1
|
Mohammadi AH, Bagheri F, Baghaei K. Chondroitin sulfate-tocopherol succinate modified exosomes for targeted drug delivery to CD44-positive cancer cells. Int J Biol Macromol 2024:133625. [PMID: 39084997 DOI: 10.1016/j.ijbiomac.2024.133625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Exosomes (Exos), natural nanovesicles released by various cell types, show potential as an effective drug delivery platform due to their intrinsic role as transporters of biomolecules between different cells. However, Exos functionalization with targeting ligands is a critical step to enhance their targeting capability, which could be challenging. In this study, Exos were modified to specifically bind to CD44-positive cells by anchoring chondroitin sulfate (CS) to their surface. Exo modification was facilitated with CS conjugation with alpha-tocopherol succinate (TOS) as an anchorage. The modified Exos were utilized for delivering curcumin (Cur) to pancreatic cancer (PC) cells. In vitro Cur release studies revealed that Exos play a crucial role in maintaining Cur within themselves, demonstrating their potential as effective carriers for drug delivery to targeted locations. Notably, Cur loaded into the modified Exos exhibited enhanced cytotoxicity compared to unmodified Exo-Cur. Meanwhile, Exo-Cur-TOS-CS induced apoptosis more effectively in AsPC-1 cells than unmodified Exos (70.2 % versus 56.9 %). It is worth mentioning that with CD44-mediated cancer-specific targeting, Exo-CS enabled increased intracellular accumulation in AsPC-1 cells, showing promise as a targeted platform for cancer therapy. These results confirm that Exo modification has a positive impact on enhancing the therapeutic efficacy and cytotoxicity of drugs.
Collapse
Affiliation(s)
- Amir Hossein Mohammadi
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran.
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia.
| |
Collapse
|
2
|
Alavijeh RK, Akhbari K. Improved Cytotoxicity and Induced Apoptosis in HeLa Cells by Co-loading Vitamin E Succinate and Curcumin in Nano-MIL-88B-NH 2. Chembiochem 2023; 24:e202300415. [PMID: 37553295 DOI: 10.1002/cbic.202300415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/10/2023]
Abstract
One of the strategies for improved therapeutic effects in cancer therapy is combination chemotherapy. In this study, a flexible nano-MOF (Fe-MIL-88B-NH2 ) was synthesized in a sonochemical process, then co-loaded with α-tocopheryl succinate (TOS) and curcumin (CCM). The anticancer activity of co-loaded Fe-MIL-88B-NH2 (Fe-MIL-88B-NH2 /TOS@CCM) against the HeLa cells was compared with that of the single-loaded counterpart (Fe-MIL-88B-NH2 @CCM). MTT analysis indicates improved cytotoxicity of Fe-MIL-88B-NH2 /TOS@CCM. The data from the cell apoptosis assay indicated more apoptosis in the case of the co-loaded nano-MOF. This study indicates the positive effect of the presence of TOS on enhancing the anticancer effect of Fe-MIL-88B-NH2 @CCM to prepare a more efficient drug delivery nanosystem.
Collapse
Affiliation(s)
- Roya Karimi Alavijeh
- School of Chemistry, College of Science, University of Tehran, Tehran, 14155-6455, Iran
| | - Kamran Akhbari
- School of Chemistry, College of Science, University of Tehran, Tehran, 14155-6455, Iran
| |
Collapse
|
3
|
Taniguchi S, Ono Y, Doi Y, Taniguchi S, Matsuura Y, Iwasaki A, Hirata N, Fukuda R, Inoue K, Yamaguchi M, Tashiro A, Egami D, Aoki S, Kondoh Y, Honda K, Osada H, Kumeta H, Saio T, Okiyoneda T. Identification of α-Tocopherol succinate as an RFFL-substrate interaction inhibitor inducing peripheral CFTR stabilization and apoptosis. Biochem Pharmacol 2023; 215:115730. [PMID: 37543348 DOI: 10.1016/j.bcp.2023.115730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/06/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023]
Abstract
The E3 ubiquitin ligase RFFL is an apoptotic inhibitor highly expressed in cancers and its knockdown suppresses cancer cell growth and sensitizes to chemotherapy. RFFL also participates in peripheral protein quality control which removes the functional cell surface ΔF508-CFTR channel and reduces the efficacy of pharmaceutical therapy for cystic fibrosis (CF). Although RFFL inhibitors have therapeutic potential for both cancer and CF, they remain undiscovered. Here, a chemical array screening has identified α-tocopherol succinate (αTOS) as an RFFL ligand. NMR analysis revealed that αTOS directly binds to RFFL's substrate-binding region without affecting the E3 enzymatic activity. Consequently, αTOS inhibits the RFFL-substrate interaction, ΔF508-CFTR ubiquitination and elimination from the plasma membrane of epithelial cells, resulting in the increased functional CFTR channel. Among the α-tocopherol (αTOL) analogs we tested, only αTOS inhibited the RFFL-substrate interaction and increased the cell surface ΔF508-CFTR, depending on RFFL expression. Similarly, the unique proapoptotic effect of αTOS was dependent on RFFL expression. Thus, unlike other αTOL analogs, αTOS acts as an RFFL protein-protein interaction inhibitor which may explain its unique biological properties among αTOL analogs. Moreover, αTOS may act as a CFTR stabilizer, a novel class of drugs that extend cell surface ΔF508-CFTR lifetime.
Collapse
Affiliation(s)
- Sachiho Taniguchi
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Yuji Ono
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Yukako Doi
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Shogo Taniguchi
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Yuta Matsuura
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Ayuka Iwasaki
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Noriaki Hirata
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Ryosuke Fukuda
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan
| | - Keitaro Inoue
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Fukuoka 820-8502, Japan
| | - Miho Yamaguchi
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Fukuoka 820-8502, Japan
| | - Anju Tashiro
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Fukuoka 820-8502, Japan
| | - Daichi Egami
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Fukuoka 820-8502, Japan
| | - Shunsuke Aoki
- Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, Fukuoka 820-8502, Japan
| | - Yasumitsu Kondoh
- Chemical Resource Development Unit, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| | - Kaori Honda
- Chemical Resource Development Unit, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| | - Hiroyuki Osada
- Chemical Resource Development Unit, RIKEN Center for Sustainable Resource Science, Saitama 351-0198, Japan
| | - Hiroyuki Kumeta
- Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Tomohide Saio
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Tsukasa Okiyoneda
- Department of Biomedical Sciences, School of Biological and Environmental Sciences, Kwansei Gakuin University, Hyogo 669-1337, Japan.
| |
Collapse
|
4
|
Boratto FA, Lages EB, Loures CMC, Sabino AP, Malachias A, Townsend DM, Branco De Barros AL, Miranda Ferreira LA, Amaral Leite E. Alpha-tocopheryl succinate and doxorubicin-loaded liposomes improve drug uptake and tumor accumulation in a murine breast tumor model. Biomed Pharmacother 2023; 165:115034. [PMID: 37356372 PMCID: PMC10720879 DOI: 10.1016/j.biopha.2023.115034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023] Open
Abstract
Liposomes composed of a rigid bilayer have high plasma stability; however, they can be challenged in efficacy due to complications in releasing the encapsulated drug as well as being internalized by the tumor cell. On the other hand, fusogenic liposomes may fuse with the plasmatic membrane and release encapsulated material directly into the cytoplasm. In a previous study, fusogenic liposomes composed of alpha-tocopheryl succinate (TS) and doxorubicin (DOX) were developed (pHSL-TS-DOX). These stabilized tumor growth and reduced toxicity compared to a commercial formulation. In the present study, we investigated whether cellular uptake or DOX accumulation in the tumor could justify the better performance of the pHSL-TS-DOX formulation. Release, deformability, and DOX plasmatic concentration studies were also carried out. pHSL-TS-DOX showed an adequate release profile and demonstrated characteristics of a deformable formulation. Data from apoptosis, cell cycle, and nuclear morphology studies have shown that the induction of cell death caused by pHSL-TS-DOX occurred more quickly. Higher DOX cellular uptake and tumor accumulation were observed when pHSL-TS-DOX was administered, demonstrating better drug delivery capacity. Therefore, better DOX uptake as well as tumor accumulation explain the great antitumor activity previously demonstrated for this formulation.
Collapse
Affiliation(s)
- Fernanda A Boratto
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo B Lages
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cristina M C Loures
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adriano P Sabino
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Angelo Malachias
- Department of Physics, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Danyelle M Townsend
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Andre Luis Branco De Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Lucas Antonio Miranda Ferreira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine Amaral Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
5
|
Sheng Q, Yuan M, Wang D, Kou Y, Liu L, Chen Y, Song S. Injectable Hydrogels of Amphiphilic Vitamin E Derivatives for Locoregional Chemotherapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:11839-11850. [PMID: 37561909 DOI: 10.1021/acs.langmuir.3c01576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Vitamin E derivatives are particularly effective in chemotherapy drug development because they are nontoxic, biocompatible, and selective. Among them, α-tocopheryl succinate (α-TOS) can act synergistically with some chemotherapeutic agents. However, its hydrophobicity limits its systemic administration, and localized formulations are not available. Herein, we developed an injectable hydrogel based on self-assembled micelles of a triblock amphiphilic derivative of α-TOS (PEG-2VES), in which doxorubicin (DOX) was encapsulated in the core of the micelles for combined chemotherapy. A molecule of α-TOS was grafted onto each end of poly(ethylene glycols) (PEGs) of different lengths. Hydrogels were prepared by dissolving the polymers or the DOX-loaded micelles in water at room temperature. The subcutaneously injected hydrogels kept their shape and sustainably released the payloads over 7 days without any noticeable inflammatory response. In vitro and in vivo results confirmed the synergistic antitumor effects of the hydrogel and loaded drug. Furthermore, DOX-loaded hydrogels showed greater therapeutic efficiency and fewer toxic side effects than DOX alone. Overall, this hydrogel acts as a multifunctional system that can deliver drug, improve the therapeutic effect, and minimize drug toxicity.
Collapse
Affiliation(s)
- Qianli Sheng
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Min Yuan
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Dan Wang
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Yuanqi Kou
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Lei Liu
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Yan Chen
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Shiyong Song
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| |
Collapse
|
6
|
Chen X, Guo L, Ma S, Sun J, Li C, Gu Z, Li W, Guo L, Wang L, Han B, Chang J. Construction of multi-program responsive vitamin E succinate-chitosan-histidine nanocarrier and its response strategy in tumor therapy. Int J Biol Macromol 2023; 246:125678. [PMID: 37414317 DOI: 10.1016/j.ijbiomac.2023.125678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
Multifunctional drug delivery carriers have emerged as a promising cancer drug delivery strategy. Here, we developed a vitamin E succinate-chitosan-histidine (VCH) multi-program responsive drug carrier. The structure was characterized by FT-IR and 1H NMR spectrum, and the DLS and SEM results showed typical nanostructures. The drug loading content was 21.0 % and the corresponding encapsulation efficiency was 66.6 %. The UV-vis and fluorescence spectra demonstrated the existence of the π-π stacking interaction between DOX and VCH. Drug release experiments implied good pH sensitivity and sustained-release effect. The DOX/VCH nanoparticles could be efficiently taken up by HepG2 cancer cells and the tumor inhibition rate was up to 56.27 %. The DOX/VCH reduced the tumor volume and weight efficiently with a TIR of 45.81 %. The histological analysis results showed that DOX/VCH could effectively inhibit tumor growth and proliferation, and there was no damage to normal organs. VCH nanocarriers could combine the advantages of VES, histidine and chitosan to achieve pH sensitivity and P-gp inhibition, and effectively improve the drug solubility, targeting and lysosomal escape. Through the program response of different micro-environment, the newly developed polymeric micelles could successfully be utilized as a multi-program responsive nanocarrier system for the treatment of cancers.
Collapse
Affiliation(s)
- Xiaotong Chen
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Lan Guo
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Saibo Ma
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Jishang Sun
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Cuiyao Li
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Zhiyang Gu
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Wenya Li
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Lili Guo
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Litong Wang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Baoqin Han
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266235, PR China
| | - Jing Chang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266235, PR China.
| |
Collapse
|
7
|
Fernandes RS, Arribada RG, Silva JO, Silva-Cunha A, Townsend DM, Ferreira LAM, Barros ALB. In Vitro and In Vivo Effect of pH-Sensitive PLGA-TPGS-Based Hybrid Nanoparticles Loaded with Doxorubicin for Breast Cancer Therapy. Pharmaceutics 2022; 14:2394. [PMID: 36365212 PMCID: PMC9696591 DOI: 10.3390/pharmaceutics14112394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/14/2022] [Accepted: 11/03/2022] [Indexed: 09/06/2023] Open
Abstract
Doxorubicin (DOX) is an antineoplastic agent clinically employed for treating breast cancer patients. Despite its effectiveness, its inherent adverse toxic side effects often limit its clinical application. To overcome these drawbacks, lipid-polymer hybrid nanoparticles (LPNP) arise as promising nanoplatforms that combine the advantages of both liposomes and polymeric nanoparticles into a single delivery system. Alpha-tocopherol succinate (TS) is a derivative of vitamin E that shows potent anticancer mechanisms, and it is an interesting approach as adjuvant. In this study, we designed a pH-sensitive PLGA-polymer-core/TPGS-lipid-shell hybrid nanoparticle, loaded with DOX and TS (LPNP_TS-DOX). Nanoparticles were physicochemically and morphologically characterized. Cytotoxicity studies, migration assay, and cellular uptake were performed in 4T1, MCF-7, and MDA-MB-231 cell lines. Antitumor activity in vivo was evaluated in 4T1 breast tumor-bearing mice. In vitro studies showed a significant reduction in cell viability, cell migration, and an increase in cellular uptake for the 4T1 cell line compared to free DOX. In vivo antitumor activity showed that LPNP-TS-DOX was more effective in controlling tumor growth than other treatments. The high cellular internalization and the pH-triggered payload release of DOX lead to the increased accumulation of the drugs in the tumor area, along with the synergic combination with TS, culminating in greater antitumor efficacy. These data support LPNP-TS-DOX as a promising drug delivery system for breast cancer treatment.
Collapse
Affiliation(s)
- Renata S. Fernandes
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, Brazil
| | - Raquel Gregório Arribada
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, Brazil
| | - Juliana O. Silva
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, Brazil
| | - Armando Silva-Cunha
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, Brazil
| | - Danyelle M. Townsend
- Department of Drug Discovery and Pharmaceutical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lucas A. M. Ferreira
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, Brazil
| | - André L. B. Barros
- Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte 31270-901, Brazil
| |
Collapse
|
8
|
Nutraceutical-Based Nanoformulations for Breast and Ovarian Cancer Treatment. Int J Mol Sci 2022; 23:ijms231912032. [PMID: 36233349 PMCID: PMC9569679 DOI: 10.3390/ijms231912032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
Different strategies have been investigated for a more satisfactory treatment of advanced breast cancer, including the adjuvant use of omega-3 polyunsaturated fatty acids (PUFAs). These nutritional compounds have been shown to possess potent anti-inflammatory and antiangiogenic activities, the capacity to affect transduction pathways/receptors involved in cell growth and to reprogram tumor microenvironment. Omega-3 PUFA-containing nanoformulations designed for drug delivery in breast cancer were shown to potentiate the effects of enclosed drugs, enhance drug delivery to target sites, and minimize drug-induced side effects. We have critically analyzed here the results of the most recent studies investigating the effects of omega-3 PUFA-containing nanoformulations in breast cancer. The anti-neoplastic efficacy of omega-3 PUFAs has also been convincingly demonstrated by using preclinical in vivo models of ovarian cancer. The results obtained are critically analyzed here and seem to provide a sufficient rationale to move to still lacking interventional clinical trials, as well as to evaluate possible advantages of enclosing omega-3 PUFAs to drug-delivery nanosystems for ovarian cancer. Future perspectives in this area are also provided.
Collapse
|
9
|
Co-delivery of doxorubicin and CRISPR/Cas9 or RNAi-expressing plasmid by chitosan-based nanoparticle for cancer therapy. Carbohydr Polym 2022; 287:119315. [DOI: 10.1016/j.carbpol.2022.119315] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/04/2022] [Accepted: 03/02/2022] [Indexed: 12/21/2022]
|
10
|
Zhou F, Ding W, Mao Q, Jiang X, Chen J, Zhao X, Xu W, Huang J, Zhong L, Sun X. The regulation of hsacirc_004413 promotes proliferation and drug resistance of gastric cancer cells by acting as a competing endogenous RNA for miR-145-5p. PeerJ 2022; 10:e12629. [PMID: 35415017 PMCID: PMC8995023 DOI: 10.7717/peerj.12629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/22/2021] [Indexed: 01/07/2023] Open
Abstract
Background Whether circRAN, which acts as a microRNA sponge, plays a role in 5-fluorouracil (5-Fu) resistant gastric cancer has not been reported. In this study, a 5-Fu resistant cell line with an IC50 of 16.59 µM was constructed. Methods Using comparative analysis of circRNA in the transcriptomics of resistant and sensitive strains, 31 differentially expressed circRNAs were detected, and the microRNA interacting with them was predicted. Results Hsacirc_004413 was selected for verification in drug resistant and sensitive cells. By interfering with hsacirc_004413 using antisense RNA, the sensitivity of drug resistant cells to 5-Fu was significantly promoted, and the apoptosis and necrosis of the cells were significantly increased. In sensitive cells, inhibition by inhibitors enhanced the resistance of cells to 5-Fu. We hypothesize that hsacirc_004413 makes gastric cancer cells resistant to 5-Fu mainly through adsorption of miR-145-5p.
Collapse
Affiliation(s)
- Fusheng Zhou
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Weiqun Ding
- Department of Gastroenterology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiqi Mao
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Xiaoyun Jiang
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Jiajie Chen
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Xianguang Zhao
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Weijia Xu
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Jiaxin Huang
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Liang Zhong
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Xu Sun
- Department of Gastroenterology, Huashan Hospital North, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Shen H, Gao Q, Liu T, Wang H, Zhang R, Zhou J, Ding S, Ye Y, Sun Z. Self-assembled tocopherol-albumin nanoparticles with full biocompatibility for chemo-photothermal therapy against breast cancer. Curr Drug Deliv 2021; 19:49-63. [PMID: 34967289 DOI: 10.2174/1567201819666211229120611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/15/2021] [Accepted: 12/19/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The combination of photothermal therapy (PTT) and chemotherapy has proven to be a promising strategy for cancer treatment. Various nanomaterials have shown great potential in combination therapy, including gold, graphene oxide, iron oxide, and other nanoparticles. However, their undefinable toxicity in vivo greatly slowed down their development for clinical applications. OBJECTIVE The present work aimed to develop a multifunctional nanoparticle for chemo-photothermal therapy composed of acknowledged biocompatible materials. METHODS A novel biocompatible nanoparticle (HIT-NPs) was self-assembled through the intrinsic interaction between D-α-tocopherol Succinate (TOS), human serum albumin (HSA) and indocyanine green (ICG). Doxorubicin (DOX) was then loaded due to the ion pairing between DOX and TOS. The feasibility of combined chemo-photothermal therapy induced by DOX-loaded HIT-NPs was carefully evaluated. RESULTS In vitro, HIT-NPs showed no cytotoxicity on human normal liver cells (HL-7702 cells) but obvious killing effects murine breast cancer cells (4T1 cells). The combined chemo-photothermal therapeutic effect on 4T1 cells was successfully obtained. DOX-loaded HIT-NPs could effectively accumulate in 4T1 subcutaneous tumors after intravenous injection, and the tumor temperature rapidly increased under laser exposure, indicating the feasibility of PTT in vivo. CONCLUSION The self-assembled HIT-NPs could provide a promising platform for combined chemo-photothermal cancer therapy with full biocompatibility.
Collapse
Affiliation(s)
- Haijun Shen
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Qianqian Gao
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Tingting Liu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Haoran Wang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ran Zhang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jie Zhou
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Sihui Ding
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhenhua Sun
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| |
Collapse
|
12
|
The apoptotic efficacy of succinic acid on renal cancer cell lines. Med Oncol 2021; 38:144. [PMID: 34687367 DOI: 10.1007/s12032-021-01577-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
Abstract
Recently, studies on the effects of non-toxic substances on cancer prophylaxis have gained value as an alternative to existing treatment options. Current studies have shown that succinic acid or its derivatives exhibit anticancer activity by inducing apoptosis. We aimed to investigate the anticancer activity of succinic acid on renal cancer for the first time in the literature. The cytotoxic activity of succinic acid on CAKI-2 and ACHN as renal cancer cell lines and MRC-5 as a healthy cell line was determined using the WST-1 cytotoxicity test. Apoptotic activity was measured by Annexin V test and cell death ELISA kit. The results showed that 25 μM and 50 μM doses of succinic acid for 24 h remarkably reduced the cell viability for CAKI-2 cells (89.77% and 90.77%) and ACHN cells (41.57% and 54.54%). Also, no significant effect was observed on the healthy cell line, as we expected. Additionally, administration of succinic acid at same doses resulted in apoptotic activity for ACHN cells (19.1 and 12.7) and CAKI-2 cells (19.85 and 29.55). ELISA results with same doses of succinic acid treatment increased the apoptotic fragment rates by 4.7 and 2.13-fold in CAKI-2 cells, and 32.92, 12.7-fold in ACHN cells. Succinic acid is a focal point for cancer treatments not only for its apoptotic success on cancer cells but also for its capacity to be metabolically active for humans. Our results suggest that succinic acid could be a potential therapeutic agent for individual cancer treatment approaches together with further molecular research.
Collapse
|
13
|
Khalili-Tanha G, Moghbeli M. Long non-coding RNAs as the critical regulators of doxorubicin resistance in tumor cells. Cell Mol Biol Lett 2021; 26:39. [PMID: 34425750 PMCID: PMC8381522 DOI: 10.1186/s11658-021-00282-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
Resistance against conventional chemotherapeutic agents is one of the main reasons for tumor relapse and poor clinical outcomes in cancer patients. Various mechanisms are associated with drug resistance, including drug efflux, cell cycle, DNA repair and apoptosis. Doxorubicin (DOX) is a widely used first-line anti-cancer drug that functions as a DNA topoisomerase II inhibitor. However, DOX resistance has emerged as a large hurdle in efficient tumor therapy. Furthermore, despite its wide clinical application, DOX is a double-edged sword: it can damage normal tissues and affect the quality of patients’ lives during and after treatment. It is essential to clarify the molecular basis of DOX resistance to support the development of novel therapeutic modalities with fewer and/or lower-impact side effects in cancer patients. Long non-coding RNAs (lncRNAs) have critical roles in the drug resistance of various tumors. In this review, we summarize the state of knowledge on all the lncRNAs associated with DOX resistance. The majority are involved in promoting DOX resistance. This review paves the way to introducing an lncRNA panel marker for the prediction of the DOX response and clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Opoku-Damoah Y, Zhang R, Ta HT, Xu ZP. Vitamin E-facilitated carbon monoxide pro-drug nanomedicine for efficient light-responsive combination cancer therapy. Biomater Sci 2021; 9:6086-6097. [PMID: 34346418 DOI: 10.1039/d1bm00941a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The quest to maximize therapeutic efficiency in cancer treatment requires innovative delivery nanoplatforms capable of employing different modules simultaneously. Combination therapy has proven to be one of the best anticancer strategies so far. Herein, we have developed a lipid-encapsulated nanoplatform that combines chemotherapy with photoresponsive gas therapy for colon cancer treatment. Carbon monoxide releasing molecules (CORMs) and vitamin E analogues (pure/pegylated α-tocopheryl succinate; α-TOS) were co-loaded into the lipid layer with core-shell upconversion nanoparticles (UCNPs), which converted 808 nm light to 360 nm photons to trigger CO release at the tumor site. This folic acid (FA)-targeting nanomedicine (Lipid/UCNP/CORM/α-TOS/FA: LUCTF) possessed a cancer-targeting ability and a light-triggered CO release ability for synergistic apoptosis of HCT116 cells via enhanced ROS generation and mitochondrial membrane breaking. In vivo data have confirmed the significantly enhanced therapeutic efficacy of LUCTF without any significant biosafety issues after intravenous administration. Thus, nanomedicine LUCTF represents a novel way for efficient cancer therapy via combining locally released CO and a compatible chemotherapeutic agent (e.g. α-TOS).
Collapse
Affiliation(s)
- Yaw Opoku-Damoah
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia. and School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia and Queensland Micro and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
15
|
Huang X, Neckenig M, Sun J, Jia D, Dou Y, Ai D, Nan Z, Qu X. Vitamin E succinate exerts anti-tumour effects on human cervical cancer cells via the CD47-SIRPɑ pathway both in vivo and in vitro. J Cancer 2021; 12:3877-3886. [PMID: 34093795 PMCID: PMC8176246 DOI: 10.7150/jca.52315] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 04/22/2021] [Indexed: 12/09/2022] Open
Abstract
Vitamin E succinate (RRR-a-tocopheryl succinate, VES) acts as a potent agent for cancer therapy and has no toxic and side effects on normal tissue cells. However, the mechanism by which VES mediates the effects are not yet fully understood. Here, we hypothesised that VES mediates antitumour activity on human cervical cancer cells via the CD47-SIRPɑ pathway in vivo and in vitro. Results indicated that the human cervical cancer HeLa cells treated with VES were more efficiently engulfed by THP-1-derived macrophages. In response to VES, the protein expression of CD47 on cell membranes and the mRNA level of CD47 in different human cervical cancer cells significantly decreased. And the level of calreticulin (CRT) mRNA in the VES-treated cells increased. By contrast, CRT protein expression was not altered. miRNA-155, miRNA-133 and miRNA-326 were up-regulated in the VES-treated HeLa cells. Knocking down miRNA-155 and miRNA-133 by RNA interference increased CD47 protein expression in the VES-treated cells. In vivo efficacy was determined in BALB/C nude mice with HeLa xenografts. Results showed that VES reduced tumour growth, increased overall survival and inhibited CD47 in the tumour transcriptionally and translationally. Furthermore, inflammatory factors (TNF-α, IL-12, IFN-γ, IL-2 and IL-10) in the spleen were altered because of VES treatment. Our results suggest that VES-induced antitumour activity is coupled to the CD47-SIRPɑ pathway in human cervical HeLa cancer cells.
Collapse
Affiliation(s)
- Xiaoli Huang
- Department of Nutrition, Qilu Hospital of Shandong University, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Markus Neckenig
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Jintang Sun
- Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Di Jia
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, Heilongjiang, China
| | - Yu Dou
- Department of Tissue Engineering and Regeneration, School and Hospital of Stomatology, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Dan Ai
- Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhaodi Nan
- Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
16
|
Vitamin E succinate with multiple functions: A versatile agent in nanomedicine-based cancer therapy and its delivery strategies. Int J Pharm 2021; 600:120457. [PMID: 33676991 DOI: 10.1016/j.ijpharm.2021.120457] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 11/20/2022]
Abstract
Vitamin E succinate (VES), a succinic acid ester of vitamin E, is one of the most effective anticancer compounds of the vitamin E family. VES can inhibit tumor growth by multiple pathways mainly involve tumor proliferation inhibition, apoptosis induction, and metastasis prevention. More importantly, the mitochondrial targeting and damaging property of VES endows it with great potential in exhibiting synergetic effect with conventional chemotherapeutic drugs and overcoming multidrug resistance (MDR). Given the lipophilicity of VES that hinders its bioavailability and therapeutic activity, nanotechnology with multiple advantages has been widely explored to deliver VES and opened up new avenues for its in vivo application. This review aims to introduce the anticancer mechanisms of VES and summarize its delivery strategies using nano-drug delivery systems. Specifically, VES-based combination therapy for synergetic anticancer effect, MDR-reversal, and oral chemotherapy improvement are highlighted. Finally, the challenges and perspectives are discussed.
Collapse
|
17
|
Ma Y, Li D, Xiao Y, Ouyang Z, Shen M, Shi X. LDH-doped electrospun short fibers enable dual drug loading and multistage release for chemotherapy of drug-resistant cancer cells. NEW J CHEM 2021. [DOI: 10.1039/d1nj02159a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
LDH-incorporated PLGA short nanofibers can be loaded with dual drugs for multistage release and chemotherapy of drug-resistant cancer cells.
Collapse
Affiliation(s)
- Yupei Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine
- International Joint Laboratory for Advanced Fiber and Low-dimension Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
| | - Du Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine
- International Joint Laboratory for Advanced Fiber and Low-dimension Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
| | - Yunchao Xiao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine
- International Joint Laboratory for Advanced Fiber and Low-dimension Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
| | - Zhijun Ouyang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine
- International Joint Laboratory for Advanced Fiber and Low-dimension Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine
- International Joint Laboratory for Advanced Fiber and Low-dimension Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine
- International Joint Laboratory for Advanced Fiber and Low-dimension Materials
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
| |
Collapse
|
18
|
Lages EB, Fernandes RS, Silva JDO, de Souza ÂM, Cassali GD, de Barros ALB, Miranda Ferreira LA. Co-delivery of doxorubicin, docosahexaenoic acid, and α-tocopherol succinate by nanostructured lipid carriers has a synergistic effect to enhance antitumor activity and reduce toxicity. Biomed Pharmacother 2020; 132:110876. [PMID: 33113428 DOI: 10.1016/j.biopha.2020.110876] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022] Open
Abstract
Doxorubicin (DOX) is widely used in cancer treatment, however, its use is often limited due to its side effects. To avoid these shortcomings, the encapsulation of DOX into nanocarriers has been suggested. Herein, we proposed a novel nanostructured lipid carrier (NLC) formulation loading DOX, docosahexaenoic acid (DHA), and α-tocopherol succinate (TS) for cancer treatment. DHA is an omega-3 fatty acid and TS is a vitamin E derivative. It has been proposed that these compounds can enhance the antitumor activity of chemotherapeutics. Thus, we hypothesized that the combination of DOX, DHA, and TS in NLC (NLC-DHA-DOX-TS) could increase antitumor efficacy and also reduce toxicity. NLC-DHA-DOX-TS was prepared using emulsification-ultrasound. DOX was incorporated after preparing the NLC, which prevented its degradation during manufacture. High DOX encapsulation efficiency was obtained due to the ion-pairing with TS. This ion-pairing increases lipophilicity of DOX and reduces its crystallinity, contributing to its encapsulation in the lipid matrix. Controlled DOX release from the NLC was observed in vitro, with increased drug release at the acidic environment. In vitro cell studies indicated that DOX, DHA, and TS have synergistic effects against 4T1 tumor cells. The in vivo study showed that NLC-DHA-DOX-TS exhibited the greatest antitumor efficacy by reducing tumor growth in 4T1 tumor-bearing mice. In addition, this formulation reduced mice mortality, prevented lung metastasis, and decreased DOX-induced toxicity to the heart and liver, which was demonstrated by hematologic, biochemical, and histologic analyses. These results indicate that NLC-DHA-DOX-TS may be a promising carrier for breast cancer treatment.
Collapse
Affiliation(s)
- Eduardo Burgarelli Lages
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renata Salgado Fernandes
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana de Oliveira Silva
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ângelo Malachias de Souza
- Department of Physics, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - André Luís Branco de Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lucas Antônio Miranda Ferreira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
19
|
Tiwari S, Sarolia J, Kansara V, Chudasama NA, Prasad K, Ray D, Aswal VK, Bahadur P. Synthesis, Colloidal Characterization and Targetability of Phenylboronic Acid Functionalized α-Tocopheryl Polyethylene Glycol Succinate in Cancer Cells. Polymers (Basel) 2020; 12:polym12102258. [PMID: 33019616 PMCID: PMC7600591 DOI: 10.3390/polym12102258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/27/2020] [Indexed: 12/20/2022] Open
Abstract
This study reports targetable micelles developed after covalent functionalization of α-tocopheryl polyethylene glycol succinate (TPGS) with amino phenylboronic acid (APBA). Nuclear magnetic resonance (NMR) and infrared (IR) spectroscopic results showed successful attachment of APBA to the hydrophilic segment of TPGS. Dynamic light scattering and small-angle neutron scattering studies revealed that the conjugate self-assembled in water to produce spherical core-shell micelles (14–20 nm) which remained stable against temperature (ca. 25–45 °C) and pH changes. The micelles could solubilize a high payload of paclitaxel (PLX) without exhibiting changes in the average size. However, at the saturation solubility, drug molecules migrated from the core to the shell region and engaged with APBA groups via π–π stacking interaction. Confocal microscopy and cell sorting analyses verified the effective translocation ability of TPGS-APBA micelles in sialic acid (SA) expressing MDA-MB-453 cells. At equivalent PLX dose, TPGS-APBA micelles showed about a twofold improvement in apoptotic death among the cells exposed for 2 h. Our findings indicate that the attachment of APBA can be a potential strategy for improving the intra-cellular localization of carriers among cancer cells expressing SA residues.
Collapse
Affiliation(s)
- Sanjay Tiwari
- Maliba Pharmacy College, Gopal-Vidyanagar Campus, Uka Tarsadia University, Surat 394350, India; (S.T.); (J.S.); (V.K.)
| | - Jayant Sarolia
- Maliba Pharmacy College, Gopal-Vidyanagar Campus, Uka Tarsadia University, Surat 394350, India; (S.T.); (J.S.); (V.K.)
| | - Vrushti Kansara
- Maliba Pharmacy College, Gopal-Vidyanagar Campus, Uka Tarsadia University, Surat 394350, India; (S.T.); (J.S.); (V.K.)
| | - Nishith A. Chudasama
- Natural Products & Green Chemistry Division, Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, India; (N.A.C.); (K.P.)
| | - Kamalesh Prasad
- Natural Products & Green Chemistry Division, Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, India; (N.A.C.); (K.P.)
| | - Debes Ray
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India; (D.R.); (V.K.A.)
| | - Vinod K Aswal
- Solid State Physics Division, Bhabha Atomic Research Centre, Mumbai 400085, India; (D.R.); (V.K.A.)
| | - Pratap Bahadur
- Department of Chemistry, Veer Narmad South Gujarat University, Surat 395007, India
- Correspondence:
| |
Collapse
|
20
|
Wan H, Liu X, Chen Y, Tang R, Yi B, Liu D. Silencing of the ARK5 gene reverses the drug resistance of multidrug-resistant SGC7901/DDP gastric cancer cells. PeerJ 2020; 8:e9560. [PMID: 32844054 PMCID: PMC7416719 DOI: 10.7717/peerj.9560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/25/2020] [Indexed: 01/12/2023] Open
Abstract
For several years, the multidrug resistance (MDR) of gastric cancer cells has been a thorny issue worldwide regarding the chemotherapy process and needs to be solved. Here, we report that the ARK5 gene could promote the multidrug resistance of gastric cancer cells in vitro and in vivo. In this study, LV-ARK5-RNAi lentivirus was used to transfect the parental cell line SGC7901 and MDR cell line SGC7901/DDP to construct a stable model of ARK5 interference. Subsequently, the cells were treated with four chemotherapeutic drugs, cisplatin (DDP), adriamycin (ADR), 5-fluorouracil (5-FU) and docetaxel (DR) and were subjected to the CCK8, colony formation, adriamycin accumulation and retention, cell apoptosis and other assays. The study found that, in vitro, the expression of ARK5 in MDR gastric cancer cells was significantly higher than that in parental cells. Additionally, when treated with different chemotherapeutic drugs, compared with parental cells, MDR cells also had a higher cell survival rate, higher colony formation number, higher drug pump rate, and lower cell apoptosis rate. Additionally, in xenograft mouse models, MDR cells with high ARK5 expression showed higher resistance to chemotherapeutic drugs than parental cells. Overall, this study revealed that silencing the ARK5 gene can effectively reverse the drug resistance of MDR gastric cancer cells to chemotherapeutic drugs, providing insights into the mechanism of this process related to its inhibition of the active pump-out ability of MDR cells.
Collapse
Affiliation(s)
- Hongtao Wan
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang, China.,Second Abdominal Surgery Department, Jiangxi Province Tumor Hospital, Nanchang, China
| | - Xiaowei Liu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang, China.,Nanchang Joint Programme, Queen Mary University of London, Nanchang, China
| | - Yanglin Chen
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang, China.,Second Abdominal Surgery Department, Jiangxi Province Tumor Hospital, Nanchang, China
| | - Ren Tang
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang, China.,Second Abdominal Surgery Department, Jiangxi Province Tumor Hospital, Nanchang, China
| | - Bo Yi
- Second Abdominal Surgery Department, Jiangxi Province Tumor Hospital, Nanchang, China
| | - Dan Liu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, Nanchang University School of Pharmaceutical Science, Nanchang, China
| |
Collapse
|
21
|
Neunert G, Tomaszewska-Gras J, Witkowski S, Polewski K. Tocopheryl Succinate-Induced Structural Changes in DPPC Liposomes: DSC and ANS Fluorescence Studies. Molecules 2020; 25:molecules25122780. [PMID: 32560228 PMCID: PMC7356869 DOI: 10.3390/molecules25122780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 01/02/2023] Open
Abstract
Recent studies show that alpha-tocopheryl succinate (TS) exhibits selective toxicity against cancer cells. In this study, we investigated the effect of TS's presence on the physico-chemical and structural properties of DPPC liposomes using fluorescence parameters (intensity, lifetime, and position of emission maximum) of 1-anilino-8-naphtalene sulphonate (ANS), differential scanning calorimetry (DSC) and zeta potential methods. Increasing the TS presence in the DPPC gel phase produced ANS fluorescence enhancement with a hypsochromic shift of the maximum. The zeta potential measurements show an increase in the negative surface charge and confirmed that this process is connected with the hydrophobic properties of dye, which becomes located deeper into the interphase region with a progressing membrane disorder. Temperature dependence studies showed that an increase in temperature increases the ANS fluorescence and shifts the ANS maximum emission from 464 to 475 nm indicating a shift from hydrophobic to a more aqueous environment. In the liquid crystalline phase, the quenching of ANS fluorescence occurs due to the increased accessibility of water to the ANS located in the glycerol region. The DSC results revealed that increasing the presence of TS led to the formation of multicomponent DSC traces, indicating the formation of intermediate structures during melting. The present results confirmed that TS embedded into the DPPC membrane led to its disruption due to destabilisation of its structure, which confirmed the measured biophysical parameters of the membrane.
Collapse
Affiliation(s)
- Grażyna Neunert
- Department of Physics and Biophysics, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, Wojska Polskiego 38/42, 60-637 Poznań, Poland;
| | - Jolanta Tomaszewska-Gras
- Department of Food Safety and Quality Management, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, Wojska Polskiego 31/33, 60-624 Poznań, Poland;
| | - Stanislaw Witkowski
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1K, 15-245 Bialystok, Poland;
| | - Krzysztof Polewski
- Department of Physics and Biophysics, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, Wojska Polskiego 38/42, 60-637 Poznań, Poland;
- Correspondence:
| |
Collapse
|
22
|
MSC-induced lncRNA HCP5 drove fatty acid oxidation through miR-3619-5p/AMPK/PGC1α/CEBPB axis to promote stemness and chemo-resistance of gastric cancer. Cell Death Dis 2020; 11:233. [PMID: 32300102 PMCID: PMC7162922 DOI: 10.1038/s41419-020-2426-z] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022]
Abstract
Chemotherapy is the first-tier treatment regime for gastric cancer (GC) patients at advance stages. Mesenchymal stem cell (MSC) cam affect drug-resistance of GC cells in tumor microenvironment, but the detailed mechanism remains poorly understood. Present study aimed to investigate the regulation of MSC-induced long non-coding RNA (lncRNA) in GC. Dysregulated lncRNAs in GC were analyzed based on GEO data. Stemness and drug-resistance of GC cells were detected by sphere formation, colony formation, CCK-8, and flow cytometry analyses. MicroRNA (miRNA)-related pathways were analyzed by online KEGG analysis tool DAVID6.8. Molecular interactions were determined by luciferase reporter assay, pulldown, RNA immunoprecipitation (RIP), chromatin immunoprecipitation (ChIP), and co-immunoprecipitation (CoIP). Results revealed that MSC co-culture improved stemness and drug-resistance of GC cells. LncRNA histocompatibility leukocyte antigen complex P5 (HCP5) was induced in GC cells by MSC co-culture, contributing to stemness and drug-resistance. Mechanistically, HCP5 sequestered miR-3619-5p and upregulated PPARG coactivator 1 alpha (PPARGC1A), increasing transcription complex Peroxisome proliferator activated receptor (PPAR) coactivator-1α (PGC1α)/CEBPB and transcriptionally inducing carnitine palmitoyltransferase 1 (CPT1), which prompted the fatty acid oxidation (FAO) in GC cells. In conclusion, MSC-induced lncRNA HCP5 drove FAO through miR-3619-5p/AMPK/PGC1α/CEBPB axis to promote stemness and chemo-resistance of GC, indicating that targeting HCP5 was a novel approach to enhancing the efficacy of chemotherapy in GC.
Collapse
|
23
|
Hama S, Okamura Y, Kamei K, Nagao S, Hayashi M, Shizuka M, Fukuzawa K, Kogure K. α-Tocopheryl succinate stabilizes the structure of tumor vessels by inhibiting angiopoietin-2 expression. Biochem Biophys Res Commun 2020; 521:947-951. [DOI: 10.1016/j.bbrc.2019.11.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/02/2019] [Indexed: 12/13/2022]
|
24
|
Boratto FA, Franco MS, Barros ALB, Cassali GD, Malachias A, Ferreira LAM, Leite EA. Alpha-tocopheryl succinate improves encapsulation, pH-sensitivity, antitumor activity and reduces toxicity of doxorubicin-loaded liposomes. Eur J Pharm Sci 2019; 144:105205. [PMID: 31874285 DOI: 10.1016/j.ejps.2019.105205] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/27/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
Doxorubicin (DOX) plays an important role in cancer treatment; however, high cardiotoxicity and low penetration in solid tumors are the main limitations of its use. Liposomal formulations have been developed to attenuate the DOX toxicity, but the technological enhancement of the liposomal formulation as well as the addition of another agent with antitumor properties, like alpha-tocopheryl succinate (TS), a semi-synthetic analog of vitamin E, could certainly bring benefits. Thus, in this study, it was proposed the development of liposomes composed of DOX and TS (pHSL-TS-DOX). A new DOX encapsulation method, without using the classic ammonium sulfate gradient with high encapsulation percentage was developed. Analysis of Small Angle X-ray Scattering (SAXS) and release study proved the pH-sensitivity of the developed formulation. It was observed stabilization of tumor growth using pHSL-TS-DOX when compared to free DOX. The toxicity tests showed the safety of this formulation since it allowed body weight initial recovery after the treatment and harmless to heart and liver, main target organs of DOX toxicity. The developed formulation also avoided the occurrence of myelosuppression, a typical adverse effect of DOX. Therefore, pHSL-TS-DOX is a promising alternative for the treatment of breast cancer since it has adequate antitumor activity and a safe toxicity profile.
Collapse
Affiliation(s)
- F A Boratto
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - M S Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - A L B Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - G D Cassali
- Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - A Malachias
- Department of Physics, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - L A M Ferreira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - E A Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| |
Collapse
|
25
|
Wei J, Long Y, Guo R, Liu X, Tang X, Rao J, Yin S, Zhang Z, Li M, He Q. Multifunctional polymeric micelle-based chemo-immunotherapy with immune checkpoint blockade for efficient treatment of orthotopic and metastatic breast cancer. Acta Pharm Sin B 2019; 9:819-831. [PMID: 31384541 PMCID: PMC6664045 DOI: 10.1016/j.apsb.2019.01.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/11/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
Immunotherapy has become a highly promising paradigm for cancer treatment. Herein, a chemo-immunotherapy was developed by encapsulating chemotherapeutic drug doxorubicin (DOX) and Toll-like receptor 7 agonist imiquimod (IMQ) in low molecular weight heparin (LMWH)-d-α-tocopheryl succinate (TOS) micelles (LT). In this process, LMWH and TOS were conjugated by ester bond and they were not only served as the hydrophilic and hydrophobic segments of the carrier, but also exhibited strong anti-metastasis effect. The direct killing of tumor cells mediated by DOX-loaded micelles (LT-DOX) generated tumor-associated antigens, initiating tumor-specific immune responses in combination with IMQ-loaded micelles (LT-IMQ). Furthermore, the blockade of immune checkpoint with programmed cell death ligand 1 (PD-L1) antibody further elevated the immune responses by up-regulating the maturation of DCs as well as the ratios of CD8+ CTLs/Treg and CD4+ Teff/Treg. Therefore, such a multifunctional strategy exhibited great potential for inhibiting the growth of orthotopic and metastatic breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Man Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | | |
Collapse
|
26
|
Lv X, Zhu Y, Ghandehari H, Yu A, Wang Y. An ROS-responsive and self-accelerating drug release nanoplatform for overcoming multidrug resistance. Chem Commun (Camb) 2019; 55:3383-3386. [PMID: 30821310 DOI: 10.1039/c9cc00358d] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
An 'on-demand' drug release and ROS-responsive nanoparticle was prepared by chemically conjugating hydrophobic α-tocopheryl succinate to hydrophilic poly(ethylene glycol) via a thioketal linker. This nanoparticle encapsulated with doxorubicin and α-tocopheryl succinate exhibited remarkable efficiency in reversing multidrug resistance both in vitro and in vivo.
Collapse
Affiliation(s)
- Xueming Lv
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China.
| | | | | | | | | |
Collapse
|
27
|
de Melo-Diogo D, Costa EC, Alves CG, Lima-Sousa R, Ferreira P, Louro RO, Correia IJ. POxylated graphene oxide nanomaterials for combination chemo-phototherapy of breast cancer cells. Eur J Pharm Biopharm 2018; 131:162-169. [DOI: 10.1016/j.ejpb.2018.08.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/03/2018] [Accepted: 08/18/2018] [Indexed: 01/08/2023]
|
28
|
Zhou Z, Lin Z, He Y, Pang X, Wang Y, Ponnusamy M, Ao X, Shan P, Tariq MA, Li P, Wang J. The Long Noncoding RNA D63785 Regulates Chemotherapy Sensitivity in Human Gastric Cancer by Targeting miR-422a. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:405-419. [PMID: 30195778 PMCID: PMC6036868 DOI: 10.1016/j.omtn.2018.05.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/24/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023]
Abstract
Gastric cancer is one of the most prevalent tumor types in the world. Chemotherapy is the most common choice for cancer treatment. However, chemotherapy resistance and adverse side effects limit its clinical applications. Aberrant expression of long noncoding RNAs (lncRNAs) has been found in various stages of gastric cancer development and progression. In this study, we identified that an oncogenic lncRNA, long intergenic non-protein-coding RNA D63785 (lncR-D63785), is highly expressed in gastric cancer tissues and cells. Silencing of lncR-D63785 inhibited cell proliferation, cell migration and invasion in gastric cancer cell lines and reduced tumor volume and size in mice. We found that the expression of lncR-D63785 was inversely correlated with microRNA 422a (miR-422a) expression, which was involved in the downregulation of expression of myocyte enhancer factor-2D (MEF2D) and drug sensitivity. Knockdown of lncR-D63785 increased the expression of miR-422a and the sensitivity of gastric cancer cells to apoptosis induced by the anticancer drug doxorubicin (DOX). This indicates that lncR-D63785 acts as a competitive endogenous RNA (ceRNA) of miR-422a and promotes chemoresistance by blocking miR-422-dependent suppression of MEF2D. Together, our results suggest that the therapeutic suppression of lncR-D63785 alone or in combination with chemotherapeutic agents may be a promising strategy for treating gastric cancer.
Collapse
Affiliation(s)
- Zhixia Zhou
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Zhijuan Lin
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, Weifang 261053, China
| | - Yuqi He
- Department of Gastroenterology, Beijing Military General Hospital, Beijing 100700, China
| | - Xin Pang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Yin Wang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Murugavel Ponnusamy
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Xiang Ao
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Peipei Shan
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Muhammad Akram Tariq
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China.
| | - Jianxun Wang
- Center for Tumor Molecular Biology, Institute for Translational Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
29
|
α- Tocopherol succinate loaded nano-structed lipid carriers improves antitumor activity of doxorubicin in breast cancer models in vivo. Biomed Pharmacother 2018; 103:1348-1354. [PMID: 29864917 DOI: 10.1016/j.biopha.2018.04.139] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 12/15/2022] Open
Abstract
Combination-based chemotherapies have been the standard treatment for multiple solid tumors since the 1960s. Combined therapies where both agents have toxicity results in dose-limiting effects. α- tocopherol succinate (TS) is an analogue of vitamin E that exhibits antitumor properties in the absence of toxicity. Hence, its combination with a frontline chemotherapy, doxorubicin (DOX) is an alternative to increase antitumor efficacy. Therefore, the aim of this work was to evaluate the antitumor activity of nanostructed lipid carriers (NLC) loaded with TS and DOX. The NLC-TS-DOX were prepared, characterized and radiolabeled with technetium-99m. Cytotoxicity studies were performed in vitro, using two breast cancer cell lines, MDA-MB-231 and 4T1. Biodistribution and antitumor activity were evaluated in 4T1 tumor-bearing mice. The results showed that NLC-TS-DOX had a small diameter (85 nm) and a long blood clearance (T1/2β = 1107.71 min) that consequently resulted in a higher tumor uptake compared to contralateral muscle for up to 48 h. Drug combination studies in MDA-MB-231 and 4T1 cells showed a combination index below 0.8 at ED50-90 for both cell lines. Interestingly, a high synergism was found at ED90. Antitumor activity showed a better control of tumor growth for animals treated with NLC-ST-DOX. The small particle size, along with the EPR effect and the controlled release of DOX from the particle, associated with the synergic combination between TS and DOX led to an increase of the antitumor efficacy. Therefore, NLC-TS-DOX can be considered a plausible alternative to improve antitumor efficacy in DOX therapeutic regimens.
Collapse
|
30
|
Liang C, Wang H, Zhang M, Cheng W, Li Z, Nie J, Liu G, Lian D, Xie Z, Huang L, Zeng X. Self-controlled release of Oxaliplatin prodrug from d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) functionalized mesoporous silica nanoparticles for cancer therapy. J Colloid Interface Sci 2018; 525:1-10. [PMID: 29679795 DOI: 10.1016/j.jcis.2018.04.058] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 04/12/2018] [Accepted: 04/15/2018] [Indexed: 12/11/2022]
Abstract
Oxaliplatin is a promising antitumor drug, but its effectiveness is limited by its side effects in vivo. In this study, we introduced an Oxaliplatin prodrug (Oxa(IV)) self-controlled release strategy, in which Oxa(IV) is encapsulated by TPGS functionalized mesoporous silica nanoparticles (MSNs), and its release is controlled by biological stimuli, such as acidic environments in tumor tissue and high concentrations of reductants in cancer cells. Despite the lack of auxiliary "gatekeepers" to MSNs, this simplified model of Oxa(IV)-MSNs-TPGS could fine-tune the movements of the drug release. Furthermore, we utilized a prodrug approach to avoid the side effects of Oxaliplatin, and we used TPGS groups to reduce multidrug resistance (MDR). Finally, the toxicity of Oxa(IV)-MSNs-TPGS to a human lung adenocarcinoma cell line (A549) in vitro was significantly lower than that of Oxaliplatin. This model demonstrates the considerable potential of a simple self-controlled release system with multiple functions.
Collapse
Affiliation(s)
- Chaoyu Liang
- Department of Chemistry, Tsinghua University, Beijing 100084, China; Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), The Shenzhen Key Lab of Gene and Antibody Therapy, and Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Heping Wang
- Department of Respiratory Diseases, Shenzhen Children's Hospital, 7019 Yitian Road, Futian District, Shenzhen 518026, China
| | - Min Zhang
- Shenzhen Xili People's Hospital, Shenzhen 518055, China
| | - Wei Cheng
- Department of Chemistry, Tsinghua University, Beijing 100084, China; Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), The Shenzhen Key Lab of Gene and Antibody Therapy, and Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Zihuang Li
- Department of Radiation Oncology, Second Clinical Medicine College of Jinan University, Shenzhen Municipal People's Hospital, Shenzhen 518020, China.
| | - Junpeng Nie
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), The Shenzhen Key Lab of Gene and Antibody Therapy, and Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Gan Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Daizheng Lian
- Department of Radiation Oncology, Second Clinical Medicine College of Jinan University, Shenzhen Municipal People's Hospital, Shenzhen 518020, China
| | - Zhenhua Xie
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), The Shenzhen Key Lab of Gene and Antibody Therapy, and Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Laiqiang Huang
- Department of Chemistry, Tsinghua University, Beijing 100084, China; Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), The Shenzhen Key Lab of Gene and Antibody Therapy, and Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China.
| | - Xiaowei Zeng
- Department of Chemistry, Tsinghua University, Beijing 100084, China; Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), The Shenzhen Key Lab of Gene and Antibody Therapy, and Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
31
|
Fu ZY. Role of ATP-binding cassette transporters, apoptosis, and long non-coding RNAs in gastric cancer multidrug resistance. Shijie Huaren Xiaohua Zazhi 2017; 25:2838-2850. [DOI: 10.11569/wcjd.v25.i32.2838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer multidrug resistance refers to the cross resistance of cancer cells to a variety of anticancer drugs, which can be primary or secondary. Several mechanisms attribute to cancer multidrug resistance. In this paper, the recent progress in the understanding of the mechanisms of multi-drug resistance of gastric cancer cells with regard to the role of adenosine triphosphate binding cassette transporters, apoptosis, and long non-coding RNAs is reviewed.
Collapse
Affiliation(s)
- Zhao-Ying Fu
- Institute of Molecular Biology and Immunology, Medical School of Yan'an University, Yan'an 716000, Shaanxi Province, China
| |
Collapse
|
32
|
Yang JZ, Huang LH, Chen R, Meng LJ, Gao YY, Ji QY, Wang Y. LIM kinase 1 serves an important role in the multidrug resistance of osteosarcoma cells. Oncol Lett 2017; 15:250-256. [PMID: 29387219 PMCID: PMC5768095 DOI: 10.3892/ol.2017.7317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/21/2017] [Indexed: 01/07/2023] Open
Abstract
Multidrug resistance (MDR) is a major challenge for the management of the majority of cancers. The precise molecular mechanisms of MDR remain elusive. In a previous study, a multidrug resistant osteosarcoma model [MG63/vincristine (VCR)] was established by intermittent exposure of MG63 cells to gradually increasing concentrations of VCR. These cells exhibited cross-resistance to multiple structurally and mechanistically unrelated chemotherapeutic agents. The development of MDR was associated with increased expression of LIM kinase 1 (LIMK1). Compared with that in normal human fetal osteoblasts (hFOB) 1.19, the messenger RNA and protein expression of LIMK1 was significantly elevated both in MG63 and U2OS osteosarcoma cells. To observe the expression pattern of LIMK1 in osteosarcoma, immunohistochemical analyses were performed on specimens derived from 6 patients. The results indicated that LIMK1 was expressed to a greater extent in the tumor parenchyma than in the mesenchyme. The role of LIMK1 in MDR was confirmed by transfecting plasmids coding LIMK1-small interfering RNA (siRNA), wild-type-LIMK1 or empty vector into MG63/VCR cells, and measuring the expression of LIMK1 and multidrug resistance protein 1 (MDR1), also known as P-gycoprotein (P-gp). The results demonstrated that the level of MDR1/P-gp was positively correlated with the level of LIMK1. This correlation was also shown with the doxorubicin efflux assay and by measuring apoptosis. Specifically, after 6 h of incubation with VCR, 25.6% of the cells transfected with the LIMK1-siRNA plasmid were apoptotic compared with 6.2% in the empty vector group and 1.3% in the group of cells transfected with the wild-type-LIMK1 plasmid. Thus, it was concluded that LIMK1 serves a key role in the MDR of osteosarcoma and functions through MDR1.
Collapse
Affiliation(s)
- Jian-Zeng Yang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China.,Department of Nuclear Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Li-Hong Huang
- Department of Physiology and Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Rui Chen
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ling-Jie Meng
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yi-Yao Gao
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Qiu-Ye Ji
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yan Wang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
33
|
Song Y, Tang C, Yin C. Enhanced antitumor efficacy of arginine modified amphiphilic nanoparticles co-delivering doxorubicin and iSur-pDNA via the multiple synergistic effect. Biomaterials 2017; 150:1-13. [PMID: 29028548 DOI: 10.1016/j.biomaterials.2017.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/01/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Arginine and α-tocopherol succinate (α-TOS) double grafted N-trimethyl chitosan chloride (TMC) nanoparticles (TAS NPs) were designed and developed for effective co-delivery of doxorubicin (DOX) and Survivin shRNA-expressing pDNA (iSur-pDNA). With DOX loading into the hydrophobic core and iSur-pDNA combining to the hydrophilic shell, TAS/DOX/pDNA NPs demonstrated favorable structural stability and sustained release properties in vitro. With the special non-clathrin-dependent endocytosis, TAS/DOX/pDNA NPs presented higher cellular uptake and mainly distributed in ER and Golgi rather than lysosomes following internalization. The in vitro nuclear localization, gene silencing efficiency, cell apoptosis, and growth inhibition of tumor cells were significantly promoted by arginine modification. In the tumor-bearing mice model, TAS/DOX/pDNA NPs possessed the maximum antitumor efficiency as compared with single delivery of DOX or iSur-pDNA. Particularly, blank TAS NPs were selectively be toxic to tumor cells as evidenced by their capabilities to inhibit proliferation and induce apoptosis of tumor cells. The promising tumor treatment of TAS/DOX/pDNA NPs via a multiple synergistic manner arising from DOX and pDNA as well as the vectors would provide a potential strategy for a dual-delivery system to improve their therapeutic efficacies.
Collapse
Affiliation(s)
- Yudong Song
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
34
|
Luo S, Wang S, Luo N, Chen F, Hu C, Zhang K. The application of aptamer 5TR1 in triple negative breast cancer target therapy. J Cell Biochem 2017; 119:896-908. [PMID: 28671278 DOI: 10.1002/jcb.26254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 06/30/2017] [Indexed: 12/11/2022]
Abstract
Chemotherapy is one of the standard strategies for treatment of breast cancer. Adriamycin (Dox) is a first-line chemotherapy agent for breast cancer. However, the gastrointestinal reactions, myocardial toxicity and other side effects caused by Dox due to its un-specific cytotoxicity limit the clinical treatment effect. To address this need, aptamer has been regarded as an ideal target molecular carrier. In the present study, we selected an aptamer 5TR1 that can specifically bind to the MUC1 protein which has been regarded as an important tumor biomarker, as well as a potential target in anticancer therapies. Dox was loaded on the modified 5TR1-GC, which specifically targets breast cancer cell MDA-MB-231. Cell viability and apoptosis assays demonstrated that the 5TR1-GC-Dox exhibited target specificity of cytotoxicity in MDA-MB-231. Moreover, in vivo xenograft study also confirmed that 5TR1-GC-Dox had a more effective effect on tumor growth inhibition and induced the apoptosis of malignant tumor cells compared to Dox. We provided a novel experimental and theoretical basis for developing an aptamer targeted drug system, thus to promote the killing effect of drugs on breast cells and to reduce the damage to normal cells and tissues for breast cancer.
Collapse
Affiliation(s)
- Shayang Luo
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Shouman Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Na Luo
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Feiyu Chen
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chun Hu
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Kejing Zhang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
35
|
Lei X, Li K, Liu Y, Wang ZY, Ruan BJ, Wang L, Xiang A, Wu D, Lu Z. Co-delivery nanocarriers targeting folate receptor and encapsulating 2-deoxyglucose and α-tocopheryl succinate enhance anti-tumor effect in vivo. Int J Nanomedicine 2017; 12:5701-5715. [PMID: 28848348 PMCID: PMC5557622 DOI: 10.2147/ijn.s135849] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A combination administration of chemical agents was highlighted to treat tumors. Recently, tumor cell has been found to be different from normal cell in metabolic manner. Most of cancer cells prefer aerobic glycolysis to mitochondrial oxidative phosphorylation (OXPHOS) to satisfy energy and biomass synthesis requirement to survive, grow and proliferate, which provides novel and potential therapeutic targets for chemotherapy. Here, 2-deoxy-d-glucose (2-DG), a potent inhibitor of glucose metabolism, was used to inhibit glycolysis of tumor cells; α-tocopheryl succinate (α-TOS), a water-insoluble vitamin E derivative, was chosen to suppress OXPHOS. Our data demonstrated that the combination treatment of 2-DG and α-TOS could significantly promote the anti-tumor efficiency in vitro compared with administration of the single drug. In order to maximize therapeutic activity and minimize negative side effects, a co-delivery nanocarrier targeting folate receptor (FR) was developed to encapsulate 2-DG and α-TOS simultaneously based on our previous work. Transmission electron microscope, dynamic light scattering method and UV-visible spectrophotometers were used to investigate morphology, size distribution and loading efficiency of the α-TOS-2-DG-loaded and FR-targeted nanoparticles (TDF NPs). The TDF NPs were found to possess a layer-by-layer shape, and the dynamic size was <100 nm. The final encapsulation efficiencies of α-TOS and 2-DG in TDF NPs were 94.3%±1.3% and 61.7%±7.7% with respect to drug-loading capacities of 8.9%±0.8% and 13.2%±2.6%, respectively. Almost no α-TOS release was found within 80 h, and release of 2-DG was sustained and slow within 72 h. The results of FR binding assay and fluorescence biodistribution revealed that TDF NPs could target FR highly expressed on tumor cell in vitro and in vivo. Further, in vivo anti-tumor experiments showed that TDF NPs had an improved biological function with less toxicity. Thus, our work indicates that the co-delivery TDF NPs have a great potential in tumor therapy.
Collapse
Affiliation(s)
- Xiaoying Lei
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Ke Li
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi.,Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an
| | - Yan Liu
- Genetic Engineering Laboratory of PLA, The Eleventh Institute of Academy of Military Medical Sciences of PLA, Changchun, Jilin, People's Republic of China
| | - Zhen Yu Wang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Ban Jun Ruan
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Li Wang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - An Xiang
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an
| | - Zifan Lu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, The Fourth Military Medical University
| |
Collapse
|
36
|
Debele TA, Lee KY, Hsu NY, Chiang YT, Yu LY, Shen YA, Lo CL. A pH sensitive polymeric micelle for co-delivery of doxorubicin and α-TOS for colon cancer therapy. J Mater Chem B 2017; 5:5870-5880. [PMID: 32264220 DOI: 10.1039/c7tb01031a] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Combination therapy through simultaneous delivery of two or more therapeutic agents using nanocarriers has emerged as an advanced tactic for cancer treatment. To ensure that two therapeutic agents can be co-delivered and rapidly release their cargo in tumor cells, a biocompatible pH-sensitive copolymer, methoxy poly(ethylene glycol)-b-poly(hydroxypropyl methacrylamide-g-α-tocopheryl succinate-g-histidine) (abbreviated as PTH), was designed and synthesized. The PTH copolymers spontaneously self-assembled into micellar-type nanoparticles in aqueous solutions and are used for co-delivery of therapeutic agents, doxorubicin (Dox) and α-TOS. During micellization, π-π stacking occurred between Dox/α-TOS and imidazole rings of PTH copolymers inducing a regular and tight arrangement of copolymers and drugs to form rod-like micelles, thus efficiently increasing the drug loading and encapsulation efficiency. The micelles enabled the rapid release of both Dox and α-TOS when the pH decreased from 7.4 to 4.5. The protein adsorption assay revealed that low amounts of IgG and BSA were adsorbed on the micelles. In vivo biodistribution demonstrated that the micelles could largely accumulate in the tumor tissues. Furthermore, drug-loaded micelles treated with HCT116 cancer cells exhibited higher cytotoxicity than normal cells, which confirmed that α-TOS exhibited a synergy effect with Dox towards cancer cells, while no recognizable side effects were observed during the treatment from organ function tests.
Collapse
Affiliation(s)
- Tilahun Ayane Debele
- Department of Biomedical Engineering, National Yang Ming University, Taipei 112, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
37
|
Karahaliloğlu Z, Kilicay E, Alpaslan P, Hazer B, Baki Denkbas E. Enhanced antitumor activity of epigallocatechin gallate–conjugated dual-drug-loaded polystyrene–polysoyaoil–diethanol amine nanoparticles for breast cancer therapy. J BIOACT COMPAT POL 2017. [DOI: 10.1177/0883911517710811] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The development of novel combination anticancer drug delivery systems is an important step to improve the effectiveness of anticancer treatment in metastatic breast cancer and to overcome increased toxicity of the currently used combination treatments. The aim of this study was to assess efficient targeting, therapeutic efficacy, and bioavailability of a combination of drugs (curcumin and α-tocopheryl succinate) loaded polystyrene–polysoyaoil–diethanol amine nanoparticles. Polystyrene–polysoyaoil–diethanol amine nanoparticles encapsulating two drugs, individually or in combination, were prepared by double-emulsion solvent evaporation method, resulting in particle size smaller than 250 nm with a surface negative charge between −30 and −40 mV. Entrapment efficiency of curcumin and α-tocopheryl succinate in the epigallocatechin gallate–conjugated dual-drug-loaded nanoparticles was found to be 68% and 80%, respectively. The release kinetics of curcumin and α-tocopheryl succinate from the nanoparticles exhibited a gradual and continuous profile followed by an initial burst behavior with a release over 20 days in vitro. Next, we have investigated the anticancer activity of nanoparticles encapsulating both the drugs and individually drug in human breast cancer cells (MDA-MB-231) using double-staining-based cell death analysis, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assessment of cytotoxicity and flow cytometer. In vitro cytotoxicity studies revealed that epigallocatechin gallate–α-tocopheryl succinate/curcumin–polystyrene–polysoyaoil–diethanol amine nanoparticles are more potent than the corresponding α-tocopheryl succinate/curcumin–polystyrene–polysoyaoil–diethanol amine nanoparticles and their single-drug-loaded forms and show a synergistic and breast tumor targeting function. Thus, here, we propose epigallocatechin gallate–conjugated curcumin and α-tocopheryl succinate–loaded polystyrene–polysoyaoil–diethanol amine nanoparticles which effectively inhibit tumor growth and reduce toxicity compared to single-drug chemotherapy.
Collapse
Affiliation(s)
| | - Ebru Kilicay
- Electronic and Automation Division, Bülent Ecevit University, Zonguldak, Turkey
| | - Pınar Alpaslan
- Department of Biomedical Engineering, TOBB University of Economics and Technology, Ankara, Turkey
| | - Baki Hazer
- Physical Chemistry Division, Bülent Ecevit University, Zonguldak, Turkey
| | | |
Collapse
|
38
|
Wu H, Liu S, Gong J, Liu J, Zhang Q, Leng X, Zhang N, Li Y. VCPA, a novel synthetic derivative of α-tocopheryl succinate, sensitizes human gastric cancer to doxorubicin-induced apoptosis via ROS-dependent mitochondrial dysfunction. Cancer Lett 2017; 393:22-32. [PMID: 28216375 DOI: 10.1016/j.canlet.2017.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 11/29/2022]
Abstract
Gastric carcinoma is a common malignant disease worldwide and has a dismal prognosis. Doxorubicin (DOX), one of the most widely used chemotherapeutic agents, has limited use because of its side effects and the development of tumor-cell resistance. Combinations of doxorubicin and non-cross-resistant agents have been required for adjuvant chemotherapy of gastric cancer. Here, we report that VCPA, a novel synthetic derivative of α-Tocopheryl Succinate, induced apoptosis via production of reactive oxygen species (ROS). When used in combination with doxorubicin, lower doses of VCPA sensitized human gastric cancer cells to DOX-induced apoptosis. The DOX/VCPA combination treatment caused an imbalance in the ratio of Bcl-2 to Bax and induced a lethal mitochondrial dysfunction. MAPKs were also activated in response to the DOX/VCPA treatment but played a protective role in DOX-induced cell death. In vivo studies further confirmed the sensitizing effect of VCPA. Combining DOX with VCPA markedly inhibited tumor growth in a tumor xenograft model of human gastric cancer. Taken together, our study revealed that VCPA, through increased ROS production, could synergize with DOX and circumvent DOX resistance in human gastric cancer cells.
Collapse
Affiliation(s)
- Han Wu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, China
| | - Shaoping Liu
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Gong
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, China
| | - Jiuyang Liu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, China
| | - Qian Zhang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, China
| | - Xiaohua Leng
- Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, China
| | - Nian Zhang
- Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yan Li
- Department of Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuhan, China; Department of Peritoneal Cancer Surgery, Cancer Center of Beijing Shijitan Hospital, The Capital Medical University, Beijing, China.
| |
Collapse
|
39
|
Liu B, Han L, Liu J, Han S, Chen Z, Jiang L. Co-delivery of paclitaxel and TOS-cisplatin via TAT-targeted solid lipid nanoparticles with synergistic antitumor activity against cervical cancer. Int J Nanomedicine 2017; 12:955-968. [PMID: 28203075 PMCID: PMC5293363 DOI: 10.2147/ijn.s115136] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cervical cancer is a major world health problem for women. Currently, cancer research focuses on improving therapy for cervical cancer using various treatment options such as co-delivery of chemotherapeutic agents by nanocarriers. PURPOSE The aim of this study was to develop trans-activating transcriptional activator (TAT)-modified solid lipid nanoparticles (SLNs) for co-delivery of paclitaxel (PTX) and α-tocopherol succinate-cisplatin prodrug (TOS-CDDP) (TAT PTX/TOS-CDDP SLNs) in order to achieve synergistic antitumor activity against cervical cancer. METHODS Lipid prodrug of CDDP (TOS-CDDP) and TAT-containing polyethylene glycol-distearoyl-phosphatidylethanolamine (TAT-PEG-DSPE) were synthesized. TAT PTX/TOS-CDDP SLNs were prepared by emulsification and solvent evaporation method. Physicochemical characteristics of SLNs such as size, morphology, and release profiles were explored. In vitro and in vivo studies were carried out to assess the efficacy of their antitumor activity in target cells. RESULTS TAT PTX/TOS-CDDP SLNs could be successfully internalized by HeLa cells and showed a synergistic effect in the suppression of cervical tumor cell growth. They exhibited high tumor tissue accumulation, superior antitumor efficiency, and much lower toxicity in vivo. CONCLUSION The present study indicates that the co-delivery system provides a promising platform as a combination therapy for the treatment of cervical cancer, and possibly other types of cancer as well.
Collapse
Affiliation(s)
- Bo Liu
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji’nan, People’s Republic of China
| | - Li Han
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji’nan, People’s Republic of China
| | - Junyan Liu
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji’nan, People’s Republic of China
| | - Shumei Han
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji’nan, People’s Republic of China
| | - Zhen Chen
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji’nan, People’s Republic of China
| | - Lixi Jiang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji’nan, People’s Republic of China
| |
Collapse
|
40
|
Yu Y, Hou L, Song H, Xu P, Sun Y, Wu K. Akt/AMPK/mTOR pathway was involved in the autophagy induced by vitamin E succinate in human gastric cancer SGC-7901 cells. Mol Cell Biochem 2016; 424:173-183. [PMID: 27796683 DOI: 10.1007/s11010-016-2853-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 10/22/2016] [Indexed: 12/18/2022]
Abstract
Vitamin E succinate (VES), a derivative of vitamin E, is a promising cancer chemopreventive agent that inhibits tumor promotion by inducing apoptotic cell death. The effects of VES on autophagy, an intricate programmed process which helps cells survive in some stressed situations by degrading some cytoplasmic material, are unclear. When human gastric cancer cells SCG-7901 were exposed to VES, both the level of microtubule-associated protein 1 light chain 3 and the yeast ATG6 homolog Beclin-1 increased, and related autophagy genes were activated, thereby suggesting that autophagy was induced by VES. We also observed that VES-induced autophagy was accompanied by the activation of AMP-activated protein kinases (AMPK). VES-induced autophagy decreased when AMPK was inhibited by using small interfering RNA (siRNA), thereby suggesting that VES-induced autophagy is mediated by AMPK. Moreover, further studies revealed that the decreased activity of mammalian target of rapamycin (mTOR) and its downstream targets P70S6K and 4EBP-1 were involved in VES-activated autophagy associated with AMPK activation. The experiments also showed that the activity of protein kinases B (Akt)-mTOR axis was inhibited by VES. VES-induced AMPK activation could be attenuated by Akt activation. Overall, our studies demonstrated that AMPK was involved in the VES-induced autophagy. Crosstalk exists between AMPK and the Akt/mTOR axis. The results elucidated the mechanism of VES-induced autophagy in human gastric cancer cells.
Collapse
Affiliation(s)
- Yang Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
| | - Liying Hou
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Huacui Song
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
| | - Peixiang Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
| | - Yue Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
| | - Kun Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150086, China.
| |
Collapse
|
41
|
Antioxidant and antitumor activity of trolox, trolox succinate, and α-tocopheryl succinate conjugates with nitroxides. Eur J Med Chem 2016; 122:127-137. [DOI: 10.1016/j.ejmech.2016.05.051] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/19/2016] [Accepted: 05/22/2016] [Indexed: 01/11/2023]
|
42
|
Yang JZ, Ma SR, Rong XL, Zhu MJ, Ji QY, Meng LJ, Gao YY, Yang YD, Wang Y. Characterization of multidrug-resistant osteosarcoma sublines and the molecular mechanisms of resistance. Mol Med Rep 2016; 14:3269-76. [DOI: 10.3892/mmr.2016.5590] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 07/08/2016] [Indexed: 11/06/2022] Open
|
43
|
Mallick A, More P, Syed MMK, Basu S. Nanoparticle-Mediated Mitochondrial Damage Induces Apoptosis in Cancer. ACS APPLIED MATERIALS & INTERFACES 2016; 8:13218-13231. [PMID: 27160664 DOI: 10.1021/acsami.6b00263] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Detouring of conventional DNA damaging anticancer drugs into mitochondria to damage mitochondrial DNA is evolving as a promising strategy in chemotherapy. Inhibiting single target in mitochondria would eventually lead to the emergence of drug resistance. Moreover, targeting mitochondria selectively in cancer cells, keeping them intact in healthy cells, remains a major challenge. Herein, triphenylphosphine (TPP)-coated positively charged 131.6 nm spherical nanoparticles (NPs) comprised of α-tocopheryl succinate (TOS, inhibitor of complex II in electron transport chain) and obatoclax (Obt, inhibitor of Bcl-2) were engineered. The TOS-TPP-Obt-NPs entered into acidic lysosomes via macropinocytosis, followed by lysosomal escape and finally homed into mitochondria over a period of 24 h. Subsequently, these TOS-TPP-Obt-NPs triggered mitochondrial outer membrane permeabilization (MOMP) by inhibiting antiapoptotic Bcl-2, leading to Cytochrome C release. These TOS-TPP-Obt-NPs mediated mitochondrial damage induced cellular apoptosis through caspase-9 and caspase-3 cleavage to show improved efficacy in HeLa cells. Moreover, TOS-TPP-Obt-NPs induced MOMP in drug-resistant triple negative breast cancer cells (MDA-MB-231), leading to remarkable efficacy, compared to the combination of free drugs in higher drug concentrations. Results presented here clearly stimulate the usage of multiple drugs to perturb simultaneously diverse targets, selectively in mitochondria, as next-generation cancer therapeutics.
Collapse
Affiliation(s)
- Abhik Mallick
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune , Pune, 411008, Maharashtra, India
| | - Piyush More
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune , Pune, 411008, Maharashtra, India
| | - Muhammed Muazzam Kamil Syed
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune , Pune, 411008, Maharashtra, India
| | - Sudipta Basu
- Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune , Pune, 411008, Maharashtra, India
| |
Collapse
|
44
|
Nanogels fabricated from bovine serum albumin and chitosan via self-assembly for delivery of anticancer drug. Colloids Surf B Biointerfaces 2016; 146:107-13. [PMID: 27262260 DOI: 10.1016/j.colsurfb.2016.05.043] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 04/13/2016] [Accepted: 05/16/2016] [Indexed: 12/13/2022]
Abstract
In this study, bovine serum albumin (BSA) and chitosan (CS) were used to prepare BSA-CS nanogels by a simple green self-assembly technique. Then the nanogels were successfully used to entrap doxorubicin hydrochloride (DOX) with an entrapment ratio of 46.3%, aiming to realize the slow-release effect and lower the cytotoxicity of DOX. The IC50 values of DOX-loaded BSA-CS (DOX-BSA-CS) and free DOX obtained by MTT assay in SGC7901 cells were 0.22 and 0.05μg/mL, respectively. The cytotoxicity of DOX significantly decreased within 24h after encapsulation by the nanogels, indicating that the loaded drug could slowly release within 24h and the BSA-CS was a good slow release system. The cellular uptake experiments indicated DOX-BSA-CS diffused faster into the cancer cell than the bare drug. The flow cytometry and TUNEL assay proved DOX-BSA-CS could induce a larger apoptosis proportion of gastric cancer cells 7901 than the bare drug and it is promising to be used for curing gastric cancer.
Collapse
|
45
|
Muhammad N, Wang X, Wang K, Zhu C, Zhu Z, Jiao Y, Guo Z. Dual-drug loaded nanoformulation with a galactosamine homing moiety for liver-targeted anticancer therapy. Dalton Trans 2016; 45:13169-78. [DOI: 10.1039/c6dt01434h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A platinum(iv) complex was prepared as a prodrug of cisplatin and co-loaded with α-tocopheryl succinate into the galactosamine-modified PLGA nanoparticle for combinational chemotherapy of liver cancer.
Collapse
Affiliation(s)
- Nafees Muhammad
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology
- School of Life Sciences
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Kun Wang
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Chengcheng Zhu
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Zhenzhu Zhu
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Yang Jiao
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| |
Collapse
|
46
|
Oliveira MS, Mussi SV, Gomes DA, Yoshida MI, Frezard F, Carregal VM, Ferreira LAM. α-Tocopherol succinate improves encapsulation and anticancer activity of doxorubicin loaded in solid lipid nanoparticles. Colloids Surf B Biointerfaces 2015; 140:246-253. [PMID: 26764108 DOI: 10.1016/j.colsurfb.2015.12.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/01/2015] [Accepted: 12/13/2015] [Indexed: 12/20/2022]
Abstract
This work aimed to develop solid lipid nanoparticles (SLN) co-loaded with doxorubicin and α-tocopheryl succinate (TS), a succinic acid ester of α-tocopherol that exhibits anticancer actions, evaluating the influence of TS on drug encapsulation efficiency. The SLN were characterized for size, zeta potential, entrapment efficiency (EE), and drug release. Studies of in vitro anticancer activity were also conducted. The EE was significantly improved from 30 ± 1% to 96 ± 2% for SLN without and with TS at 0.4%, respectively. In contrast, a reduction in particle size from 298 ± 1 to 79 ± 1 nm was observed for SLN without and with TS respectively. The doxorubicin release data show that SLN provide a controlled drug release. The in vitro studies showed higher cytotoxicity for doxorubicin-TS-loaded SLN than for free doxorubicin in breast cancer cells. These findings suggest that TS-doxorubicin-loaded SLN is a promising alternative for the treatment of cancer.
Collapse
Affiliation(s)
- Mariana S Oliveira
- Department of Phamaceutics, Faculty of Pharmacy, Federal University of Minas Gerais (UFMG), Brazil
| | - Samuel V Mussi
- Department of Phamaceutics, Faculty of Pharmacy, Federal University of Minas Gerais (UFMG), Brazil
| | - Dawidson A Gomes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Brazil
| | - Maria Irene Yoshida
- Department of Chemistry, Institute of Exact Sciences, Federal University of Minas Gerais (UFMG), Brazil
| | - Frederic Frezard
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av Antônio Carlos, 6627, Campus Pampulha, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Virgínia M Carregal
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Av Antônio Carlos, 6627, Campus Pampulha, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Lucas A M Ferreira
- Department of Phamaceutics, Faculty of Pharmacy, Federal University of Minas Gerais (UFMG), Brazil.
| |
Collapse
|
47
|
Lee KY, Chiang YT, Hsu NY, Yang CY, Lo CL, Ku CA. Vitamin E containing polymer micelles for reducing normal cell cytotoxicity and enhancing chemotherapy efficacy. Acta Biomater 2015; 24:286-96. [PMID: 26087112 DOI: 10.1016/j.actbio.2015.06.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 05/13/2015] [Accepted: 06/10/2015] [Indexed: 02/03/2023]
Abstract
An α-tocopheryl succinate (α-TOS) containing diblock copolymer micellar system was used to deliver doxorubicin (Dox), an anticancer drug, for HCT116 colon cancer therapy. The α-TOS containing diblock copolymers were synthesized by conjugation of α-TOS molecules and a mPEG-b-PHEMA hydrophilic diblock copolymer by ester bonds. The Dox-loaded polymeric micelles were then obtained by solvent exchange process. In acidic surroundings such as endosomes or secondary lysosomes, the structures of the Dox-loaded polymeric micelles deformed and released the drug loads. Additionally, Dox-loaded polymeric micelles enhanced the cytotoxicity of Dox and α-TOS to cancer cells in vitro. Dox-loaded polymeric micelles also showed an exceptional tumor inhibiting effect in vivo. This study indicates that the α-TOS containing polymeric micelle system can be used as a drug carrier for cancer therapy.
Collapse
Affiliation(s)
- Kuan-Yi Lee
- Department of Biomedical Engineering, National Yang Ming University, Taipei 112, Taiwan
| | - Yi-Ting Chiang
- Department of Biomedical Engineering, National Yang Ming University, Taipei 112, Taiwan
| | - Ning-Yu Hsu
- Department of Biomedical Engineering, National Yang Ming University, Taipei 112, Taiwan
| | - Chieh-Yu Yang
- Department of Biomedical Engineering, National Yang Ming University, Taipei 112, Taiwan
| | - Chun-Liang Lo
- Department of Biomedical Engineering, National Yang Ming University, Taipei 112, Taiwan; Biophotonics & Molecular Imaging Research Center (BMIRC), National Yang Ming University, Taipei 112, Taiwan; Biomedical Engineering Research Center, National Yang Ming University, Taipei 112, Taiwan.
| | - Chen-An Ku
- Taiwan Textile Research Institute, New Taipei City 23674, Taiwan
| |
Collapse
|
48
|
Hou L, Li Y, Song H, Zhang Z, Sun Y, Zhang X, Wu K. Protective Macroautophagy Is Involved in Vitamin E Succinate Effects on Human Gastric Carcinoma Cell Line SGC-7901 by Inhibiting mTOR Axis Phosphorylation. PLoS One 2015; 10:e0132829. [PMID: 26168048 PMCID: PMC4500415 DOI: 10.1371/journal.pone.0132829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/19/2015] [Indexed: 12/26/2022] Open
Abstract
Vitamin E succinate (VES), a potential cancer therapeutic agent, potently induces apoptosis and inhibits the growth of various cancer cells. Autophagy has been supposed to promote cancer cell survival or trigger cell death, depending on particular cancer types and tumor microenvironments. The role of autophagy in the growth suppressive effect of VES on gastric cancer cell is basically unknown. We aimed to determine whether and how autophagy affected the VES-induced inhibition of SGC-7901 human gastric carcinoma cell growth. SGC-7901 cells were treated with VES or pre-treated with autophagy inhibitor, chloroquine (CQ) and 3-methyladenine (3-MA). Electron microscopy, fluorescence microscopy and Western blot were used to study whether VES induced autophagy reaction in SGC-7901 cells. Western blot evaluated the activities of the mammalian target of rapamycin (mTOR) axis. Then we used 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and flow cytometry to detect the level of cell viability and apoptosis. Collectively, our data indeed strongly support our hypothesis that VES treatment produced cytological variations that depict autophagy, increased the amount of intracellular green fluorescent protein—microtubule associated protein 1 light chain 3 (GFP-LC3) punctate fluorescence and the number of autophagic vacuoles. It altered the expression of endogenous autophagy marker LC3. VES activated the suppression of mTOR through inhibiting upstream regulators p38 MAPK and Akt. mTOR suppression consequently inhibited the activation of mTOR downstream targets p70S6K and 4E-BP-1. The activation of the upstream mTOR inhibitor AMPK had been up-regulated by VES. The results showed that pre-treatment SGC-7901 with autophagy inhibitors before VES treatment could increase the capacity of VES to reduce cell viability and to provoke apoptosis. In conclusion, VES-induced autophagy participates in SGC-7901 cell protection by inhibiting mTOR axis phosphorylation. Our findings not only strengthen our understanding of the roles of autophagy in cancer biology, but may also be useful for developing new treatments for gastric cancer patients.
Collapse
Affiliation(s)
- Liying Hou
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Yuze Li
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
- Department of the Fourth Internal Medicine, The Fourth Hospital of Heilongjiang Province, Harbin, China
| | - Huacui Song
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Zhihong Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
- Food Processing Institute, Heilongjiang Academy of Agricultural Sciences, Harbin, China
| | - Yanpei Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Xuguang Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
- Department of Internal Medicine, Hematology and Oncology, Harbin Children’s Hospital, Harbin, China
| | - Kun Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
- * E-mail:
| |
Collapse
|
49
|
San YZ, Liu Y, Zhang Y, Shi PP, Zhu YL. Peroxisome proliferator-activated receptor-γ agonist inhibits the mammalian target of rapamycin signaling pathway and has a protective effect in a rat model of status epilepticus. Mol Med Rep 2015; 12:1877-83. [PMID: 25891824 PMCID: PMC4464309 DOI: 10.3892/mmr.2015.3641] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 03/12/2015] [Indexed: 01/01/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPAR-γ) has a protective role in several neurological diseases. The present study investigated the effect of the PPAR-γ agonist, pioglitazone, on the mammalian target of rapamycin (mTOR) signaling pathway in a rat model of pentylenetetrazol (PTZ)-induced status epilepticus (SE). The investigation proceeded in two stages. First, the course of activation of the mTOR signaling pathway in PTZ-induced SE was examined to determine the time-point of peak activity, as reflected by phopshorylated (p)-mTOR/mTOR and p-S6/S6 ratios. Subsequently, pioglitazone was administrated intragastrically to investigate its effect on the mTOR signaling pathway, through western blot and immunochemical analyses. The levels of the interleukin (IL)-1β and IL-6 inflammatory cytokines were detected using ELISA, and neuronal loss was observed via Nissl staining. In the first stage of experimentation, the mTOR signaling pathway was activated, and the p-mTOR/mTOR and p-S6/S6 ratios peaked on the third day. Compared with the vehicle treated-SE group, pretreatment with pioglitazone was associated with the loss of fewer neurons, lower levels of IL-1β and IL-6, and inhibition of the activation of the mTOR signaling pathway. Therefore, the mTOR signaling pathway was activated in the PTZ-induced SE rat model, and the PPAR-γ agonist, pioglitazone, had a neuroprotective effect, by inhibiting activation of the mTOR pathway and preventing the increase in the levels of IL-1β and IL-6.
Collapse
Affiliation(s)
- Yong-Zhi San
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yu Liu
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yu Zhang
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Ping-Ping Shi
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yu-Lan Zhu
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
50
|
Mallick A, More P, Ghosh S, Chippalkatti R, Chopade BA, Lahiri M, Basu S. Dual drug conjugated nanoparticle for simultaneous targeting of mitochondria and nucleus in cancer cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:7584-98. [PMID: 25811662 DOI: 10.1021/am5090226] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Effective targeting of mitochondria has emerged as an alternative strategy in cancer chemotherapy. However, considering mitochondria's crucial role in cellular energetics, metabolism and signaling, targeting mitochondria with small molecules would lead to severe side effects in cancer patients. Moreover, mitochondrial functions are highly dependent on other cellular organelles like nucleus. Hence, simultaneous targeting of mitochondria and nucleus could lead to more effective anticancer strategy. To achieve this goal, we have developed sub 200 nm particles from dual drug conjugates derived from direct tethering of mitochondria damaging drug (α- tocopheryl succinate) and nucleus damaging drugs (cisplatin, doxorubicin and paclitaxel). These dual drug conjugated nanoparticles were internalized into the acidic lysosomal compartments of the HeLa cervical cancer cells through endocytosis and induced apoptosis through cell cycle arrest. These nanoparticles damaged mitochondrial morphology and triggered the release of cytochrome c. Furthermore, these nanoparticles target nucleus to induce DNA damage, fragment the nuclear morphology and damage the cytoskeletal protein tubulin. Therefore, these dual drug conjugated nanoparticles can be successfully used as a platform technology for simultaneous targeting of multiple subcellular organelles in cancer cells to improve the therapeutic efficacy of the free drugs.
Collapse
Affiliation(s)
- Abhik Mallick
- †Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune, Pune, 411021, Maharashtra India
| | - Piyush More
- †Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune, Pune, 411021, Maharashtra India
- §Institute of Bioinformatics and Biotechnology (IBB), University of Pune, Pune, 411007, Maharashtra India
| | - Sougata Ghosh
- §Institute of Bioinformatics and Biotechnology (IBB), University of Pune, Pune, 411007, Maharashtra India
| | - Rohan Chippalkatti
- §Institute of Bioinformatics and Biotechnology (IBB), University of Pune, Pune, 411007, Maharashtra India
- ΦDepartment of Biology, Indian Institute of Science Education and Research (IISER)-Pune, Pune, 411021, Maharashtra India
| | - Balu A Chopade
- ⊥Department of Microbiology, Savitribai Phule Pune University, Pune, 411007, Maharashtra India
| | - Mayurika Lahiri
- ΦDepartment of Biology, Indian Institute of Science Education and Research (IISER)-Pune, Pune, 411021, Maharashtra India
| | - Sudipta Basu
- †Department of Chemistry, Indian Institute of Science Education and Research (IISER)-Pune, Pune, 411021, Maharashtra India
| |
Collapse
|