proliferation, induced apoptosis, and suppressed cell invasion and migration in the MDA-MB-231 cells. Down-regulation of the key Hsp90-dependent tumor-driving molecules, such as RIP1 and MMP-9, by 17-DR may be related to these effects. Taken together, our results suggest that 17-DR has potential as a therapeutic agent for the treatment of TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Breast Neoplasms , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Cell Line, Tumor , Cell Physiological Phenomena/drug effects , Down-Regulation/drug effects , Female , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
7.
Anticancer Drugs ; 25(4): 447-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24492287

ABSTRACT

The hexokinase inhibitor 3-bromopyruvate (3-BrPA) can inhibit glycolysis in tumor cells to reduce ATP production, resulting in apoptosis. However, as 3-BrPA is an alkylating agent, its cytotoxic action may be induced by other molecular mechanisms. The results presented here reveal that 3-BrPA-induced apoptosis is caspase independent. Further, 3-BrPA induces the generation of reactive oxygen species in MDA-MB-231 cells, leading to mitochondria-mediated apoptosis. These results suggest that caspase-independent apoptosis may be induced by the generation of reactive oxygen species. In this study, we also demonstrated that 3-BrPA induces apoptosis through the downregulation of myeloid cell leukemia-1 (Mcl-1) in MDA-MB-231 breast cancer cells. The results of Mcl-1 knockdown indicate that Mcl-1 plays an important role in 3-BrPA-induced apoptosis. Further, the upregulation of Mcl-1 expression in 3-BrPA-treated MDA-MB-231 cells significantly increases cell viability. In addition, 3-BrPA treatment resulted in the downregulation of p-Akt, suggesting that 3-BrPA may downregulate Mcl-1 through the phosphoinositide-3-kinase/Akt pathway. These findings indicate that 3-BrPA induces apoptosis in breast cancer cells by downregulating Mcl-1 through the phosphoinositide-3-kinase/Akt signaling pathway.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Hexokinase/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pyruvates/pharmacology , Breast Neoplasms/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chromones/pharmacology , Down-Regulation , Female , Humans , Morpholines/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oncogene Protein v-akt/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(2): 351-5, 2013 Apr.
Article in Chinese | MEDLINE | ID: mdl-23628031

ABSTRACT

This study was purposed to investigate the effects of 2-deoxy-D-glucose (2-DG) on sensitizing HL-60 cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis and its possible mechanism. The proliferative inhibition of HL-60 cells treated with different concentrations of 2-DG and TRAIL was measured by MTT assay. The cells were treated with 2-DG, TRAIL, and 2-DG combined with TRAIL at the concentration < IC50 value, i.e. 10 mmol/L for 2-DG and 100 ng/ml for TRAIL. Apoptosis was analyzed by flow cytometry with PI staining; the expression of RIP1, GRP78, and PARP was analyzed by Western blot; the activity of caspase-3 was detected by special detection kit. The results showed that the combined treatment of HL-60 cells for 48 h induced an apoptotic rate of (45.1 ± 4.3)%, which was significantly higher than that of treated with 2-DG or TRAIL alone; at the same time, the combined treatment potentiated the expression of GRP78 and caspase-3 activity, and down-regulated the expression of RIP1. It is concluded that 2-DG can sensitize HL-60 cells to TRAIL-induced apoptosis, which may be correlated with excessive endoplasmic reticulum stress response, down-regulation of RIP1, and increase of caspase-3 activity.


Subject(s)
Apoptosis/drug effects , Deoxyglucose/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Caspase 3/metabolism , Endoplasmic Reticulum Chaperone BiP , HL-60 Cells , Heat-Shock Proteins/metabolism , Humans , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism
9.
Cancer Biol Ther ; 7(5): 651-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18245952

ABSTRACT

Urokinase (uPA) and its receptor (uPAR) play an important role in tumor growth and metastasis, and overexpression of these molecules is strongly correlated with poor prognosis in a variety of malignant tumors. In this study, ATF-Fc, an antibody-like molecule comprising the amino-terminal fragment of human uPA (ATF) linked to the Fc fragment of human IgG1 via a flexible linker was developed. Its antitumor activities were evaluated in vitro and in vivo. The results showed that ATF-Fc had obvious cytotoxic effect on several types of tumor cells, which is dependent on cellular expression of uPAR and its Fc fragment. Treatment with ATF-Fc caused a significant suppression on tumor growth and metastasis of xenograft human tumors (MCF-7 breast cancer and BGC-823 gastric cancer) in athymic nude mice. Furthermore, we demonstrated that ATF-Fc had an anti-angiogenesis activity both in vitro and in vivo. In conclusion, we provided a novel therapeutic antibody-like molecule in the management of a variety of solid tumors by disrupting the uPA/uPAR interaction.


Subject(s)
Immunoglobulin Fc Fragments/chemistry , Neoplasms/therapy , Receptors, Cell Surface/chemistry , Urokinase-Type Plasminogen Activator/chemistry , Amino Acid Sequence , Animals , Antibodies/chemistry , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Immunoglobulin G/chemistry , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Metastasis , Neovascularization, Pathologic , Receptors, Urokinase Plasminogen Activator
10.
J Natl Cancer Inst ; 99(20): 1551-5, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17925540

ABSTRACT

Tumor endothelial marker 8 (TEM8) was discovered as a cell membrane protein that is predominantly expressed in tumor endothelium and identified as a receptor for anthrax toxin. We developed an antibody-like molecule that consists of the protective antigen (PA)-binding domain of human TEM8 linked to the Fc portion of human immunoglobulin G1 (TEM8-Fc). This engineered protein bound to PA in a divalent cation-dependent manner and efficiently protected J774A.1 macrophage-like cells against anthrax toxin challenge in a dose-dependent manner. TEM8-Fc suppressed the growth and metastasis of xenograft human tumors in athymic nude mice (control versus 10 mg/kg TEM8-Fc, mean tumor weight: LS-180, 1.72 versus 0.16 g, difference = 1.56 g, 95% confidence interval [CI] = 0.96 to 2.16 g; P<.001; MCF-7, 1.12 versus 0.08 g, difference = 1.04 g, 95% CI = 0.77 to 1.31 g; P<.001; HepG2, 1.28 versus 0.35 g, difference = 0.93 g, 95% CI = 0.60 to 1.25 g; P<.001). Furthermore, TEM8 interacted with the M2 isoenzyme of pyruvate kinase (M2-PK), which has an important role in tumor growth and metastasis. TEM8-Fc is a novel therapeutic antibody-like agent in the management of solid tumors that may act by trapping M2-PK.


Subject(s)
Antineoplastic Agents/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/pharmacology , Membrane Proteins/drug effects , Membrane Proteins/pharmacology , Neoplasm Proteins/drug effects , Pyruvate Kinase/drug effects , Receptors, Cell Surface/drug effects , Animals , Antibodies , Breast Neoplasms/drug therapy , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Drug Design , Female , Humans , Immunoprecipitation , Liver Neoplasms/drug therapy , Male , Membrane Proteins/metabolism , Mice , Mice, Nude , Microfilament Proteins , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/pharmacology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL