1
|
Xu X, Huang Z, Han H, Yu Z, Ye L, Zhao Z, Qian Y, Li Y, Zhao R, Zhang T, Liu Y, Cai J, Lin S, Zhai E, Chen J, Cai S. N 7-methylguanosine tRNA modification promotes gastric cancer progression by activating SDHAF4-dependent mitochondrial oxidative phosphorylation. Cancer Lett 2025; 615:217566. [PMID: 39965707 DOI: 10.1016/j.canlet.2025.217566] [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: 07/23/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
N7-methylguanosine (m7G) tRNA modification is closely implicated in tumor occurrence and development. However, the precise function and molecular mechanisms of m7G tRNA modification in gastric cancer (GC) remain unclear. In this study, we evaluated the expression and function of methyltransferase-like 1 (METTL1) and WD repeat domain 4 (WDR4) in GC and elucidated the mechanisms underlying the role of METTL1/WDR4-mediated m7G tRNA modifications in promoting GC progression. Upregulation of m7G methyltransferase complex proteins, METTL1 and WDR4, in GC tissues significantly correlates with poor patient prognosis. Functionally, METTL1 and WDR4 facilitate GC progression in vitro and in vivo. Mechanistically, METTL1 knockdown reduces the expression of m7G-modified tRNAs and attenuates the translation of oncogenes enriched in pathways associated with oxidative phosphorylation. Furthermore, METTL1 strengthens mitochondrial electron transport chain complex II (ETC II) activity by promoting succinate dehydrogenase assembly factor 4 (SDHAF4) translation, thereby accelerating GC metabolism and progression. Forced expression of SDHAF4 and chemical modulators of ETC II could reverse the effects of METTL1 on mouse GC. Collectively, our findings delineate the oncogenic role and molecular mechanisms of METTL1/WDR4-mediated m7G tRNA modifications in GC progression, suggesting METTL1/WDR4 and its downstream signaling axis as potential therapeutic targets for GC.
Collapse
Affiliation(s)
- Xiang Xu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Zhixin Huang
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Hui Han
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Zihan Yu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Linying Ye
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Zeyu Zhao
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Yan Qian
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, Guangdong, China
| | - Risheng Zhao
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Tianhao Zhang
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Yinan Liu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Junchao Cai
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510000, Guangdong, China
| | - Shuibin Lin
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Ertao Zhai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| | - Jianhui Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China; Department of General Surgery, Guangxi Hospital Division of the First Affiliated Hospital, Sun Yat-sen University, Nanning, 530000, Guangxi, China.
| | - Shirong Cai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
2
|
Lei L, Song Y, Yang L, Wang Y, Xia X, Zhang Y, Zhang X, Zhang X, Duggal I, He B, Peppas NA, Cao J, Gao H. Triethylamine-mediated protonation-deprotonation unlocks dual-drug self assembly to suppress breast cancer progression and metastasis. Proc Natl Acad Sci U S A 2025; 122:e2416796122. [PMID: 39874295 PMCID: PMC11804646 DOI: 10.1073/pnas.2416796122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
Carrier-free nanomedicines exhibited significant potential in elevating drug efficacy and safety for tumor management, yet their self assembly typically relied on chemical modifications of drugs or the incorporation of surfactants, thereby compromising the drug's inherent pharmacological activity. To address this challenge, we proposed a triethylamine (TEA)-mediated protonation-deprotonation strategy that enabled the adjustable-proportion self assembly of dual drugs without chemical modification, achieving nearly 100% drug loading capacity. Molecular dynamic simulations, supported by experiment evidence, elucidated the underlying self-assembly mechanism. Specifically, TEA facilitated the deprotonation of Doxorubicin (Dox) and α-Tocopherol succinate (α-tos), causing Dox to transition from a hydrophilic to a hydrophobic state, while simultaneously increasing the hydrophilicity of α-tos. This allowed for a fine-tuned balance between the hydrophilic and hydrophobic properties of the two compounds, enabling their precise self assembly into a carrier-free nanomedicine (DT) with a tailored drug ratio. The engineered DT demonstrated the ability to accumulate at the tumor sites and release its therapeutic drugs in a controlled manner. The combination of Dox and α-tos synergistically generated reactive oxygen species and modulated the expression of tumor matrix metalloproteinase-9, leading to superior antitumor efficacy without significant metastasis, while maintaining excellent safety profiles. Our findings provided unique perspectives on the design of carrier-free nanomedicine for cancer therapy, thereby laying a solid foundation for its potential clinical translation.
Collapse
Affiliation(s)
- Lei Lei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Yujun Song
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Lianyi Yang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Yazhen Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Yiwei Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| | - Xiaoxian Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Xuequan Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Ishaan Duggal
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX78712
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Nicholas A. Peppas
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX78712
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX78712
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX78712
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX78712
- Department of Surgery and Perioperative Care, Dell Medical School, Austin, TX78712
- Department of Pediatrics, Dell Medical School, Austin, TX78712
| | - Jun Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu610064, People’s Republic of China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, People’s Republic of China
| |
Collapse
|
3
|
Cui YH, Wei J, Fan H, Li W, Zhao L, Wilkinson E, Peterson J, Xie L, Zou Z, Yang S, Applebaum MA, Kline J, Chen J, He C, He YY. Targeting DTX2/UFD1-mediated FTO degradation to regulate antitumor immunity. Proc Natl Acad Sci U S A 2024; 121:e2407910121. [PMID: 39661064 DOI: 10.1073/pnas.2407910121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Here, we show that vitamin E succinate (VES) acts as a degrader for the m6A RNA demethylase fat mass and obesity-associated protein (FTO), thus suppressing tumor growth and resistance to immunotherapy. FTO is ubiquitinated by its E3 ligase DTX2, followed by UFD1 recruitment and subsequent degradation in the proteasome. VES binds to FTO and DTX2, leading to enhanced FTO-DTX2 interaction, FTO ubiquitination, and degradation in FTO-dependent tumor cells. VES suppressed tumor growth and enhanced antitumor immunity and response to immunotherapy in vivo in mouse models. Genetic FTO knockdown or VES treatment increased m6A methylation in the LIF (Leukemia Inhibitory Factor) gene and decreased LIF mRNA decay, and thus sensitized melanoma cells to T cell-mediated cytotoxicity. Taken together, our findings reveal the underlying molecular mechanism for FTO protein degradation and identify a dietary degrader for FTO that inhibits tumor growth and overcomes immunotherapy resistance.
Collapse
Affiliation(s)
- Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Jiangbo Wei
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Hao Fan
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Wenlong Li
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Lijie Zhao
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Emma Wilkinson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637
| | - Jack Peterson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- The College, University of Chicago, Chicago, IL 60637
| | - Lishi Xie
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Zhongyu Zou
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Mark A Applebaum
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637
| | - Justin Kline
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Jing Chen
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Chuan He
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
- HHMI, University of Chicago, Chicago, IL 60637
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
4
|
Ma G, Chong W, Qi Y, Lu Z, Zhang Z, Nian B, Hu Y. Can vitamin E ester derivatives be excellent alternatives of vitamin E: state of art. Bioprocess Biosyst Eng 2023; 46:1695-1709. [PMID: 37555945 DOI: 10.1007/s00449-023-02918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023]
Abstract
Vitamin E (VE) is a natural antioxidant which is widely used in the food fields, while the shortcomings of easy oxidative inactivation and poor water solubility limit its application. Vitamin E esters' (VEEs) derivatives, such as vitamin E acetate (VEA), are more stable and easier to be absorbed while have similar biological activities and physiological functions compared with VE. In this systematic review, the digestion, absorption and physiological function of VEEs were summarized. To promote their further industrial applications, the synthesis strategies of VEEs were also summarized in-depth. In particular, as a new generation of green solvents, ionic liquids (ILs) have been widely used in enzymatic reactions due to the stabilization and activation of enzymes. Their applications in enzymatic synthesis of VEEs were summarized and discussed. Finally, several future perspectives for developing more efficiency strategies of VEEs synthesis, such as enzyme engineering and design of novel ILs, were also discussed.
Collapse
Affiliation(s)
- Guangzheng Ma
- State Key Laboratory of Materials-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Technology University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Wenya Chong
- State Key Laboratory of Materials-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Technology University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Yuan Qi
- State Key Laboratory of Materials-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Technology University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Zeping Lu
- State Key Laboratory of Materials-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Technology University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Zihan Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Technology University, Nanjing, 210009, Jiangsu Province, People's Republic of China
| | - Binbin Nian
- State Key Laboratory of Materials-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Technology University, Nanjing, 210009, Jiangsu Province, People's Republic of China.
| | - Yi Hu
- State Key Laboratory of Materials-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Technology University, Nanjing, 210009, Jiangsu Province, People's Republic of China.
| |
Collapse
|
5
|
Fadaka AO, Akinsoji T, Klein A, Madiehe AM, Meyer M, Keyster M, Sikhwivhilu LM, Sibuyi NRS. Stage-specific treatment of colorectal cancer: A microRNA-nanocomposite approach. J Pharm Anal 2023; 13:1235-1251. [PMID: 38174117 PMCID: PMC10759263 DOI: 10.1016/j.jpha.2023.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 01/05/2024] Open
Abstract
Colorectal cancer (CRC) is among the leading causes of cancer mortality. The lifetime risk of developing CRC is about 5% in adult males and females. CRC is usually diagnosed at an advanced stage, and at this point therapy has a limited impact on cure rates and long-term survival. Novel and/or improved CRC therapeutic options are needed. The involvement of microRNAs (miRNAs) in cancer development has been reported, and their regulation in many oncogenic pathways suggests their potent tumor suppressor action. Although miRNAs provide a promising therapeutic approach for cancer, challenges such as biodegradation, specificity, stability and toxicity, impede their progression into clinical trials. Nanotechnology strategies offer diverse advantages for the use of miRNAs for CRC-targeted delivery and therapy. The merits of using nanocarriers for targeted delivery of miRNA-formulations are presented herein to highlight the role they can play in miRNA-based CRC therapy by targeting different stages of the disease.
Collapse
Affiliation(s)
- Adewale Oluwaseun Fadaka
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, 7535, South Africa
| | - Taiwo Akinsoji
- School of Medicine, Southern Illinois University, Springfield, IL, 62702, USA
| | - Ashwil Klein
- Plant Omics Laboratory, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, 7535, South Africa
| | - Abram Madimabe Madiehe
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, 7535, South Africa
- Nanobiotechnology Research Group, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, 7535, South Africa
| | - Mervin Meyer
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, 7535, South Africa
| | - Marshall Keyster
- Environmental Biotechnology Laboratory, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, 7535, South Africa
| | - Lucky Mashudu Sikhwivhilu
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Advanced Materials Division, Mintek, Johannesburg, 2125, South Africa
- Department of Chemistry, Faculty of Science, Engineering and Agriculture, University of Venda, Thohoyandou, 0950, South Africa
| | - Nicole Remaliah Samantha Sibuyi
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, Faculty of Natural Sciences, University of the Western Cape, Bellville, 7535, South Africa
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Advanced Materials Division, Mintek, Johannesburg, 2125, South Africa
| |
Collapse
|
6
|
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
|
7
|
Suksiriworapong J, Achayawat C, Juangrattanakamjorn P, Taresco V, Crucitti VC, Sakchaisri K, Bunsupa S. Modification of Poly(Glycerol Adipate) with Tocopherol and Cholesterol Modulating Nanoparticle Self-Assemblies and Cellular Responses of Triple-Negative Breast Cancer Cells to SN-38 Delivery. Pharmaceutics 2023; 15:2100. [PMID: 37631315 PMCID: PMC10459774 DOI: 10.3390/pharmaceutics15082100] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/27/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
This study aimed to fabricate new variations of glycerol-based polyesters by grafting poly(glycerol adipate) (PGA) with hydrophobic bioactive moieties, tocopherol (TOC), and cholesterol (CHO). Their effects on nanoparticle (NP) formation, drug release, and cellular responses in cancer and normal cells were evaluated. CHO and TOC were successfully grafted onto PGA backbones with 30% and 50% mole grafting. Increasing the percentage of mole grafting in both molecules increased the glass transition temperature and water contact angle of the final polymers but decreased the critical micelle concentration of the formulated particles. PGA-TOC NPs reduced the proliferation of MDA-MB-231 cancer cells. However, they enhanced the proliferation of primary dermal fibroblasts within a specific concentration range. PGA-CHO NPs minimally affected the growth of cancer and normal cells. Both types of NPs did not affect apoptosis or the cell cycle of cancer cells. PGA-CHO and PGA-TOC NPs were able to entrap SN-38, a hydrophobic anticancer drug, with a particle size <200 nm. PGA-CHO NPs had a higher drug loading capacity and a greater drug release than PGA-TOC NPs. However, SN-38-loaded PGA-TOC NPs showed higher toxicity than SN-38 and SN-38-loaded PGA-CHO NPs due to the combined effects of antiproliferation and higher cellular uptake. Compared with SN-38, the drug-loaded NPs more profoundly induced sub-G1 in the cell cycle analysis and apoptosis of cancer cells in a similar pattern. Therefore, PGA-CHO and PGA-TOC polymers have potential applications as delivery systems for anticancer drugs.
Collapse
Affiliation(s)
| | - Chittin Achayawat
- Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | | | - Vincenzo Taresco
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Valentina Cuzzucoli Crucitti
- Centre for Additive Manufacturing and Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, UK
| | - Krisada Sakchaisri
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Somnuk Bunsupa
- Department of Pharmacognosy, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
8
|
Qi Z, Shi J, Song Y, Deng Y. A novel micellar carrier to reverse multidrug resistance of tumours: TPGS derivatives with end-grafted cholesterol. J Drug Target 2023; 31:537-553. [PMID: 37092957 DOI: 10.1080/1061186x.2023.2205614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
D-α-tocopherol polyethylene glycol succinate (TPGS) has good biocompatibility, low immunogenicity, prolonged circulation time, and it can reverse multidrug resistance of tumours. However, the micelle concentration (CMC) of TPGS is too high (0.2 mg/mL) to develop the formulation of the micelle. In this study, TPGS was modified with cholesterol to obtain a new carrier material, TPGS-CHMC. The CMC of TPGS-CHMC was 2 μg/mL, which was extremely lower than that of TPGS. Docetaxel (DTX)-loaded TPGS-CHMC micelles (TPGS-CHMC/DTX) exhibited an average size of approximately 13 nm, a zeta potential of approximately -4.66 mV, and high encapsulation efficiency (99.2 ± 0.6%). TPGS-CHMC reduced mitochondrial membrane potential and cell membrane fluidity in paclitaxel-resistant ovarian cancer cells (A2780/T). In vivo, DiR-loaded TPGS-CHMC micelles were selectively distributed in A2780/T tumour-bearing nude mice. In A2780/T tumour-bearing nude mice, TPGS-CHMC/DTX micelles displayed significantly higher anti-tumour activity and less toxicity than the free DTX solution. In summary, TPGS-CHMC has various advantages, and provides a new option for developing functional polymeric micelles.
Collapse
Affiliation(s)
- Zhaowei Qi
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jia Shi
- The first affiliated hospital of Jinzhou medical university, Jinzhou, Liaoning, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| |
Collapse
|
9
|
Ray MN, Ozono M, Nakao M, Sano S, Kogure K. Only one carbon difference determines the pro-apoptotic activity of α-tocopheryl esters. FEBS J 2023; 290:1027-1048. [PMID: 36083714 DOI: 10.1111/febs.16623] [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: 02/21/2022] [Revised: 07/19/2022] [Accepted: 09/08/2022] [Indexed: 11/27/2022]
Abstract
α-Tocopheryl succinate (TS), a redox-silent succinyl ester of natural α-Tocopherol, has emerged as a novel anti-cancer agent. However, the underlying mechanism is unclear. We found that the terminal dicarboxylic moiety of tocopheryl esters contributes to apoptosis induction and thus cytotoxicity. To further examine this relationship, we compared the pro-apoptotic activity of TS, which has four carbon atoms in the terminal dicarboxylic moiety, to that of a newly synthesized, tocopheryl glutarate (Tglu), which has five. Cytotoxicity assays in vitro confirmed that TS stimulated apoptosis, while Tglu was non-cytotoxic. In investigating biological mechanisms leading to these opposing effects, we found that TS caused an elevation of intracellular superoxide, but Tglu did not. TS increased intracellular Ca2+ in cultured cells, suggesting induction of endoplasmic reticulum (ER) stress; however, Tglu did not affect Ca2+ homeostasis. 1,4,5-trisphosphate (IP3 ) receptor antagonist 2-Aminoethyl diphenylborinate (2-APB) decreased TS-induced intracellular Ca2+ , restored mitochondrial activity and cell viability in TS-treated cells, establishing the ER-mitochondria relationship in apoptosis induction. Moreover, real-time PCR, immunostaining and Western blotting assays revealed that TS downregulated glucose-regulated protein 78 (GRP78), which maintains ER homeostasis and promotes cell survival. Conversely, Tglu upregulates GRP78. Taken together, our results suggest a model in which TS-mediated superoxide production and GRP78 inhibition induce ER stress, which elevates intracellular Ca2+ and depolarizes mitochondria, leading to apoptosis. Because Tglu does not affect superoxide generation and increases GRP78 expression, it inhibits ER stress and is thereby non-cytotoxic. Our research provides insight into the structure-activity relationship of tocopheryl esters regarding the induction of apoptosis.
Collapse
Affiliation(s)
- Manobendro Nath Ray
- Department of Pharmaceutical Health Chemistry, Graduate School of Pharmaceutical Sciences, Tokushima University, Japan
| | - Mizune Ozono
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Japan
| | - Michiyasu Nakao
- Department of Molecular Medicinal Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Japan
| | - Shigeki Sano
- Department of Molecular Medicinal Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Japan
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Japan
| |
Collapse
|
10
|
Mondal SK, Jinka S, Shankar G, Srinivas R, Banerjee R. Modification of α-Tocopherol Succinate with a Tumor-targeting Peptide Conjugate Enhances the Antitumor Efficacy of a Paclitaxel-loaded Lipid Aggregate. Chem Asian J 2023; 18:e202201136. [PMID: 36482874 DOI: 10.1002/asia.202201136] [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: 11/09/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Paclitaxel (PTX) is a widely used chemotherapeutic agent in the clinic. However, its clinical benefit is limited due to its low water solubility, off-target toxicity, and for being a multidrug-resistant (MDR) substrate. To overcome these limitations in this study, a tumor-targeting peptide (CRGDK peptide, a ligand for NRP-1 receptor) conjugate of α-tocopheryl succinate (α-TOS) was synthesized and modified on PTX-loaded lipid aggregate (TL-PTX) to leverage the benefits of α-TOS, which include a) anti-cancer activity, b) increased PTX loading, and c) inhibition of MDR activity. Use of peptide conjugate of α-TOS (α-TOS-CRGDK) in lipid aggregate increased PTX entrapment efficiency by 20%, helped in NRP-1 specific cellular uptake and significantly enhanced apoptotic and cell killing activity (p <0.01) of PTX compared to control formulation (CL-PTX) by inhibiting MDR-activity in melanoma resulting in ∼70% increment in overall survival of melanoma tumor-bearing mice. In conclusion, CRGDK- α-TOS conjugate in association with PTX-loaded liposome provided a unique NRP-1 targeted, drug-resistant reversing anticancer regimen for treating aggressive melanoma.
Collapse
Affiliation(s)
- Sujan Kumar Mondal
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, India
- Department of Radiology, Michigan State University, East Lansing, Michigan (USA
| | - Sudhakar Jinka
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, India
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gajji Shankar
- Mass Spectrometry Division, CSIR - Indian Institute of Chemical Technology (CSIRIICT), Uppal Road, Tarnaka, Hyderabad, 500 007, Telangana State, India
| | - Ragampeta Srinivas
- Mass Spectrometry Division, CSIR - Indian Institute of Chemical Technology (CSIRIICT), Uppal Road, Tarnaka, Hyderabad, 500 007, Telangana State, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad, India
| |
Collapse
|
11
|
Marotta C, Giorgi E, Binacchi F, Cirri D, Gabbiani C, Pratesi A. An overview of recent advancements in anticancer Pt(IV) prodrugs: New smart drug combinations, activation and delivery strategies. Inorganica Chim Acta 2023. [DOI: 10.1016/j.ica.2023.121388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
12
|
Date T, Kuche K, Chaudhari D, Ghadi R, Sahel DK, Chitkara D, Jain S. Hitting Multiple Cellular Targets in Triple-Negative Breast Cancer Using Dual-Action Cisplatin(IV) Prodrugs for Safer Synergistic Chemotherapy. ACS Biomater Sci Eng 2022; 8:2349-2362. [PMID: 35522530 DOI: 10.1021/acsbiomaterials.1c01582] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Triple-negative breast cancer (TNBC) cells show improved sensitivity for cisplatin therapy due to their defective DNA damage repair system. However, the clinical utilization of cisplatin is limited by dose-dependent systemic toxicities and chemoresistance. Cisplatin Pt(IV) derivatives having kinetically inert octahedral geometry provide an effective strategy to overcome these limitations. Upon cellular reduction, these derivatives release cisplatin and axial ligands, acting as dual-action prodrugs. Hereby, we have developed three cisplatin(IV) conjugates using distinct bioactive axial moieties (valproate, tocopherol, and chlorambucil), which can synergistically complement cisplatin activity and attack multiple cellular targets. The designed derivatives showcased enhanced antiproliferative activity and improved therapeutic synergism along with a noteworthy cisplatin dose reduction index in a panel of six cancer cells. These Pt(IV) derivatives remarkably improved cellular drug uptake and showed lower dependency on copper transporter 1 (Ctr1) for uptake than cisplatin. The results of enhanced in vitro activity were well corroborated by in vivo efficacy testing in the 4T1 cell-based TNBC model, showcasing ∼2-7-folds higher tumor volume reduction for Pt(IV) derivatives than cisplatin. In addition, the designed derivatives significantly reduced the nephrotoxicity risk involved in cisplatin therapy, indicated by systemic toxicity biomarkers and organ histopathology. The results indicated that cisplatin(IV) derivatives could open new avenues for safer synergistic chemotherapy in TNBC.
Collapse
Affiliation(s)
- Tushar Date
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali 160062, Punjab, India
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali 160062, Punjab, India
| | - Dasharath Chaudhari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali 160062, Punjab, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali 160062, Punjab, India
| | - Deepak Kumar Sahel
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector 67, Mohali 160062, Punjab, India
| |
Collapse
|
13
|
Marathe S, Shadambikar G, Mehraj T, Sulochana SP, Dudhipala N, Majumdar S. Development of α-Tocopherol Succinate-Based Nanostructured Lipid Carriers for Delivery of Paclitaxel. Pharmaceutics 2022; 14:1034. [PMID: 35631620 PMCID: PMC9145488 DOI: 10.3390/pharmaceutics14051034] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 12/21/2022] Open
Abstract
The management of retinoblastoma (RB) involves the use of invasive treatment regimens. Paclitaxel (PTX), an effective antineoplastic compound used in the treatment of a wide range of malignant tumors, poses treatment challenges due to systemic toxicity, rapid elimination, and development of resistance. The goal of this work was to develop PTX-loaded, α-tocopherol succinate (αTS)-based, nanostructured lipid carrier (NLCs; αTS-PTX-NLC) and PEGylated αTS-PTX-NLC (αTS-PTX-PEG-NLC) to improve ocular bioavailability. The hot homogenization method was used to prepare the NLCs, and repeated measures ANOVA analysis was used for formulation optimization. αTS-PTX-NLC and αTS-PTX-PEG-NLC had a mean particle size, polydispersity index and zeta potential of 186.2 ± 3.9 nm, 0.17 ± 0.03, −33.2 ± 1.3 mV and 96.2 ± 3.9 nm, 0.27 ± 0.03, −39.15 ± 3.2 mV, respectively. The assay and entrapment efficiency of both formulations was >95.0%. The NLC exhibited a spherical shape, as seen from TEM images. Sterilized (autoclaved) formulations were stable for up to 60 days (last time point checked) under refrigerated conditions. PTX-NLC formulations exhibited an initial burst release and 40% drug release, overall, in 48 h. The formulations exhibited desirable physicochemical properties and could lead to an effective therapeutic option in the management of RB.
Collapse
Affiliation(s)
- Sushrut Marathe
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS 38677, USA; (S.M.); (G.S.); (T.M.); (S.P.S.); (N.D.)
| | - Gauri Shadambikar
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS 38677, USA; (S.M.); (G.S.); (T.M.); (S.P.S.); (N.D.)
| | - Tabish Mehraj
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS 38677, USA; (S.M.); (G.S.); (T.M.); (S.P.S.); (N.D.)
| | - Suresh P. Sulochana
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS 38677, USA; (S.M.); (G.S.); (T.M.); (S.P.S.); (N.D.)
| | - Narendar Dudhipala
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS 38677, USA; (S.M.); (G.S.); (T.M.); (S.P.S.); (N.D.)
| | - Soumyajit Majumdar
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS 38677, USA; (S.M.); (G.S.); (T.M.); (S.P.S.); (N.D.)
- Research Institute of Pharmaceutical Sciences, University of Mississippi, Oxford, MS 38677, USA
| |
Collapse
|
14
|
Kumbhar PS, Nadaf S, Manjappa AS, Jha NK, Shinde SS, Chopade SS, Shete AS, Disouza JI, Sambamoorthy U, Kumar SA. D-ɑ-tocopheryl polyethylene glycol succinate: A review of multifarious applications in nanomedicines. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
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
|
16
|
Fialova JL, Raudenska M, Jakubek M, Kejik Z, Martasek P, Babula P, Matkowski A, Filipensky P, Masarik M. Novel Mitochondria-targeted Drugs for Cancer Therapy. Mini Rev Med Chem 2021; 21:816-832. [PMID: 33213355 DOI: 10.2174/1389557520666201118153242] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/08/2020] [Accepted: 10/16/2020] [Indexed: 11/22/2022]
Abstract
The search for mitochondria-targeted drugs has dramatically risen over the last decade. Mitochondria are essential organelles serving not only as a powerhouse of the cell but also as a key player in cell proliferation and cell death. Their central role in the energetic metabolism, calcium homeostasis and apoptosis makes them an intriguing field of interest for cancer pharmacology. In cancer cells, many mitochondrial signaling and metabolic pathways are altered. These changes contribute to cancer development and progression. Due to changes in mitochondrial metabolism and changes in membrane potential, cancer cells are more susceptible to mitochondria-targeted therapy. The loss of functional mitochondria leads to the arrest of cancer progression and/or a cancer cell death. Identification of mitochondrial changes specific for tumor growth and progression, rational development of new mitochondria-targeted drugs and research on delivery agents led to the advance of this promising area. This review will highlight the current findings in mitochondrial biology, which are important for cancer initiation, progression and resistance, and discuss approaches of cancer pharmacology with a special focus on the anti-cancer drugs referred to as 'mitocans'.
Collapse
Affiliation(s)
- Jindriska Leischner Fialova
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Martina Raudenska
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Milan Jakubek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, CZ-121 08 Prague, Czech Republic
| | - Zdenek Kejik
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, CZ-121 08 Prague, Czech Republic
| | - Pavel Martasek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, CZ-121 08 Prague, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Adam Matkowski
- Department of Pharmaceutical Biology and Botany, Wroclaw Medical University, 50556 Borowska 211, Poland
| | - Petr Filipensky
- Department of Urology, St. Anne's Faculty Hospital, CZ-65691 Brno, Czech Republic
| | - Michal Masarik
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| |
Collapse
|
17
|
Liang L, Peng Y, Qiu L. Mitochondria-targeted vitamin E succinate delivery for reversal of multidrug resistance. J Control Release 2021; 337:117-131. [PMID: 34274383 DOI: 10.1016/j.jconrel.2021.07.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 07/11/2021] [Accepted: 07/14/2021] [Indexed: 12/31/2022]
Abstract
Inducing mitochondrial malfunction is an appealing strategy to overcome tumor multidrug resistance (MDR). Reported here a versatile mitochondrial-damaging molecule, vitamin E succinate (VES), is creatively utilized to assist MDR reversal of doxorubicin hydrochloride (DOX·HCl) via a nanovesicle platform self-assembled from amphiphilic polyphosphazenes containing pH-sensitive 1H-benzo-[d]imidazol-2-yl) methanamine (BIMA) groups. Driven by multiple non-covalent interactions, VES is fully introduced into the hydrophobic membrane of DOX·HCl-loaded nanovesicles with loading content of 23.5%. The incorporated VES also offers robust anti-leakage property toward DOX·HCl under normal physiological conditions. More importantly, upon release within acidic tumor cells, VES can target mitochondria and result in various dysfunctions including excessive generation of reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨm) loss, and inhibited adenosine triphosphate (ATP) synthesis, which contribute to cell apoptosis and insufficient energy supply for drug efflux pumps. Consequently, the killing-effect of DOX·HCl is significantly enhanced toward drug resistant cancer cells at the optimal mass ratio of DOX·HCl to VES. Further in vivo antitumor investigation on nude mice bearing xenograft drug-resistant human chronic myelogenous leukemia K562/ADR tumors verifies the extremely enhanced anti-tumor efficacy of the dual drug-loaded nanovesicle with the tumor inhibition rate (TIR) of 82.38%. Collectively, this study provides a s safe, facile and promising strategy for both precise drug delivery and MDR eradication to improve cancer therapy.
Collapse
Affiliation(s)
- Lina Liang
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yan Peng
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Liyan Qiu
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
18
|
Zheng RR, Zhao LP, Liu LS, Deng FA, Chen XY, Jiang XY, Wang C, Yu XY, Cheng H, Li SY. Self-delivery nanomedicine to overcome drug resistance for synergistic chemotherapy. Biomater Sci 2021; 9:3445-3452. [PMID: 33949456 DOI: 10.1039/d1bm00119a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Multidrug resistance (MDR) is one of the prime reasons for the failure of cancer chemotherapy, which continues to be a great challenge to be solved. In this work, α-tocopherol succinate (α-TOS) and doxorubicin (DOX)-based self-delivery nanomedicine (designated as α-TD) is prepared to combat drug resistance for cancer synergistic chemotherapy. Carrier-free α-TD possesses a fairly high drug loading rate and improves the cellular uptake via the endocytosis pathway. More importantly, the apoptotic inducer α-TOS could elevate the reactive oxygen species (ROS) generation, disrupt mitochondrial function and reduce adenosine 5'-triphosphate (ATP) production, which facilitate the intracellular drug retention while decreasing its efflux. As a result, α-TD achieves a considerable synergistic chemotherapeutic effect against drug resistant cancer cells. Moreover, it also exhibits a preferable inhibitory effect on tumor growth with a low system toxicity in vivo. This synergistic drug self-delivery strategy would open a new window for developing carrier-free nanomedicine for overcoming drug resistance in cancer therapy.
Collapse
Affiliation(s)
- Rong-Rong Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | - Lin-Ping Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | - Ling-Shan Liu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Fu-An Deng
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | - Xia-Yun Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | - Xue-Yan Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | - Chang Wang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | - Hong Cheng
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Shi-Ying Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| |
Collapse
|
19
|
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
|
20
|
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
|
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
|
Yan G, Chen R, Xiong N, Song J, Wang X, Tang R. pH-sensitive small molecule nanodrug self-assembled from amphiphilic vitamin B6-E analogue conjugate for targeted synergistic cancer therapy. Colloids Surf B Biointerfaces 2020; 191:111000. [PMID: 32247946 DOI: 10.1016/j.colsurfb.2020.111000] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023]
Abstract
To promote the targeted cancer therapy, the pH-sensitive small molecule nanodrug self-assembled from amphiphilic vitamin B6-E analogue conjugate was successfully constructed. Herein, water-soluble vitamin B6 with pKa (5.6) was chemically conjugated to lipid-soluble vitamin E succinate (α-TOS), which showed selective cancer cell killing ability and this amphiphilic small molecule vitamin conjugate could self-assemble to be free nanoparticles (NPs) and doxorubicin-loaded NPs (α-TOS-B6-NPs-DOX). The small molecule nanodrugs could perform the following characteristic: (i) stability in the sodium dodecyl sulfonate (SDS) solution and long-term storage stability in PBS via surface negative charge; (ii) tumor accumulation by enhanced penetration and retention (EPR) effect; (iii) improved cellular internalization by means of vitamin B6 transporting membrane carrier (VTC); and (iv) facilitating endosomal escape and rapid drug release for synergistic toxicity to tumor cells via charge reversal and ester hydrolysis at intracellular pH and/or esterase. Moreover, α-TOS-B6-NPs-DOX exhibited long blood circulation stability and significant tumor accumulation and inhibition with the decreased side effects in vivo. Thus, the pH-sensitive small molecule nanodrug self-assembled from amphiphilic vitamin B6-E analogue conjugate could be the potential drug carriers in targeted synergistic cancer therapy.
Collapse
Affiliation(s)
- Guoqing Yan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Ran Chen
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Nanchi Xiong
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Jiayu Song
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province, 230601, PR China.
| |
Collapse
|
23
|
Wu Y, Liu J, Movahedi F, Gu W, Xu T, Xu ZP. Enhanced Prevention of Breast Tumor Metastasis by Nanoparticle-Delivered Vitamin E in Combination with Interferon-Gamma. Adv Healthc Mater 2020; 9:e1901706. [PMID: 32052565 DOI: 10.1002/adhm.201901706] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/16/2020] [Indexed: 12/15/2022]
Abstract
Preventing cancer metastasis is one of the remaining challenges in cancer therapy. As an efficient natural product, alpha-tocopheryl succinate (α-TOS), the most effective form of vitamin E, holds great anticancer potential. To improve its efficacy and bioavailability, lipid-coated calcium carbonate/phosphate (LCCP) nanoparticles (NPs) with folic acid and PEG modification are synthesized for efficient delivery of α-TOS to 4T1 cancer cells. The optimized LCCP-FA NPs (NP-TOS15) show an α-TOS loading efficiency of around 60%, and enhanced uptake by 4T1 metastatic cancer cells. Consequently, NP-TOS15 significantly enhance the anticancer effect in combination with interferon-gamma (IFN-γ) in terms of apoptosis facilitation and migration inhibition. Importantly, NP-TOS15 upregulate the anticancer immunity via downregulating program death ligand 1 (PD-L1) expression that is initially induced by IFN-γ, and remarkably prevent the lung metastasis, particularly in combination with IFN-γ. Further investigation reveals that this combination therapy also modulates the cytotoxic lymphocyte infiltration into the tumor microenvironment for tumor elimination. Taken together, the NP delivery of α-TOS in combination with IFN-γ provides an applicable strategy for cancer therapy.
Collapse
Affiliation(s)
- Yilun Wu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Fatemeh Movahedi
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Tiefeng Xu
- The First Affiliated Hospital of Hainan Medical University Cancer Institute of Hainan Medical University Haikou Hainan 570102 China
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| |
Collapse
|
24
|
Mitophagy contributes to alpha-tocopheryl succinate toxicity in GSNOR-deficient hepatocellular carcinoma. Biochem Pharmacol 2020; 176:113885. [PMID: 32112881 DOI: 10.1016/j.bcp.2020.113885] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
The downregulation of the denitrosylating enzyme S-nitrosoglutathione reductase (GSNOR, EC:1.1.1.284), is a feature of hepatocellular carcinoma (HCC). This condition causes mitochondrial rearrangements that sensitize these tumors to mitochondrial toxins, in particular to the mitochondrial complex II inhibitor alpha-tocopheryl succinate (αTOS). It has also been reported the GSNOR depletion impairs the selective degradation of mitochondria through mitophagy; however, if this contributes to GSNOR-deficient HCC cell sensitivity to αTOS and can be applied to anticancer therapies, is still not known. Here, we provide evidence that GSNOR-deficient HCC cells show defective mitophagy which contributes to αTOS toxicity. Mitophagy inhibition by Parkin (EC: 2.3.2.31) depletion enhances αTOS anticancer effects, thus suggesting that this drug could be effective in treating mitophagy-defective tumors.
Collapse
|
25
|
Cheng X, Zeng X, Zheng Y, Fang Q, Wang X, Wang J, Tang R. pH-sensitive pluronic micelles combined with oxidative stress amplification for enhancing multidrug resistance breast cancer therapy. J Colloid Interface Sci 2020; 565:254-269. [PMID: 31978788 DOI: 10.1016/j.jcis.2020.01.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/11/2020] [Accepted: 01/11/2020] [Indexed: 11/25/2022]
Abstract
Multidrug resistance (MDR) is one of the major obstacles to clinical cancer chemotherapy. Herein, we designed new pH-sensitive pluronic micelles with the synergistic effects of oxidative therapy and MDR reversal. Pluronic (P123) was modified with α-tocopheryl succinate (α-TOS) via an acid-labile ortho ester (OE) linkage to give a pH-sensitive copolymer (POT). Self-assembled POT micelles exhibited desirable size (~80 nm), excellent anti-dilution ability, high drug loading (~85%), acid-triggered degradation and drug release behaviours. In vitro cell experiments verified that POT micelles could significantly reverse MDR through suppressing the function of drug effluxs mediated by P123 and induce more reactive oxygen species (ROS) generation mediated by α-TOS, resulting in enhanced cytotoxicity and apoptosis in MDR cells. In vivo studies further revealed that DOX-loaded POT micelles (POT-DOX) possessed the highest drug accumulation (3.03% ID/g at 24 h) and the strongest tumour growth inhibition (TGI 83.48%). Pathological analysis also indicated that POT-DOX could induce more apoptosis or necrosis at the site of tumour without distinct damage to normal tissues. Overall, these smart POT micelles have great potential as promising nano-carriers for MDR reversal and cancer treatment.
Collapse
Affiliation(s)
- Xu Cheng
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Xiaoli Zeng
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Yan Zheng
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Qin Fang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Jun Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui Province 230601, PR China.
| |
Collapse
|
26
|
Kaneko S, Yamazaki T, Kohno K, Sato A, Kato K, Yano T. Combination Effect of Bowman-Birk Inhibitor and α-Tocopheryl Succinate on Prostate Cancer Stem-Like Cells. J Nutr Sci Vitaminol (Tokyo) 2020; 65:272-277. [PMID: 31257268 DOI: 10.3177/jnsv.65.272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The reoccurrence of androgen-dependent prostate cancer after anti-androgen therapy mainly depends on prostate cancer stem-like cells. To reduce the risk, it is important to delete the cancer stem-like cells. Furthermore, to induce differentiation of cancer stem-like cells is critical to abrogate stemness of the cells. Therefore, we tried to investigate a possibility on the establishment of a new effective therapy to eradicate the cancer stem-like cells via the induction of differentiation in this study. Prostate cancer stem-like cells from an androgen-dependent prostate cancer cell line (LNCaP cell) had severe resistance against an anti-androgen therapeutic agent. We selected Bowman-Birk inhibitor (BBI) from soybeans reported as a chemopreventive agent in prostate cancer to differentiate the caner stem-like cells and α-tocopheryl succinate (TOS) known as a mitocan to induce effectively cytotoxic effect against the cancer stem-like cells. In fact, only TOS treatment had cytotoxic effect against the cancer stem-like cells, but the addition of BBI treatment to the cells treated with TOS reinforced TOS-mediated cytotoxicity in the cancer stem-like cells. This reinforcement coincided with the combination-enhanced apoptosis in the stem-like cells. Also, we confirmed caspase9-caspase3 cascade mainly contributed to the enhancement of the cytotoxicity in the stem-like cells caused by the combination, indicating that the reinforcement of BBI on TOS-mediated apoptosis via mitochondria related to the enhancing cytotoxic effect of the combination on the prostate cancer stem-like cells. Overall, it seems that the combination is an effective new approach to reduce the reoccurrence of prostate cancer targeting prostate cancer stem cells.
Collapse
Affiliation(s)
- Saki Kaneko
- Graduate School of Food and Nutritional Sciences, Toyo University
| | | | - Kakeru Kohno
- Graduate School of Food and Nutritional Sciences, Toyo University.,Research Institute of Life Innovation, Toyo University
| | - Ayami Sato
- Research Institute of Life Innovation, Toyo University
| | - Kazunori Kato
- Graduate School of Science and Engineerin, Toyo University
| | - Tomohiro Yano
- Graduate School of Food and Nutritional Sciences, Toyo University.,Research Institute of Life Innovation, Toyo University
| |
Collapse
|
27
|
Czupiel PP, Delplace V, Shoichet MS. Cationic block amphiphiles show anti-mitochondrial activity in multi-drug resistant breast cancer cells. J Control Release 2019; 305:210-219. [PMID: 31071370 DOI: 10.1016/j.jconrel.2019.04.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022]
Abstract
Currently, there are limited treatment options for multi-drug resistant breast cancer. Lipid-modified cationic peptides have the potential to reach the mitochondria, which are attractive targets for the treatment of multi-drug resistant (MDR) breast cancer; yet, little is known about their mitochondrial targeting and anti-cancer activity. Interestingly, lipid-modified cationic peptides, typically used as gene transfection agents, exhibit similar structural features to mitochondrial targeted peptides. Using octahistidine-octaarginine (H8R8) as a model cationic peptide for cell penetration and endosomal escape, we explored the anti-cancer potential of lipid-modified cationic peptides as a function of amphiphilicity, biodegradability and lipid structure. We found that cationic peptides modified with a lipid that is at least 12 carbons in length exhibit potent anti-cancer activity in the low micromolar range in both EMT6/P and EMT6/AR-1 breast cancer cells. Comparing degradable and non-degradable linkers, as well as L- and D-amino acid sequences, we found that the anti-cancer activity is mostly independent of the biodegradation of the lipid-modified cationic peptides. Two candidates, stearyl-H8R8 (Str-H8R8) and vitamin E succinate-H8R8 (VES-H8R8) were cytotoxic to cancer cells by mitochondria depolarization. We observed increased reactive oxygen species (ROS) production, reduced cell bioenergetics and drug efflux, triggering apoptosis and G1 cell cycle arrest. Compared to Str-H8R8, VES-H8R8 showed enhanced cancer cell selectivity and drug efflux inhibition, thereby serving as a potential novel therapeutic agent. This study deepens our understanding of lipid-modified cationic peptides and uncovers their potential in multi-drug resistant breast cancer.
Collapse
Affiliation(s)
- Petro P Czupiel
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Vianney Delplace
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada; Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada.
| |
Collapse
|
28
|
Wałejko P, Dobrzycki Ł, Ratkiewicz A, Socha P, Witkowski S, Cyrański MK. An X-ray and Natural Bond Orbital (NBO) structural study of α-tocopheryl and 2,2,5,7,8-pentamethylchroman-6-yl succinates. JOURNAL OF SAUDI CHEMICAL SOCIETY 2019. [DOI: 10.1016/j.jscs.2018.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
29
|
Du X, Yin S, Zhou F, Du X, Xu J, Gu X, Wang G, Li J. Reduction-sensitive mixed micelles for selective intracellular drug delivery to tumor cells and reversal of multidrug resistance. Int J Pharm 2018; 550:1-13. [DOI: 10.1016/j.ijpharm.2018.08.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/31/2018] [Accepted: 08/12/2018] [Indexed: 12/17/2022]
|
30
|
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]
|
31
|
Han S, Sun R, Su H, Lv J, Xu H, Zhang D, Fu Y. Delivery of docetaxel using pH-sensitive liposomes based on D-α-tocopheryl poly(2-ethyl-2-oxazoline) succinate: Comparison with PEGylated liposomes. Asian J Pharm Sci 2018; 14:391-404. [PMID: 32104468 PMCID: PMC7032253 DOI: 10.1016/j.ajps.2018.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/21/2018] [Accepted: 07/25/2018] [Indexed: 12/22/2022] Open
Abstract
This study aimed to investigate the ability of the novel materials D-α-tocopheryl poly(2-ethyl-2-oxazoline) succinate (TPOS) to construct pH-sensitive liposomes. TPOS was initially synthesized and characterized by TLC, FTIR, and 1H-NMR. The buffering capacity of polyethylene glycol- distearoyl phosphatidylethanolamine (PEG-DSPE) and TPOS was determined by acid-base titration, and TPOS displayed a slower downtrend and gentler slope of titration curve than PEG-DSPE within pH 7.4–5.0. Studies on the in vitro drug release demonstrated that TPOS modified docetaxel (DOC) liposomes (TPOS-DOC-L) had a slower drug-release rate at pH 7.4 similar to PEGylated-DOC liposomes (PEG-DOC-L), whereas the release rate reached approximately 86.92% ± 1.69% at pH 6.4. In vitro cellular uptake assays by microplate reader, and flow cytometry revealed that TPOS modified coumarin 6 liposomes (TPOS-C6-L) had stronger cellular uptake at pH 6.4 than that at pH 7.4 (P < 0.01). Conversely, for PEGylated C6 liposomes (PEG-C6-L) and conventional C6 liposomes (C6-L), very similar cellular uptakes were exhibited at different pH values. Confocal laser scanning microscopy images showed that PEG-C6-L and C6-L were mainly located in lysosomes. By contrast, TPOS-C6-L showed broader cytoplasmic release and distribution at 4 h. MTT assay showed that the cytotoxicity of TPOS-DOC-L was similar to that of PEG-DOC-L and conventional DOC liposomes (DOC-L) at the same DOC concentration and at pH 7.4, but was much lower than those at pH 6.4 after 48 h of incubation. The apoptosis of PEG-DOC-L and DOC-L had no remarkable improvement with decreased pH from 7.4 to 6.4. Meanwhile, TPOS-DOC-L significantly induced the apoptosis of HeLa cells with decreased pH. Therefore, TPOS can be a biomaterial for the construction of a pH-sensitive drug delivery system.
Collapse
Affiliation(s)
- Shu Han
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Ruiyang Sun
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Hong Su
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Jing Lv
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Huan Xu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Di Zhang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Yuanshan Fu
- Department of Anatomy, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
32
|
Hepatocellular carcinomas are promoted by tocopheryl acetate but eliminated by tocopheryl succinate. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
33
|
Battogtokh G, Cho YY, Lee JY, Lee HS, Kang HC. Mitochondrial-Targeting Anticancer Agent Conjugates and Nanocarrier Systems for Cancer Treatment. Front Pharmacol 2018; 9:922. [PMID: 30174604 PMCID: PMC6107715 DOI: 10.3389/fphar.2018.00922] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Abstract
The mitochondrion is an important intracellular organelle for drug targeting due to its key roles and functions in cellular proliferation and death. In the last few decades, several studies have revealed mitochondrial functions, attracting the focus of many researchers to work in this field over nuclear targeting. Mitochondrial targeting was initiated in 1995 with a triphenylphosphonium-thiobutyl conjugate as an antioxidant agent. The major driving force for mitochondrial targeting in cancer cells is the higher mitochondrial membrane potential compared with that of the cytosol, which allows some molecules to selectively target mitochondria. In this review, we discuss mitochondria-targeting ligand-conjugated anticancer agents and their in vitro and in vivo behaviors. In addition, we describe a mitochondria-targeting nanocarrier system for anticancer drug delivery. As previously reported, several agents have been known to have mitochondrial targeting potential; however, they are not sufficient for direct application for cancer therapy. Thus, many studies have focused on direct conjugation of targeting ligands to therapeutic agents to improve their efficacy. There are many variables for optimal mitochondria-targeted agent development, such as choosing a correct targeting ligand and linker. However, using the nanocarrier system could solve some issues related to solubility and selectivity. Thus, this review focuses on mitochondria-targeting drug conjugates and mitochondria-targeted nanocarrier systems for anticancer agent delivery.
Collapse
Affiliation(s)
| | | | | | | | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| |
Collapse
|
34
|
Martín-Saldaña S, Palao-Suay R, Aguilar MR, García-Fernández L, Arévalo H, Trinidad A, Ramírez-Camacho R, San Román J. pH-sensitive polymeric nanoparticles with antioxidant and anti-inflammatory properties against cisplatin-induced hearing loss. J Control Release 2017; 270:53-64. [PMID: 29197586 DOI: 10.1016/j.jconrel.2017.11.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 11/28/2022]
Abstract
Polymeric nanoparticles (NPs) based on smart synthetic amphiphilic copolymers are used to transport and controlled release dexamethasone in the inner ear to protect against the ototoxic effect of cisplatin. The NPs were based on a mixture of two pseudo-block polymer drugs obtained by free radical polymerization: poly(VI-co-HEI) and poly(VP-co-MVE) or poly(VP-co-MTOS), being VI 1-vinylimidazole, VP N-vinylpyrrolidone, and HEI, MVE and MTOS the methacrylic derivatives of ibuprofen, α-tocopherol and α-tocopheryl succinate, respectively. The NPs were obtained by nanoprecipitation with appropriate hydrodynamic properties, and isoelectric points that matched the pH of inflamed tissue. The NPs were tested both in vitro (using HEI-OC1 cells) and in vivo (using a murine model) with good results. Although the concentration of dexamethasone administered in the NPs is around two orders of magnitude lower that the conventional treatment for intratympanic administration, the NPs protected from the cytotoxic effect of cisplatin when the combination of the appropriate properties in terms of size, zeta potential, encapsulation efficiency and isoelectric point were achieved. To the best of our knowledge this is the first time that pH sensitive NPs are used to protect from cisplatin-induced hearing loss by intratympanic administration.
Collapse
Affiliation(s)
- Sergio Martín-Saldaña
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Ear Research Group, Hospital UniversitarioPuerta de Hierro Majadahonda, Health Research Institute Puerta de Hierro, Madrid, Spain
| | - Raquel Palao-Suay
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| | - María Rosa Aguilar
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain.
| | - Luis García-Fernández
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| | - Humberto Arévalo
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Almudena Trinidad
- Ear Research Group, Hospital UniversitarioPuerta de Hierro Majadahonda, Health Research Institute Puerta de Hierro, Madrid, Spain
| | - Rafael Ramírez-Camacho
- Ear Research Group, Hospital UniversitarioPuerta de Hierro Majadahonda, Health Research Institute Puerta de Hierro, Madrid, Spain
| | - Julio San Román
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| |
Collapse
|
35
|
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
|
36
|
Palao-Suay R, Aguilar MR, Parra-Ruiz FJ, Martín-Saldaña S, Rohner NA, Thomas SN, San Román J. Multifunctional decoration of alpha-tocopheryl succinate-based NP for cancer treatment: effect of TPP and LTVSPWY peptide. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:152. [PMID: 28861765 DOI: 10.1007/s10856-017-5963-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/16/2017] [Indexed: 06/07/2023]
Abstract
Active targeting not only of a specific cell but also a specific organelle maximizes the therapeutic activity minimizing adverse side effects in healthy tissues. The present work describes the synthesis, characterization, and in vitro biological activity of active targeting nanoparticles (NP) for cancer therapy based on α-tocopheryl succinate (α-TOS), a well-known mitocan, that selectively induces apoptosis of cancer cells and proliferalting endothelial cells. Human epidermal growth factor receptor 2 (HER2) targeting peptide LTVSPWY (PEP) and triphenylphosphonium lipophilic cation (TPP) were conjugated to a previously optimized RAFT block copolymer that formed self-assembled NP of appropriate size for this application and low polydispersity by self-organized precipitation method. PEP and TPP were included in order to target not only HER2 positive cancer cells, but also the mitochondria of these cancer cells, respectively. The in vitro experiments demonstrated the faster incorporation of the active-targeting NP and the higher accumulation of TPP-bearing NP in the mitochondria of MDA-MB-453 HER2 positive cancer cells compared to non-decorated NP. Moreover, the encapsulation of additional α-TOS in the hydrophobic core of the NP was achieved with high efficiencies. The loaded NP presented higher cytotoxicity than unloaded NP but preserved their selectivity against cancer cells in a range of tested concentrations.
Collapse
Affiliation(s)
- Raquel Palao-Suay
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, ICTP-CSIC, Juan de la Cierva, 3, 28006, Madrid, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| | - María Rosa Aguilar
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, ICTP-CSIC, Juan de la Cierva, 3, 28006, Madrid, Spain.
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain.
| | - Francisco J Parra-Ruiz
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, ICTP-CSIC, Juan de la Cierva, 3, 28006, Madrid, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| | - Sergio Martín-Saldaña
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, ICTP-CSIC, Juan de la Cierva, 3, 28006, Madrid, Spain
| | - Nathan A Rohner
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, 30332, GA, USA
| | - Susan N Thomas
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, 30332, GA, USA
| | - Julio San Román
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, ICTP-CSIC, Juan de la Cierva, 3, 28006, Madrid, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| |
Collapse
|
37
|
Kilicay E, Karahaliloglu Z, Alpaslan P, Hazer B, Denkbas EB. In vitro evaluation of antisense oligonucleotide functionalized core-shell nanoparticles loaded with α-tocopherol succinate. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:1762-1785. [PMID: 28696185 DOI: 10.1080/09205063.2017.1354670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Antisense oligonucleotide (ASO)-conjugated-α-tocopherol succinate (TCS)-loaded-poly(lactic acid)-g-poly(ethylene glycol) nanoparticles (ASO-TCS-PLA-PEG NPs), with the ratio of polymer/TCS of 10:2.5, 10:5, 10:7 (w/w) were prepared for targeting cancer therapy. The amphiphilic PLA, amino terminated PEG graft copolymers were synthesized by ring opening polymerization reaction. Nanoparticles were produced by using double emulsion (w/o/w) solvent evaporation method. ASO-TCS-PLA-PEG NPs demonstrated satisfactory encapsulation and loading efficiency and size distribution. The short-term stability studies were carried out at 4 and 25 °C for 30 days to assess their mean particle size, polydispersity index and zeta potential. The cellular uptake and extended cytoplasmic retention of the NPs in A549 human lung carcinoma and L929 mouse fibroblast cells were examined by fluorescence and confocal microscopy. In human lung cancer cells, ASO-TCS-PLA-PEG NPs exhibited better cellular internalization, cytotoxicity and apoptotic and necrotic effects compared to healthy cell line, L929. These findings showed that ASO-modified nanoparticles could serve as a promising nanocarrier for targeted tumor cells.
Collapse
Affiliation(s)
- Ebru Kilicay
- a Zonguldak Vocational High School, Bülent Ecevit University , Zonguldak , Turkey
| | - Zeynep Karahaliloglu
- b Faculty of Science, Biology Department , Aksaray University , Aksaray , Turkey
| | - Pınar Alpaslan
- c Department of Biomedical Engineering , TOBB University of Economics and Technology , Ankara , Turkey
| | - Baki Hazer
- d Physical Chemistry Division, Chemistry Department , Bülent Ecevit University , Zonguldak , Turkey
| | - Emir Baki Denkbas
- e Biochemistry Division, Department of Chemistry , Hacettepe University , Ankara , Turkey
| |
Collapse
|
38
|
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
|
39
|
Tuguntaev RG, Chen S, Eltahan AS, Mozhi A, Jin S, Zhang J, Li C, Wang PC, Liang XJ. P-gp Inhibition and Mitochondrial Impairment by Dual-Functional Nanostructure Based on Vitamin E Derivatives To Overcome Multidrug Resistance. ACS APPLIED MATERIALS & INTERFACES 2017; 9:16900-16912. [PMID: 28463476 PMCID: PMC5545886 DOI: 10.1021/acsami.7b03877] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Vitamin E derivatives possess many essential features for drug-delivery applications, such as biocompatibility, stability, improvement of water solubility of hydrophobic compounds, anticancer activity, and the ability to overcome multidrug resistance (MDR). Herein, vitamin E derivatives are used to overcome MDR through a combined P-glycoprotein (P-gp) inhibition and mitochondrial impairment strategy. A novel nanomicellar drug-delivery system as a carrier for doxorubicin (DOX) was developed, in which d-α-tocopheryl polyethylene glycol 1000 succinate was used as a P-gp inhibitor, α-tocopheryl succinate was introduced as a mitochondrial disrupting agent, and d-α-tocopheryl polyethylene glycol 2000 succinate was used as the main building block of micelles. The optimal ratio between the components of the nanocarrier was determined. The resultant DOX-loaded mixed micelles exhibited a suitable size of 52.08 nm, high drug-loading encapsulation efficiency (>98%), high stability, and pH-dependent drug release. In vitro experiments demonstrated a significantly increased cytotoxic activity of DOX-loaded mixed micelles against resistant MCF-7/Adr cells (45-fold higher than DOX after 48 h of treatment). In vivo studies revealed superior antitumor efficiency with less cardio- and hepatotoxicities of DOX-loaded micelles compared with that of free DOX. These results highlight that the developed DOX-loaded mixed micelles have a promising potential to overcome MDR in chemotherapy for clinical usage.
Collapse
Affiliation(s)
- Ruslan G. Tuguntaev
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Shizhu Chen
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
| | - Ahmed Shaker Eltahan
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Anbu Mozhi
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Shubin Jin
- Beijing Municipal Institute of Labour Protection, No. 55 Taoranting Road, Xicheng District, Beijing 100054, P. R. China
| | - Jinchao Zhang
- College of Chemistry & Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, P. R. China
| | - Chan Li
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Corresponding Authors: . Phone: +86-010-82545569. Fax: +86-010-62656765 (C.L.). (X.-J.L.)
| | - Paul C. Wang
- Fu Jen Catholic University, Taipei, 24205, Taiwan
- Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington, D.C. 20060, United States
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- Laboratory of Controllable Nanopharmaceuticals, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Corresponding Authors: . Phone: +86-010-82545569. Fax: +86-010-62656765 (C.L.). (X.-J.L.)
| |
Collapse
|
40
|
Hou Y, Yao C, Ling L, Du Y, He R, Ismail M, Zhang Y, Fu Z, Li X. Novel dual VES phospholipid self-assembled liposomes with an extremely high drug loading efficiency. Colloids Surf B Biointerfaces 2017; 156:29-37. [PMID: 28499202 DOI: 10.1016/j.colsurfb.2017.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 03/30/2017] [Accepted: 05/01/2017] [Indexed: 01/16/2023]
Abstract
Vitamin E succinate (VES), a unique selective anti-cancer drug, has attracted much attention for its ability to induce apoptosis in various cancer cells. Importantly, it has been reported that VES is largely non-toxic to normal cells. However, poor aqueous solubility and bioavailability extensively restricted its clinical utility. In this report, dual VES phospholipid conjugate (di-VES-GPC) prodrug based liposomes were prepared in order to develop an efficient delivery system for VES. Di-VES-GPC was first synthesized by conjugating VES with l-α-glycerophosphorylcholine (GPC) using N,N'-dicyclohexylcarbodiimide (DCC) as a coupling agent. The di-VES-GPC prodrug was able to self-assemble into liposomes by reverse-phase evaporation method. The structure of the liposomes was characterized by dynamic light scattering (DLS), transmission electron microscopy (TEM) and cryo-TEM. The results showed that di-VES-GPC assembled liposomes were spherical with an average diameter approximately 183nm. Cryo-TEM data confirmed the formation of multilamellar liposomes with the bilayer thickness about 5nm by the assembly of the conjugate without any excipient. The VES drug loading highly reaches up to 82.8wt% in the liposomes after a simple calculation. Furthermore, the in vitro release behavior of di-VES-GPC liposomes was evaluated in different media. It was found that the liposomes could release free VES at a weakly acidic microenvironment but exhibited good stability under a simulated biological condition. The cellular uptake and intracellular drug release tests demonstrated that di-VES-GPC liposomes could be internalized effectively and converted into parent drug VES in cancer cells. Furthermore, in vitro antitumor activities of the di-VES-GPC liposomes were evaluated by MTT assay and flow cytometry. It was revealed that the liposomes presented comparable cytotoxicities to free VES. Taken together, the di-VES-GPC liposomes might provide an excellent formulation of VES which have potential in the treatment of cancers.
Collapse
Affiliation(s)
- Yongpeng Hou
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Chen Yao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| | - Longbing Ling
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Ruiyu He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Muhammad Ismail
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Ying Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Zhenglin Fu
- National Center for Protein Science, Shanghai 200000, China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| |
Collapse
|
41
|
Savitskaya MA, Onischenko GE. α-Tocopheryl Succinate Affects Malignant Cell Viability, Proliferation, and Differentiation. BIOCHEMISTRY (MOSCOW) 2017; 81:806-18. [PMID: 27677550 DOI: 10.1134/s0006297916080034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The widespread occurrence of malignant tumors motivates great attention to finding and investigating effective new antitumor preparations. Such preparations include compounds of the vitamin E family. Among them, α-tocopheryl succinate (vitamin E succinate (VES)) has the most pronounced antitumor properties. In this review, various targets and mechanisms of the antitumor effect of vitamin E succinate are characterized. It has been shown that VES has multiple intracellular targets and effects, and as a result VES is able to induce apoptosis in tumor cells, inhibit their proliferation, induce differentiation, prevent metastasizing, and inhibit angiogenesis. However, VES has minimal effects on normal cells and tissues. Due to the variety of targets and selectivity of action, VES is a promising agent against malignant neoplasms. More detailed studies in this area can contribute to development of effective and safe chemotherapeutic preparations.
Collapse
Affiliation(s)
- M A Savitskaya
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia.
| | | |
Collapse
|
42
|
Rohlenova K, Sachaphibulkij K, Stursa J, Bezawork-Geleta A, Blecha J, Endaya B, Werner L, Cerny J, Zobalova R, Goodwin J, Spacek T, Alizadeh Pesdar E, Yan B, Nguyen MN, Vondrusova M, Sobol M, Jezek P, Hozak P, Truksa J, Rohlena J, Dong LF, Neuzil J. Selective Disruption of Respiratory Supercomplexes as a New Strategy to Suppress Her2 high Breast Cancer. Antioxid Redox Signal 2017; 26:84-103. [PMID: 27392540 PMCID: PMC5206771 DOI: 10.1089/ars.2016.6677] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS Expression of the HER2 oncogene in breast cancer is associated with resistance to treatment, and Her2 may regulate bioenergetics. Therefore, we investigated whether disruption of the electron transport chain (ETC) is a viable strategy to eliminate Her2high disease. RESULTS We demonstrate that Her2high cells and tumors have increased assembly of respiratory supercomplexes (SCs) and increased complex I-driven respiration in vitro and in vivo. They are also highly sensitive to MitoTam, a novel mitochondrial-targeted derivative of tamoxifen. Unlike tamoxifen, MitoTam efficiently suppresses experimental Her2high tumors without systemic toxicity. Mechanistically, MitoTam inhibits complex I-driven respiration and disrupts respiratory SCs in Her2high background in vitro and in vivo, leading to elevated reactive oxygen species production and cell death. Intriguingly, higher sensitivity of Her2high cells to MitoTam is dependent on the mitochondrial fraction of Her2. INNOVATION Oncogenes such as HER2 can restructure ETC, creating a previously unrecognized therapeutic vulnerability exploitable by SC-disrupting agents such as MitoTam. CONCLUSION We propose that the ETC is a suitable therapeutic target in Her2high disease. Antioxid. Redox Signal. 26, 84-103.
Collapse
Affiliation(s)
- Katerina Rohlenova
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic
| | | | - Jan Stursa
- 2 School of Medical Science, Griffith University , Southport, Australia .,3 Prague Institute of Chemical Technology , Prague, Czech Republic .,4 Biomedical Research Center, University Hospital , Hradec Kralove, Czech Republic
| | | | - Jan Blecha
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic
| | - Berwini Endaya
- 2 School of Medical Science, Griffith University , Southport, Australia
| | - Lukas Werner
- 4 Biomedical Research Center, University Hospital , Hradec Kralove, Czech Republic
| | - Jiri Cerny
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic
| | - Renata Zobalova
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic .,2 School of Medical Science, Griffith University , Southport, Australia
| | - Jacob Goodwin
- 2 School of Medical Science, Griffith University , Southport, Australia
| | - Tomas Spacek
- 5 Institute of Physiology , Prague, Czech Republic
| | | | - Bing Yan
- 2 School of Medical Science, Griffith University , Southport, Australia
| | - Maria Nga Nguyen
- 2 School of Medical Science, Griffith University , Southport, Australia
| | - Magdalena Vondrusova
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic
| | - Margaryta Sobol
- 6 Institute of Molecular Genetics , Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Jezek
- 5 Institute of Physiology , Prague, Czech Republic
| | - Pavel Hozak
- 6 Institute of Molecular Genetics , Czech Academy of Sciences, Prague, Czech Republic
| | - Jaroslav Truksa
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic
| | - Jakub Rohlena
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic
| | - Lan-Feng Dong
- 2 School of Medical Science, Griffith University , Southport, Australia
| | - Jiri Neuzil
- 1 Institute of Biotechnology , Czech Academy of Sciences, BIOCEV, Vestec, Prague-West, Czech Republic .,2 School of Medical Science, Griffith University , Southport, Australia
| |
Collapse
|
43
|
Qu Q, Ma X, Zhao Y. Anticancer Effect of α-Tocopheryl Succinate Delivered by Mitochondria-Targeted Mesoporous Silica Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2016; 8:34261-34269. [PMID: 27998109 DOI: 10.1021/acsami.6b13974] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondria targeted mesoporous silica nanoparticles (MSNPs) having an average diameter of 68 nm were fabricated and then loaded with hydrophobic anticancer agent α-tocopheryl succinate (α-TOS). The property of targeting mitochondria was achieved by the surface functionalization of triphenylphosphonium (TPP) on MSNPs, since TPP is an effective mitochondria-targeting ligand. Intracellular uptake and mitochondria targeting of fabricated MSNPs were evaluated in HeLa and HepG2 cancerous cell lines as well as HEK293 normal cell line. In addition, various biological assays were conducted with the aim to investigate the effectiveness of α-TOS delivered by the functional MSNPs, including studies of cytotoxicity, mitochondria membrane potential, intracellular adenosine triphosphate (ATP) production, and apoptosis. On the basis of these experiments, high anticancer efficiency of α-TOS delivered by mitochondria targeted MSNPs was demonstrated, indicating a promising application potential of MSNP-based platform in mitochondria targeted delivery of anticancer agents.
Collapse
Affiliation(s)
- Qiuyu Qu
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, 637371 Singapore
| | - Xing Ma
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, 637371 Singapore
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University , 21 Nanyang Link, 637371 Singapore
- School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
44
|
Palao-Suay R, Aguilar MR, Parra-Ruiz FJ, Maji S, Hoogenboom R, Rohner NA, Thomas SN, Román JS. Enhanced Bioactivity of α-Tocopheryl Succinate Based Block Copolymer Nanoparticles by Reduced Hydrophobicity. Macromol Biosci 2016; 16:1824-1837. [PMID: 27739627 PMCID: PMC5518931 DOI: 10.1002/mabi.201600259] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/04/2016] [Indexed: 12/25/2022]
Abstract
Well-structured amphiphilic copolymers are necessary to obtain self-assembled nanoparticles (NPs) based on synthetic polymers. Highly homogeneous and monodispersed macromolecules obtained by controlled polymerization have successfully been used for this purpose. However, disaggregation of the organized macromolecules is desired when a bioactive element, such as α-tocopheryl succinate, is introduced in self-assembled NPs and this element must be exposed or released to exert its action. The aim of this work is to demonstrate that the bioactivity of synthetic NPs based on defined reversible addition-fragmentation chain transfer polymerization copolymers can be enhanced by the introduction of hydrophilic comonomers in the hydrophobic segment. The amphiphilic terpolymers are based on poly(ethylene glycol) (PEG) as hydrophilic block, and a hydrophobic block based on a methacrylic derivative of α-tocopheryl succinate (MTOS) and small amounts of 2-hydroxyethyl methacrylate (HEMA) (PEG-b-poly(MTOS-co-HEMA)). The introduction of HEMA reduces hydrophobicity and introduces "disorder" both in the homogeneous blocks and the compact core of the corresponding NPs. These NPs are able to encapsulate additional α-tocopheryl succinate (α-TOS) with high efficiency and their biological activity is much higher than that described for the unmodified copolymers, proposedly due to more efficient degradation and release of α-TOS, demonstrating the importance of the hydrophilic-hydrophobic balance.
Collapse
Affiliation(s)
- Raquel Palao-Suay
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006, Madrid, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - María Rosa Aguilar
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006, Madrid, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Francisco J Parra-Ruiz
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006, Madrid, Spain
| | - Samarendra Maji
- Supramolecular Chemistry Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, 9000, Ghent, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281-S4, 9000, Ghent, Belgium
| | - Nathan A Rohner
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, 30332, GA, USA
| | - Susan N Thomas
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, 30332, GA, USA
| | - Julio San Román
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, Juan de la Cierva 3, 28006, Madrid, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Av. Monforte de Lemos 3-5, 28029, Madrid, Spain
| |
Collapse
|
45
|
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
|
46
|
Niki E. Oxidative stress and antioxidants: Distress or eustress? Arch Biochem Biophys 2016; 595:19-24. [PMID: 27095209 DOI: 10.1016/j.abb.2015.11.017] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 10/14/2015] [Accepted: 10/14/2015] [Indexed: 01/06/2023]
Abstract
There is a growing consensus that reactive oxygen species (ROS) are not just associated with various pathologies, but that they act as physiological redox signaling messenger with important regulatory functions. It is sometimes stated that "if ROS is a physiological signaling messenger, then removal of ROS by antioxidants such as vitamins E and C may not be good for human health." However, it should be noted that ROS acting as physiological signaling messenger and ROS removed by antioxidants are not the same. The lipid peroxidation products of polyunsaturated fatty acids and cholesterol induce adaptive response and enhance defense capacity against subsequent oxidative insults, but it is unlikely that these lipid peroxidation products are physiological signaling messenger produced on purpose. The removal of ROS and inhibition of lipid peroxidation by antioxidants should be beneficial for human health, although it has to be noted also that they may not be an effective inhibitor of oxidative damage mediated by non-radical oxidants. The term ROS is vague and, as there are many ROS and antioxidants which are different in chemistry, it is imperative to explicitly specify ROS and antioxidant to understand the effects and role of oxidative stress and antioxidants properly.
Collapse
Affiliation(s)
- Etsuo Niki
- Health Research Institute, National Institute of Advanced Industrial Science & Technology, Takamatsu 761-0395, Japan; Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| |
Collapse
|
47
|
Muddineti OS, Ghosh B, Biswas S. Current trends in the use of vitamin E-based micellar nanocarriers for anticancer drug delivery. Expert Opin Drug Deliv 2016; 14:715-726. [DOI: 10.1080/17425247.2016.1229300] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Omkara Swami Muddineti
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Telangana, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Telangana, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Telangana, India
| |
Collapse
|
48
|
Yu LY, Su GM, Chen CK, Chiang YT, Lo CL. Specific Cancer Cytosolic Drug Delivery Triggered by Reactive Oxygen Species-Responsive Micelles. Biomacromolecules 2016; 17:3040-7. [PMID: 27536957 DOI: 10.1021/acs.biomac.6b00916] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cytosolic drug delivery, a major route in cancer therapy, is limited by the lack of efficient and safe endosomal escape techniques. Herein, we demonstrate a reactive oxygen species (ROS)-responsive micelle composed of methoxy polyethylene glycol-b-poly(diethyl sulfide) (mPEG-PS) copolymers which can induce specific endosome escape in cancer cells by changes in the hydrophobicity of copolymers. Owing to the more ROS levels in cancer cells than normal cells, the copolymers can be converted into more hydrophilic and insert into and destabilize the cancer intracellular endosome membrane after cellular uptake. More importantly, we show that acid-intolerant drugs successfully maintain their bioactivity and cause selective cytotoxicity for cancer cells over normal cells. Our results suggest that the endosomal escape induced by hydrophobic-hydrophilic exchange of copolymers has great potential to locally and efficiently deliver biological agents (e.g., proteins and genes) in the cancer cell cytosol.
Collapse
Affiliation(s)
| | | | | | - Yi-Ting Chiang
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan, Republic of China
| | | |
Collapse
|
49
|
Hou L, Zhang H, Xu P, Zhang L, Zhang X, Sun Y, Huang X, Wu K. Effect of vitamin E succinate on the expression of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor in gastric cancer cells and CD4(+) T cells. MOLECULAR BIOSYSTEMS 2016; 11:3119-28. [PMID: 26378383 DOI: 10.1039/c5mb00350d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gastric malignancy, which shows poor prognosis, is one of the most frequent causes of cancer-associated deaths. Vitamin E succinate (VES) inhibits cell proliferation and induces apoptosis in a concentration- and time-dependent manner. We explored the effect of VES on the expression of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor in gastric cancer cells and CD4(+) T cells. On one hand, VES dose-dependently regulated the expression of the TRAIL receptor in gastric cancer cells. Moreover, the activation of the TRAIL receptor, death receptor 4 (DR4), and death receptor 5 (DR5) in gastric cancer cells increased for up to 12 h. On the other hand, the expression of TRAIL protein in human CD4(+) T cells was obviously upregulated in the presence of VES. On the basis of these findings, we combined VES and human CD4(+) T cells to induce apoptosis of MKN28 human gastric cancer cells. The results showed that VES induced higher gastric cancer cell apoptosis when combined with human CD4(+) T cells than when applied alone. We conclude that VES can induce the expression of TRAIL receptor in gastric cancer cells, as well as the expression of TRAIL in CD4(+) T cells. Overall, our results provide a theoretical basis for future immunotherapy studies.
Collapse
Affiliation(s)
- Liying Hou
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, 150081 Harbin, China.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Rizza S, Montagna C, Cardaci S, Maiani E, Di Giacomo G, Sanchez-Quiles V, Blagoev B, Rasola A, De Zio D, Stamler JS, Cecconi F, Filomeni G. S-nitrosylation of the Mitochondrial Chaperone TRAP1 Sensitizes Hepatocellular Carcinoma Cells to Inhibitors of Succinate Dehydrogenase. Cancer Res 2016; 76:4170-82. [PMID: 27216192 DOI: 10.1158/0008-5472.can-15-2637] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 04/20/2016] [Indexed: 11/16/2022]
Abstract
S-nitrosoglutathione reductase (GSNOR) represents the best-documented denitrosylase implicated in regulating the levels of proteins posttranslationally modified by nitric oxide on cysteine residues by S-nitrosylation. GSNOR controls a diverse array of physiologic functions, including cellular growth and differentiation, inflammation, and metabolism. Chromosomal deletion of GSNOR results in pathologic protein S-nitrosylation that is implicated in human hepatocellular carcinoma (HCC). Here we identify a metabolic hallmark of aberrant S-nitrosylation in HCC and exploit it for therapeutic gain. We find that hepatocyte GSNOR deficiency is characterized by mitochondrial alteration and by marked increases in succinate dehydrogenase (SDH) levels and activity. We find that this depends on the selective S-nitrosylation of Cys(501) in the mitochondrial chaperone TRAP1, which mediates its degradation. As a result, GSNOR-deficient cells and tumors are highly sensitive to SDH inhibition, namely to α-tocopheryl succinate, an SDH-targeting molecule that induced RIP1/PARP1-mediated necroptosis and inhibited tumor growth. Our work provides a specific molecular signature of aberrant S-nitrosylation in HCC, a novel molecular target in SDH, and a first-in-class therapy to treat the disease. Cancer Res; 76(14); 4170-82. ©2016 AACR.
Collapse
Affiliation(s)
- Salvatore Rizza
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Costanza Montagna
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Simone Cardaci
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Emiliano Maiani
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Virginia Sanchez-Quiles
- Department of Biochemistry and Molecular Biology and the Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology and the Villum Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Andrea Rasola
- CNR Institute of Neuroscience and Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Daniela De Zio
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine, Case Western Reserve University and Harrington Discovery Institute, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Francesco Cecconi
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark. Department of Biology, University of Rome Tor Vergata, Rome, Italy. IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppe Filomeni
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark. Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|