1
|
Li Y, Liao Z, Wei X, Xiao X, Hu J. Epifriedelanol enhances adriamycin-induced cytotoxicity towards K562/ADM cells by down regulating of P-gp and MRP2. Xenobiotica 2022; 52:389-396. [PMID: 35582915 DOI: 10.1080/00498254.2022.2079441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
1. Multidrug resistance (MDR) is a critical issue during chemotherapy of cancers. Epifriedelanol (Epi) is the effective compounds from the Root Bark of Ulmus davidiana. This study aims to investigate the effect of Epi on MDR and its potential mechanism in the adriamycin (Adr)-resistant K562/ADM cells.2. The effect of Epi on MDR, P-glycoprotein (P-gp) and multidrug resistance-associated proteins (MRPs) were investigated in the adriamycin (Adr)-resistant K562/ADM cells. In addition, the alterations of nuclear receptor pregnane X receptor (PXR) and constitutive androstane receptor (CAR) mRNA expression levels in K562/ADM cells after Epi treatment were also examined.3. Epi significantly enhanced Adr-induced cytotoxicity towards K562/ADM cells. Combination of Epi and Adr can significantly reduce the 50% inhibitory concentration (IC50) of K562/ADM cells to Adr. The reversal fold was 1.83 and 3.64 after treated with Epi at 10 and 20 μM, respectively. The intracellular accumulation of Adr was significant increased after exposure to Epi at 5-20 μM compared with the control group. Furthermore, Epi treatment significantly decreased the mRNA and protein expression of P-gp and MRP2 in K562/ADM cells.4. The present study demonstrated that Epi could enhance Adr-induced cytotoxicity towards K562/ADM cells accompanied by the down-regulation of P-gp and MRP2.
Collapse
Affiliation(s)
- Yuhua Li
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhengzheng Liao
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiaohua Wei
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiong Xiao
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jinfang Hu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
2
|
Thorat ND, Bauer J, Tofail SAM, Gascón Pérez V, Bohara RA, Yadav HM. Silica nano supra-assembly for the targeted delivery of therapeutic cargo to overcome chemoresistance in cancer. Colloids Surf B Biointerfaces 2019; 185:110571. [PMID: 31683204 DOI: 10.1016/j.colsurfb.2019.110571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
Cancer cells become resistant over the period to chemotherapeutic drugs and pose a challenging impediment for oncologists in providing effective treatment. Nanomedicine allows to overcome chemoresistance and is the focus of our investigation. Silica nanostructures have been highlighted as an interesting drug delivery platform in vitro and in vivo applications. Here we show the validity of nanomedicine approach for targeted chemotherapeutic cargo delivery to overcome chemoresistance in cancer cells both in vitro and in vivo. For demonstrating the concept, we functionalised ∼100 nm long porous silica nanoparticles (∼20 nm diameter ordered pore structure) by conjugating anticancer drug, cytochrome c enzyme and dual-function anticancer aptamer AS1411 in single supra-assembled nanocargos. The supra-assembly on the porous silica nanostructure allows for a high loading of catalytic enzyme cytochrome c, anticancer drug and aptamer. The silica supra-assembly is characterized by transmission electron microscopy (TEM) and Brunauer-Emmett-Teller (BET) surface area analysis. Conjugation of cargoes has been monitored at each step by UV-vis and Fluorescence spectroscopy. Finally, the constructed supra-assembled nanocarrier tested on chemoresistance colon cancer (HCT116) cells. A pH-responsive, intracellular theranostic cargo delivery has been achieved and the triple action of the nanocargo made an efficient killing of drug resistance colon cancer cells in vitro (∼ 92% cell death) through triplex therapy effects by supressing the P-glycoprotein (P-gp) level. Furthermore, in vivo animal toxicity studies demonstrated, the supra-assembled nanocargos have encouraging safety index to be used in cancer therapy and drug delivery applications.
Collapse
Affiliation(s)
- Nanasaheb D Thorat
- Modelling Simulation and Innovative Characterisation (MOSAIC), Department of Physics and Bernal Institute, University of Limerick, Limerick, V94 T9PX, Ireland; Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, wybrzeże Stanisława Wyspiańskiego 27, Wrocław 50-370, Poland
| | - Joanna Bauer
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wroclaw University of Science and Technology, wybrzeże Stanisława Wyspiańskiego 27, Wrocław 50-370, Poland
| | - Syed A M Tofail
- Modelling Simulation and Innovative Characterisation (MOSAIC), Department of Physics and Bernal Institute, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Victoria Gascón Pérez
- Chemical Sciences Department, Bernal Institute, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Raghvendra A Bohara
- Centre for Interdisciplinary Research, D. Y. Patil University, Kolhapur, 416006, India; CURAM, Centre for Research in Medical Devices, National University of Ireland Galway, Ireland
| | - Hemraj M Yadav
- Department of Energy and Materials Engineering, Dongguk University, Seoul, 04620, South Korea.
| |
Collapse
|
3
|
Yin M, Su Z, Cui B, Han Y, Dai H, Yu X. A New Type of Nanogel Carrier based on Mixed Pluronic Loaded with Low-Dose Antitumor Drugs. JOURNAL OF WUHAN UNIVERSITY OF TECHNOLOGY-MATER. SCI. ED. 2019; 34:960-967. [DOI: 10.1007/s11595-019-2144-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/12/2018] [Indexed: 01/06/2025]
|
4
|
Tian L, Zhang Y, Wang Y, Dang R, Fu Z, Gu B, Wen N. Triptolide reduces proliferation and enhances apoptosis in drug-resistant human oral cancer cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1204-1213. [PMID: 31933935 PMCID: PMC6947074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/27/2018] [Indexed: 06/10/2023]
Abstract
Triptolide (TPL) is a traditional Chinese medicine that possesses anti-multidrug resistance (MDR) properties against various cancers, including oral cancer. However, the functional roles of TPL in oral cancer cells and its potential ability to overcome MDR have not fully evaluated. Therefore, in this study we used oral cancer cell line SAS to establish Taxol-resistant cell line SAS/Taxol and investigated the effects of TPL on MDR, proliferation, and apoptosis of SAS/Taxol cells. We first demonstrated that TPL overcame MDR in SAS/Taxol cells. In addition, TPL induced prominent proliferation inhibition, cell cycle arrest, and apoptosis of SAS/Taxol cells. Furthermore, the pro-apoptotic effect of TPL on SAS/Taxol cells was dependent on intrinsic and extrinsic apoptotic pathways involved in the activation of caspases. Consistently, TPL successfully hampered oral tumor growth by inducing cell apoptosis in a xenograft mouse model. Overall, these results indicated that TPL circumvented MDR of SAS/Taxol cells by inhibition of proliferation and induction of apoptosis which was partly mediated by the intrinsic and extrinsic apoptotic pathways, suggesting the potential therapeutic value of TPL on Taxol-resistant human oral cancer.
Collapse
Affiliation(s)
- Li Tian
- Department of Anesthesiology and Perioperative Medicine, State Key Discipline of Cell Biology, Xijing Hospital, The Fourth Military Medical UniversityXi’an 710018, Shaanxi, China
- Institute of Stomatology, Chinese PLA General HospitalBeijing 100039, China
| | - Yang Zhang
- Department of Orthopedics, State Key Discipline of Cell Biology, Xijing Hospital, The Fourth Military Medical UniversityXi’an 710018, Shaanxi, China
| | - Yu Wang
- Department of Oncology, State Key Discipline of Cell Biology, Xijing Hospital, The Fourth Military Medical UniversityXi’an 710018, Shaanxi, China
| | - Ruijie Dang
- Institute of Stomatology, Chinese PLA General HospitalBeijing 100039, China
| | - Zhiguang Fu
- Institute of Stomatology, Chinese PLA General HospitalBeijing 100039, China
| | - Bin Gu
- Institute of Stomatology, Chinese PLA General HospitalBeijing 100039, China
| | - Ning Wen
- Institute of Stomatology, Chinese PLA General HospitalBeijing 100039, China
| |
Collapse
|
5
|
Shang HS, Lu HF, Lee CH, Chiang HS, Chu YL, Chen A, Lin YF, Chung JG. Quercetin induced cell apoptosis and altered gene expression in AGS human gastric cancer cells. ENVIRONMENTAL TOXICOLOGY 2018; 33:1168-1181. [PMID: 30152185 DOI: 10.1002/tox.22623] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 05/20/2023]
Abstract
Quercetin is one of the natural components from natural plant and it induces cell apoptosis in many human cancer cell lines. However, no available reports show that quercetin induces apoptosis and altered associated gene expressions in human gastric cancer cells, thus, we investigated the effect of quercetin on the apoptotic cell death and associated gene expression in human gastric cancer AGS cells. Results indicated that quercetin induced cell morphological changes and reduced total viability via apoptotic cell death in AGS cells. Furthermore, results from flow cytometric assay indicated that quercetin increased reactive oxygen species (ROS) production, decreased the levels of mitochondrial membrane potential (ΔΨm ), and increased the apoptotic cell number in AGS cells. Results from western blotting showed that quercetin decreased anti-apoptotic protein of Mcl-1, Bcl-2, and Bcl-x but increased pro-apoptotic protein of Bad, Bax, and Bid. Furthermore, quercetin increased the gene expressions of TNFRSF10D (Tumor necrosis factor receptor superfamily, member 10d, decoy with truncated death domain), TP53INP1 (tumor protein p53 inducible nuclear protein 1), and JUNB (jun B proto-oncogene) but decreased the gene expression of VEGFB (vascular endothelial growth factor B), CDK10 (cyclin-dependent kinase 10), and KDELC2 (KDEL [Lys-Asp-Glu-Leu] containing 2) that are associated with apoptosis pathways. Thus, those findings may offer more information regarding the molecular, gene expression, and signaling pathway for quercetin induced apoptotic cell death in human gastric cancer cells.
Collapse
Affiliation(s)
- Hung-Sheng Shang
- Graduate Institute of Clinical of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsu-Feng Lu
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan
- Department of Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Ching-Hsiao Lee
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Han-Sun Chiang
- Graduate Institute of Basic Medicine, Fu-Jen Catholic University, New Taipei city, Taiwan
| | - Yung-Lin Chu
- International Master's Degree Program in Food Science, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Ann Chen
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yuh-Feng Lin
- Graduate Institute of Clinical of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Shuang Ho Hospital, New Taipei City, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
6
|
Broad targeting of triptolide to resistance and sensitization for cancer therapy. Biomed Pharmacother 2018; 104:771-780. [DOI: 10.1016/j.biopha.2018.05.088] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/06/2018] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
|
7
|
Ma YS, Yao CN, Liu HC, Yu FS, Lin JJ, Lu KW, Liao CL, Chueh FS, Chung JG. Quercetin induced apoptosis of human oral cancer SAS cells through mitochondria and endoplasmic reticulum mediated signaling pathways. Oncol Lett 2018; 15:9663-9672. [PMID: 29928342 PMCID: PMC6004715 DOI: 10.3892/ol.2018.8584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/17/2018] [Indexed: 12/27/2022] Open
Abstract
Oral cancer is a cause of cancer-associated mortality worldwide and the treatment of oral cancer includes radiation, surgery and chemotherapy. Quercetin is a component from natural plant products and it has been demonstrated that quercetin is able to induce cytotoxic effects through induction of cell apoptosis in a number of human cancer cell lines. However, there is no available information to demonstrate that quercetin is able to induce apoptosis in human oral cancer cells. In the present study, the effect of quercetin on the cell death via the induction of apoptosis in human oral cancer SAS cells was investigated using flow cytometry, Annexin V/propidium iodide (PI) double staining, western blotting and confocal laser microscopy examination, to test for cytotoxic effects at 6–48 h after treatment with quercetin. The rate of cell death increased with the duration of quercetin treatment based on the results of a cell viability assay, increased Annexin V/PI staining, increased reactive oxygen species and Ca2+ production, decreased the levels of mitochondrial membrane potential (ΔΨm), increased proportion of apoptotic cells and altered levels of apoptosis-associated protein expression in SAS cells. The results from western blotting revealed that quercetin increased Fas, Fas-Ligand, fas-associated protein with death domain and caspase-8, all of which associated with cell surface death receptor. Furthermore, quercetin increased the levels of activating transcription factor (ATF)-6α, ATF-6β and gastrin-releasing peptide-78 which indicated an increase in endoplasm reticulum stress, increased levels of the pro-apoptotic protein BH3 interacting-domain death antagonist, and decreased levels of anti-apoptotic proteins B-cell lymphoma (Bcl) 2 and Bcl-extra large which may have led to the decreases of ΔΨm. Additionally, confocal microscopy suggested that quercetin was able to increase the expression levels of cytochrome c, apoptosis-inducing factor and endonuclease G, which are associated with apoptotic pathways. Therefore, it is hypothesized that quercetin may potentially be used as a novel anti-cancer agent for the treatment of oral cancer in future.
Collapse
Affiliation(s)
- Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 84001, Taiwan, R.O.C.,Department of Chinese Medicine, E-Da Hospital, Kaohsiung 82445, Taiwan, R.O.C
| | - Chien-Ning Yao
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Hsin-Chung Liu
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Fu-Shun Yu
- School of Dentistry, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Jen-Jyh Lin
- Division of Cardiology, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Kung-Wen Lu
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Ching-Lung Liao
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Fu-Shin Chueh
- Department of Health and Nutrition Biotechnology, Asia University, Wufeng, Taichung 41354, Taiwan, R.O.C
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan, R.O.C.,Department of Biotechnology, Asia University, Wufeng, Taichung 41354, Taiwan, R.O.C
| |
Collapse
|
8
|
Toward targeted therapy in chemotherapy-resistant pancreatic cancer with a smart triptolide nanomedicine. Oncotarget 2016; 7:8360-72. [PMID: 26840019 PMCID: PMC4884998 DOI: 10.18632/oncotarget.7073] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023] Open
Abstract
Chemoresistance is the major impediment for treating pancreatic cancer. Herb-derived compound triptolide (TP) can inhibit proliferation of chemo-resistant pancreatic cancer (CPC) cell lines through multiple mechanisms, which exhibited superior anticancer efficacy compared with gemcitabine. However, toxicity due to non-specific exposure to healthy tissues hindered its clinical translation. Herein we successfully achieved targeting CPC cells and avoiding exposure to healthy tissues for TP by nucleolin-specific aptamer (AS1411) mediated polymeric nanocarrier. We conjugated AS1411 aptamer to carboxy terminated poly(ethylene glycol)–block–poly(d, l-lactide) (HOOC-PEG-PDLLA), then prepared AS1411-PEG-PDLLA micelle loading TP (AS-PPT) through solid dispersion technique. AS-PPT showed more antitumor activity than TP and equivalent specific binding ability with gemcitabine-resistant human pancreatic cancer cell (MIA PaCa-2) to AS1411 aptamer in vitro. Furthermore, we studied the distribution of AS-PPT (Cy3-labed TP) at tissue and cellular levels using biophotonic imaging technology. The results showed AS1411 facilitated TP selectively accumulating in tumor tissues and targeting CPC cells. The lifetime of the MIA PaCa-2 cell-bearing mice administrated with AS-PPT was efficiently prolonged than that of the mice subjected to the clinical anticancer drug Gemzar®in vivo. Such work provides a new strategy for overcoming the drug resistance of pancreatic cancer.
Collapse
|
9
|
Xie CQ, Zhou P, Zuo J, Li X, Chen Y, Chen JW. Triptolide exerts pro-apoptotic and cell cycle arrest activity on drug-resistant human lung cancer A549/Taxol cells via modulation of MAPK and PI3K/Akt signaling pathways. Oncol Lett 2016; 12:3586-3590. [PMID: 27900040 DOI: 10.3892/ol.2016.5099] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/09/2016] [Indexed: 01/06/2023] Open
Abstract
Multidrug resistance (MDR) is a major obstacle in the effective chemotherapeutic treatment of cancers. Triptolide (TPL) is a diterpenoid isolated from Tripterygium wilfordii Hook. f., a traditional Chinese medicine. It was demonstrated in our previous study that TPL exerts anti-MDR cancers on various MDR cell lines (including A549/Taxol, MCF-7/ADR and Bel7402/5-Fu). The present study was designed to investigate its anti-proliferative activity on A549/Taxol cells, and explore the underlying mechanism of action. The anti-proliferative activity of TPL on A549/Taxol cells was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Its pro-apoptosis and cell cycle arrest activities were analyzed by flow cytometry. Western blot assay was employed to investigate the levels of mitogen-activated protein kinases (MAPKs) and apoptosis-related proteins in cells. TPL efficiently suppressed the proliferation of A549/Taxol cells. Co-treatment with MAPK inhibitors in the MTT assay indicated that the extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) pathways were involved in the process. Upregulation of p-p38, p-ERK, p-GSK-3β, Bax and cleaved caspases-3 and -9, and downregulation of p-JNK, p-Akt and Bcl-2 were observed upon treatment with TPL in the A549/Taxol cells. The results from flow cytometry assay revealed that TPL induced apoptosis and S-phase arrest in A549/Taxol cells. This occurred as a result of the upregulation of p-ERK and p-GSK-3β, and the downregulation of p-JNK and p-Akt, and was responsible for the subsequent anti-proliferative activity.
Collapse
Affiliation(s)
- Chen Qiong Xie
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Ping Zhou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Jian Zuo
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Xiang Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China; Jiangsu Key Laboratory for Chinese Material Medical Processing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Yong Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| | - Jian Wei Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China; Jiangsu Key Laboratory for TCM Formulae Research, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, P.R. China
| |
Collapse
|
10
|
Jiang QW, Cheng KJ, Mei XL, Qiu JG, Zhang WJ, Xue YQ, Qin WM, Yang Y, Zheng DW, Chen Y, Wei MN, Zhang X, Lv M, Chen MW, Wei X, Shi Z. Synergistic anticancer effects of triptolide and celastrol, two main compounds from thunder god vine. Oncotarget 2016; 6:32790-804. [PMID: 26447544 PMCID: PMC4741730 DOI: 10.18632/oncotarget.5411] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/25/2015] [Indexed: 02/07/2023] Open
Abstract
Triptolide and celastrol are two main active compounds isolated from Thunder God Vine with the potent anticancer activity. However, the anticancer effect of triptolide in combination with celastrol is still unknown. In the present study, we demonstrated that the combination of triptolide with celastrol synergistically induced cell growth inhibition, cell cycle arrest at G2/M phase and apoptosis with the increased intracellular ROS accumulation in cancer cells. Pretreatment with ROS scavenger N-acetyl-L-cysteine dramatically blocked the apoptosis induced by co-treatment with triptolide and celastrol. Treatment with celastrol alone led to the decreased expressions of HSP90 client proteins including survivin, AKT, EGFR, which was enhanced by the addition of triptolide. Additionally, the celastrol-induced expression of HSP70 and HSP27 was abrogated by triptolide. In the nude mice with xenograft tumors, the lower-dose combination of triptolide with celastrol significantly inhibited the growth of tumors without obvious toxicity. Overall, triptolide in combination with celastrol showed outstanding synergistic anticancer effect in vitro and in vivo, suggesting that this beneficial combination may offer a promising treatment option for cancer patients.
Collapse
Affiliation(s)
- Qi-Wei Jiang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Ke-Jun Cheng
- Chemical Biology Center, Lishui Institute of Agricultural Sciences, Lishui, Zhejiang, China.,National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiao-Long Mei
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Jian-Ge Qiu
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Wen-Ji Zhang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - You-Qiu Xue
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Wu-Ming Qin
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Yang Yang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Di-Wei Zheng
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Yao Chen
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Meng-Ning Wei
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xu Zhang
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Min Lv
- Institute of Materia Medica, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mei-Wan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xing Wei
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Wu X, Ma J, Ye Y, Lin G. Transporter modulation by Chinese herbal medicines and its mediated pharmacokinetic herb–drug interactions. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1026:236-253. [DOI: 10.1016/j.jchromb.2015.11.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 10/22/2022]
|
12
|
Zhang Y, Li J, Lei X, Zhang T, Liu G, Yang M, Liu M. Influence of Verapamil on Pharmacokinetics of Triptolide in Rats. Eur J Drug Metab Pharmacokinet 2015; 41:449-56. [DOI: 10.1007/s13318-015-0275-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
13
|
pH-sensitive pullulan-based nanoparticle carrier for adriamycin to overcome drug-resistance of cancer cells. Carbohydr Polym 2014; 111:908-17. [DOI: 10.1016/j.carbpol.2014.05.057] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/13/2014] [Accepted: 05/13/2014] [Indexed: 12/23/2022]
|
14
|
Li XJ, Jiang ZZ, Zhang LY. Triptolide: progress on research in pharmacodynamics and toxicology. JOURNAL OF ETHNOPHARMACOLOGY 2014; 155:67-79. [PMID: 24933225 DOI: 10.1016/j.jep.2014.06.006] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/05/2014] [Accepted: 06/05/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii Hook. f. (Tripterygium wilfordii), also known as Huangteng and gelsemium elegan, is a traditional Chinese medicine that has been marketed in China as Tripterygium wilfordii glycoside tablets. Triptolide (TP), an active component in Tripterygium wilfordii extracts, has been used to treat various diseases, including lupus, cancer, rheumatoid arthritis and nephritic syndrome. This review summarizes recent developments in the research on the pharmacodynamics, pharmacokinetics, pharmacy and toxicology of TP, with a focus on its novel mechanism of reducing toxicity. This review provides insight for future studies on traditional Chinese medicine, a field that is both historically and currently important. MATERIALS AND METHODS We included studies published primarily within the last five years that were available in online academic databases (e.g., PubMed, Google Scholar, CNKI, SciFinder and Web of Science). RESULTS TP has a long history of use in China because it displays multiple pharmacological activities, including anti-rheumatism, anti-inflammatory, anti-tumor and neuroprotective properties. It has been widely used for the treatment of various diseases, such as rheumatoid arthritis, nephritic syndrome, lupus, Behcet׳s disease and central nervous system diseases. Recently, numerous breakthroughs have been made in our understanding of the pharmacological efficacy of TP. Although TP has been marketed as a traditional Chinese medicine, its multi-organ toxicity prevents it from being widely used in clinical practice. CONCLUSIONS Triptolide, a biologically active natural product extracted from the root of Tripterygium wilfordii, has shown promising pharmacological effects, particularly as an anti-tumor agent. Currently, in anti-cancer research, more effort should be devoted to investigating effective anti-tumor targets and confirming the anti-tumor spectrum and clinical indications of novel anti-tumor pro-drugs. To apply TP appropriately, with high efficacy and low toxicity, the safety and non-toxic dose range for specific target organs and diseases should be determined, the altered pathways and mechanisms of exposure need to be clarified, and an early warning system for toxicity needs to be established. With further in-depth study of the efficacy and toxicity of TP, we believe that TP will become a promising multi-use drug with improved clinical efficacy and safety in the future.
Collapse
Affiliation(s)
- Xiao-Jiaoyang Li
- Jiangsu Center of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhen-Zhou Jiang
- Jiangsu Center of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China.
| | - Lu-yong Zhang
- Jiangsu Center of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
15
|
Hsp90 inhibitor BIIB021 enhances triptolide-induced apoptosis of human T-cell acute lymphoblastic leukemia cells in vitro mainly by disrupting p53-MDM2 balance. Acta Pharmacol Sin 2013; 34:1545-53. [PMID: 24241349 DOI: 10.1038/aps.2013.124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/02/2013] [Indexed: 01/02/2023] Open
Abstract
AIM To investigate the effects of BIIB021, an inhibitor of heat shock protein 90 (Hsp90) alone or in combination with triptolide (TPL) on T-cell acute lymphoblastic leukemia (T-ALL) and the mechanisms of action. METHODS Human T-ALL cells line Molt-4 was examined. The cell viability was measured using MTT assay. Apoptotic cells were studied with Hoechst 33258 staining. Cell apoptosis and cell cycle were analyzed using flow cytometry with Annexin V/PI staining and PI staining, respectively. The levels of multiple proteins, including Akt, p65, CDK4/6, p18, Bcl-2 family proteins, MDM2, and p53, were examined with Western blotting. The level of MDM2 mRNA was determined using RT-PCR. RESULTS Treatment of Molt-4 cells with BIIB021 (50-800 nmol/L) inhibited the cell growth in a dose-dependent manner (the IC50 value was 384.6 and 301.8 nmol/L, respectively, at 48 and 72 h). BIIB021 dose-dependently induced G0/G1 phase arrest, followed by apoptosis of Molt-4 cells. Furthermore, BIIB021 increased the expression of p18, decreased the expression of CDK4/6, and activated the caspase pathway in Molt-4 cells. Moreover, BIIB021 (50-400 nmol/L) dose-dependently decreased the phospho-MDM2 and total MDM2 protein levels, but slightly increased the phospho-p53 and total p53 protein levels, whereas TPL (5-40 nmol/L) dose-dependently enhanced p53 activation without affecting MDM2 levels. Co-treatment with BIIB021 and TPL showed synergic inhibition on Molt-4 cell growth. The co-treatment disrupted p53-MDM2 balance, thus markedly enhanced p53 activation. In addition, the co-treatment increased the expression of Bak and Bim, followed by increased activation of caspase-9. CONCLUSION The combination of BIIB021 and TPL may provide a novel strategy for treating T-ALL by overcoming multiple mechanisms of apoptosis resistance.
Collapse
|
16
|
Lu F, Hou YQ, Song Y, Yuan ZJ. TFPI-2 downregulates multidrug resistance protein in 5-FU-resistant human hepatocellular carcinoma BEL-7402/5-FU cells. Anat Rec (Hoboken) 2012; 296:56-63. [PMID: 23125179 DOI: 10.1002/ar.22611] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 08/22/2012] [Indexed: 12/20/2022]
Abstract
Tissue factor pathway inhibitor-2 (TFPI-2) is known to induce apoptosis and to suppress tumor metastasis in several types of cancer cells. However, there is little known about its reversal effect on chemoresistant tumor cells. This study investigated the effect of TFPI-2 in 5-fluorouracil (5-FU)-resistant human hepatocellular cancer BEL-7402/5-FU cells in vitro. We constructed TFPI-2 overexpression BEL-7402/5-FU cell lines and explored resistance index (RI) of 5-FU, function of the P-glycoprotein (P-gp) efflux pump, and the mRNA and protein expression of drug resistance gene, including multidrug resistance gene (MDR1), lung-resistance protein (LRP), multidrug resistance-associated protein (MRP1), glutathione-S-transferase-π (GST-π), excision repair cross-complementing gene 1 (ERCC1), and p38 phosphorylation. We found that TFPI-2 improved the RI of 5-FU and inhibited P-gp function. Western blotting and real-time PCR revealed that TFPI-2 also decreased mRNA and protein expression of MDR1, LRP, MRP1, GST-π, and ERCC1, whereas p38 phosphorylation was increased. We considered that TFPI-2 reduces 5-FU resistance in BEL-7402/5-FU cells, and the mechanism appears to involve p38-mediated downregulation of drug resistance gene expression such as MDR1, LRP, MRP1, GST-π, and ERCC1.
Collapse
Affiliation(s)
- Fei Lu
- Microsurgical Ward Section, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | | | | | | |
Collapse
|