1
|
Li M, Fang G, Zahid F, Saleem R, Ishrat G, Ali Z, Naeem M, Din FU. Co-delivery of paclitaxel and curcumin loaded solid lipid nanoparticles for improved targeting of lung cancer: In vitro and in vivo investigation. Heliyon 2024; 10:e30290. [PMID: 38720725 PMCID: PMC11076978 DOI: 10.1016/j.heliyon.2024.e30290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
The objective of this study was to develop nanotechnology-mediated paclitaxel (PAC) and curcumin (CUR) co-loaded solid lipid nanoparticles (PAC-CUR-SLNs) for the treatment of lung cancer, which is a leading cause of death worldwide. Around 85 % cases of lungs cancer constitute non-small cell lung cancer (NSCLC). PAC-CUR-SLNs were prepared via high pressure homogenization. The in vitro drug release of PAC-CUR-SLNs was checked followed by their in vitro cytotoxic investigation using adenocarcinomic human alveolar basal epithelial cells (A549) cell lines. Anticancer effects along with side effects of the synergistic delivery of PAC-CUR-SLNs were studied in vivo, using BALB/c mice. PAC-CUR-SLNs were nano sized (190 nm), homogeneously disseminated particles with %IE of both PAC and CUR above 94 %. PAC-CUR-SLNs released PAC and CUR in a controlled fashion when compared with free drug suspensions. The cytotoxicity of PAC-CUR-SLNs was higher than individual drug-loaded SLNs and pure drugs. Moreover, the co-delivery displayed synergistic effect, indicating potential of PAC-CUR-SLNs in lung cancer treatment. In vivo tumor investigation of PAC-CUR-SLNs exhibited 12-fold reduced tumor volume and almost no change in body weight of BALB/c mice, when compared with the experimental groups including control group. The inhibition of tumor rate on day 28 was 82.7 % in the PAC-CUR-SLNs group, which was significantly higher than the pure drugs and monotherapies. It can be concluded that, encapsulating the co-loaded antitumor drugs like PAC-CUR in SLNs may help in improved targeting of the tumor with enhanced anticancer effect.
Collapse
Affiliation(s)
- Mao Li
- Guangxi Higher Education Key Laboratory for the Research of Du-related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Gang Fang
- Guangxi Higher Education Key Laboratory for the Research of Du-related Diseases in Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Fatima Zahid
- Department of Pharmacy Quaid-i-Azam University, 45320, Islamabad, Pakistan
- Nanomedicine Research Group, Department of Pharmacy Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Raheela Saleem
- College of Pharmacy, Liaquat University of Medical and Health Sciences Jamshoro, Pakistan
| | - Ghazala Ishrat
- Department of Pharmaceutics, Faculty of Pharmacy, Salim Habib University, Karachi, Pakistan
| | - Zakir Ali
- Department of Pharmacy Quaid-i-Azam University, 45320, Islamabad, Pakistan
- Nanomedicine Research Group, Department of Pharmacy Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Muhammad Naeem
- National University of Medical Sciences, Rawalpindi, Pakistan
| | - Fakhar ud Din
- Department of Pharmacy Quaid-i-Azam University, 45320, Islamabad, Pakistan
- Nanomedicine Research Group, Department of Pharmacy Quaid-i-Azam University, 45320, Islamabad, Pakistan
| |
Collapse
|
2
|
Pham DD, Pham TH, Bui TH, Britikova EV, Britikov VV, Bocharov EV, Usanov SA, Phan VC, Le TBT. In vitro and in vivo anti-tumor effect of Trichobakin fused with urokinase-type plasminogen activator ATF-TBK. Mol Biol Rep 2024; 51:130. [PMID: 38236367 DOI: 10.1007/s11033-023-09036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Trichobakin (TBK), a member of type I ribosome-inactivating proteins (RIPs), was first successfully cloned from Trichosanthes sp Bac Kan 8-98 in Vietnam. Previous study has shown that TBK acts as a potential protein synthesis inhibitor; however, the inhibition efficiency and specificity of TBK on cancer cells remain to be fully elucidated. METHODS AND RESULTS In this work, we employed TBK and TBK conjugated with a part of the amino-terminal fragment (ATF) of the urokinase-type plasminogen activator (uPA), which contains the Ω-loop that primarily interacts with urokinase-type plasminogen activator receptor, and can be a powerful carrier in the drug delivery to cancer cells. Four different human tumor cell lines and BALB/c mice bearing Lewis lung carcinoma cells (LLC) were used to evaluate the role of TBK and ATF-TBK in the inhibition of tumor growth. Here we showed that the obtained ligand fused RIP (ATF-TBK) reduced the growth of four human cancer cell lines in vitro in the uPA receptor level-dependent manner, including the breast adenocarcinoma MDA-MB 231 cells and MCF7 cells, the prostate carcinoma LNCaP cells and the hepatocellular carcinoma HepG2 cells. Furthermore, the conjugate showed anti-tumor activity and prolonged the survival time of tumor-bearing mice. The ATF-TBK also did not cause the death of mice with doses up to 48 mg/kg, and they were not significantly distinct on parameters of hematology and serum biochemistry between the control and experiment groups. CONCLUSIONS In conclusion, ATF-TBK reduced the growth of four different human tumor cell lines and inhibited lung tumor growth in a mouse model with little side effects. Hence, the ATF-TBK may be a target to consider as an anti-cancer agent for clinical trials.
Collapse
Affiliation(s)
- Dan Duc Pham
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Thi Hue Pham
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Thi Huyen Bui
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Elena V Britikova
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141, Minsk, Belarus
| | - Vladimir V Britikov
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141, Minsk, Belarus
| | - Eduard V Bocharov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia, 117997
| | - Sergey A Usanov
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, 220141, Minsk, Belarus
| | - Van Chi Phan
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam
| | - Thi Bich Thao Le
- Institute of Biotechnology (IBT), Vietnam Academy of Science and Technology (VAST), 18, Hoang Quoc Viet Road, Cau Giay, Hanoi, Vietnam.
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam.
| |
Collapse
|
3
|
Jia W, Yuan J, Li S, Cheng B. The role of dysregulated mRNA translation machinery in cancer pathogenesis and therapeutic value of ribosome-inactivating proteins. Biochim Biophys Acta Rev Cancer 2023; 1878:189018. [PMID: 37944831 DOI: 10.1016/j.bbcan.2023.189018] [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: 09/14/2023] [Revised: 10/17/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
Dysregulated protein synthesis is a hallmark of tumors. mRNA translation reprogramming contributes to tumorigenesis, which is fueled by abnormalities in ribosome formation, tRNA abundance and modification, and translation factors. Not only malignant cells but also stromal cells within tumor microenvironment can undergo transformation toward tumorigenic phenotypes during translational reprogramming. Ribosome-inactivating proteins (RIPs) have garnered interests for their ability to selectively inhibit protein synthesis and suppress tumor growth. This review summarizes the role of dysregulated translation machinery in tumor development and explores the potential of RIPs in cancer treatment.
Collapse
Affiliation(s)
- Wentao Jia
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
4
|
Sharma A, Gupta S, Sharma NR, Paul K. Expanding role of ribosome-inactivating proteins: From toxins to therapeutics. IUBMB Life 2023; 75:82-96. [PMID: 36121739 DOI: 10.1002/iub.2675] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/26/2022] [Indexed: 02/02/2023]
Abstract
Ribosome-inactivating proteins (RIPs) are toxic proteins with N-glycosidase activity. RIPs exert their action by removing a specific purine from 28S rRNA, thereby, irreversibly inhibiting the process of protein synthesis. RIPs can target both prokaryotic and eukaryotic cells. In bacteria, the production of RIPs aid in the process of pathogenesis whereas, in plants, the production of these toxins has been attributed to bolster defense against insects, viral, bacterial and fungal pathogens. In recent years, RIPs have been engineered to target a particular cell type, this has fueled various experiments testing the potential role of RIPs in many biomedical applications like anti-viral and anti-tumor therapies in animals as well as anti-pest agents in engineered plants. In this review, we present a comprehensive study of various RIPs, their mode of action, their significance in various fields involving plants and animals. Their potential as treatment options for plant infections and animal diseases is also discussed.
Collapse
Affiliation(s)
- Anuj Sharma
- Department of Biochemistry, DAV University, Jalandhar, Punjab, India
| | - Shelly Gupta
- Department of Biochemistry, School of Biosciences and Bioengineering, Lovely Professional University, Phagwara, Punjab, India
| | - Neeta Raj Sharma
- School of Biosciences and Bioengineering, Lovely Professional University, Phagwara, Punjab, India
| | - Karan Paul
- Department of Biochemistry, DAV University, Jalandhar, Punjab, India
| |
Collapse
|
5
|
Ahmadi Moghaddam Y, Maroufi A, Zareei S, Irani M. Computational design of fusion proteins against ErbB2-amplified tumors inspired by ricin toxin. Front Mol Biosci 2023; 10:1098365. [PMID: 36936983 PMCID: PMC10018397 DOI: 10.3389/fmolb.2023.1098365] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Although the anti-cancer activity of ricin is well-known, its non-specific targeting challenges the development of ricin-derived medicines. In the present study, novel potential ribosome-inactivating fusion proteins (RIPs) were computationally engineered by incorporation of an ErbB2-dependant penetrating peptide (KCCYSL, MARAKE, WYSWLL, MARSGL, MSRTMS, and WYAWML), a linker (either EAAAK or GGGGS) and chain A of ricin which is responsible for the ribosome inactivation. Molecular dynamics simulations assisted in making sure that the least change is made in conformation and dynamic behavior of ricin chain A in selected chimeric protein (CP). Moreover, the potential affinity of the selected CPs against the ligand-uptaking ErbB2 domain was explored by molecular docking. The results showed that two CPs (CP2 and 10) could bind the receptor with the greatest affinity.
Collapse
Affiliation(s)
- Yasser Ahmadi Moghaddam
- Department of Plant Production and Genetics, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran
- *Correspondence: Yasser Ahmadi Moghaddam,
| | - Asad Maroufi
- Department of Plant Production and Genetics, Faculty of Agriculture, University of Kurdistan, Sanandaj, Iran
| | - Sara Zareei
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mehdi Irani
- Department of Chemistry, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| |
Collapse
|
6
|
The Updated Review on Plant Peptides and Their Applications in Human Health. Int J Pept Res Ther 2022; 28:135. [PMID: 35911180 PMCID: PMC9326430 DOI: 10.1007/s10989-022-10437-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Biologically active plant peptides, consisting of secondary metabolites, are compounds (amino acids) utilized by plants in their defense arsenal. Enzymatic processes and metabolic pathways secrete these plant peptides. They are also known for their medicinal value and have been incorporated in therapeutics of major human diseases. Nevertheless, its limitations (low bioavailability, high cytotoxicity, poor absorption, low abundance, improper metabolism, etc.) have demanded a need to explore further and discover other new plant compounds that overcome these limitations. Keeping this in mind, therapeutic plant proteins can be excellent remedial substitutes for bodily affliction. A multitude of these peptides demonstrates anti-carcinogenic, anti-microbial, anti-HIV, and neuro-regulating properties. This article's main aim is to list out and report the status of various therapeutic plant peptides and their prospective status as peptide-based drugs for multiple diseases (infectious and non-infectious). The feasibility of these compounds in the imminent future has also been discussed.
Collapse
|
7
|
Advances on Delivery of Cytotoxic Enzymes as Anticancer Agents. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123836. [PMID: 35744957 PMCID: PMC9230553 DOI: 10.3390/molecules27123836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022]
Abstract
Cancer is one of the most serious human diseases, causing millions of deaths worldwide annually, and, therefore, it is one of the most investigated research disciplines. Developing efficient anticancer tools includes studying the effects of different natural enzymes of plant and microbial origin on tumor cells. The development of various smart delivery systems based on enzyme drugs has been conducted for more than two decades. Some of these delivery systems have been developed to the point that they have reached clinical stages, and a few have even found application in selected cancer treatments. Various biological, chemical, and physical approaches have been utilized to enhance their efficiencies by improving their delivery and targeting. In this paper, we review advanced delivery systems for enzyme drugs for use in cancer therapy. Their structure-based functions, mechanisms of action, fused forms with other peptides in terms of targeting and penetration, and other main results from in vivo and clinical studies of these advanced delivery systems are highlighted.
Collapse
|
8
|
Comparative Proteomic Analysis of Drug Trichosanthin Addition to BeWo Cell Line. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27051603. [PMID: 35268705 PMCID: PMC8911981 DOI: 10.3390/molecules27051603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022]
Abstract
Trichosanthin (TCS) is a traditional Chinese herbal medicine used to treat some gynecological diseases. Its effective component has diverse biological functions, including antineoplastic activity. The human trophoblast cell line BeWo was chosen as an experimental model for in vitro testing of a drug screen for anticancer properties of TCS. The MTT method was used in this study to get a primary screen result. The result showed that 100 mM had the best IC50 value. Proteomics analysis was then performed for further investigation of the drug effect of TCS on the BeWo cell line. In this differential proteomic expression analysis, the total proteins extracted from the BeWo cell line and their protein expression level after the drug treatment were compared by 2DE. Then, 24 unique three-fold differentially expressed proteins (DEPs) were successfully identified by MALDI-TOF/TOF MS. Label-free proteomics was run as a complemental method for the same experimental procedure. There are two proteins that were identified in both the 2DE and label-free methods. Among those identified proteins, bioinformatics analysis showed the importance of pathway and signal transduction and gives us the potential possibility for the disease treatment hypothesis.
Collapse
|
9
|
Ran G, Feng XL, Xie YL, Zheng QY, Guo PP, Yang M, Feng YL, Ling C, Zhu LQ, Zhong C. The use of miR122 and its target sequence in adeno-associated virus-mediated trichosanthin gene therapy. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2021; 19:515-525. [PMID: 34538767 DOI: 10.1016/j.joim.2021.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Plant-derived cytotoxic transgene expression, such as trichosanthin (tcs), regulated by recombinant adeno-associated virus (rAAV) vector is a promising cancer gene therapy. However, the cytotoxic transgene can hamper the vector production in the rAAV producer cell line, human embryonic kidney (HEK293) cells. Here, we explored microRNA-122 (miR122) and its target sequence to limit the expression of the cytotoxic gene in the rAAV producer cells. METHODS A miR122 target (122T) sequence was incorporated into the 3' untranslated region of the tcs cDNA sequence. The firefly luciferase (fluc) transgene was used as an appropriate control. Cell line HEK293-mir122 was generated by the lentiviral vector-mediated genome integration of the mir122 gene in parental HEK293 cells. The effects of miR122 overexpression on cell growth, transgene expression, and rAAV production were determined. RESULTS The presence of 122T sequence significantly reduced transgene expression in the miR122-enriched Huh7 cell line (in vitro), fresh human hepatocytes (ex vivo), and mouse liver (in vivo). Also, the normal liver physiology was unaffected by delivery of 122T sequence by rAAV vectors. Compared with the parental cells, the miR122-overexpressing HEK293-mir122 cell line showed similar cell growth rate and expression of transgene without 122T, as well as the ability to produce liver-targeting rAAV vectors. Fascinatingly, the yield of rAAV vectors carrying the tcs-122T gene was increased by 77.7-fold in HEK293-mir122 cells. Moreover, the tcs-122T-containing rAAV vectors significantly reduced the proliferation of hepatocellular carcinoma cells without affecting the normal liver cells. CONCLUSION HEK293-mir122 cells along with the 122T sequence provide a potential tool to attenuate the cytotoxic transgene expression, such as tcs, during rAAV vector production.
Collapse
Affiliation(s)
- Gai Ran
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA
| | - Xi-Lin Feng
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yi-Lin Xie
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Qing-Yun Zheng
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Peng-Peng Guo
- Department of Traditional Chinese Medicine, Chinese People's Liberation Army 971 Hospital, Qingdao 266071, Shandong Province, China
| | - Ming Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Ying-Lu Feng
- Department of Traditional Chinese Medicine, Chinese People's Liberation Army 971 Hospital, Qingdao 266071, Shandong Province, China
| | - Chen Ling
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA
| | - Li-Qing Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Chen Zhong
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China.
| |
Collapse
|
10
|
Setayesh-Mehr Z, Poorsargol M. Toxic proteins application in cancer therapy. Mol Biol Rep 2021; 48:3827-3840. [PMID: 33895972 DOI: 10.1007/s11033-021-06363-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/16/2021] [Indexed: 12/19/2022]
Abstract
Ribosome inactivating proteins (RIPs) as family of anti-cancer drugs recently received much attention due to their interesting anti-cancer mechanism. In spite of small drugs, RIPs use the large-size effect (LSE) to prevent the efflux process governed by drug resistance transporters (DRTs) which prevents inside of the cells against drug transfection. There are many clinical translation obstacles that severely restrict their applications especially their delivery approach to the tumor cells. As the main goal of this review, we will focus on trichosanthin (TCS) and gelonin (Gel) and other types, especially scorpion venom-derived RIPs to clarify that they are struggling with what types of bio-barriers and these challenges could be solved in cancer therapy science. Then, we will try to highlight recent state-of-the-arts in delivery of RIPs for cancer therapy.
Collapse
Affiliation(s)
- Zahra Setayesh-Mehr
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran.
| | - Mahdiye Poorsargol
- Department of Chemistry, Faculty of Sciences, University of Zabol, Zabol, Iran
| |
Collapse
|
11
|
Zhu Z, Ying Z, Zeng M, Zhang Q, Liao G, Liang Y, Li C, Zhang C, Wang X, Jiang W, Luan P, Sha O. Trichosanthin cooperates with Granzyme B to restrain tumor formation in tongue squamous cell carcinoma. BMC Complement Med Ther 2021; 21:88. [PMID: 33750370 PMCID: PMC7944607 DOI: 10.1186/s12906-021-03266-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 02/28/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Tongue squamous cell carcinoma (TSCC) is a common type of oral cancer, with a relatively poor prognosis and low post-treatment survival rate. Various strategies and novel drugs to treat TSCC are emerging and under investigation. Trichosanthin (TCS), extracted from the root tubers of Tian-Hua-Fen, has been found to have multiple biological and pharmacological functions, including inhibiting the growth of cancer cells. Granzyme B (GrzB) is a common toxic protein secreted by natural killer cells and cytotoxic T cells. Our group has reported that TCS combined with GrzB might be a superior approach to inhibit liver tumor progression, but data relating to the use of this combination to treat TSCC remain limited. The aim of this study was to examine the effectiveness of TCS on TSCC processes and underlying mechanisms. METHODS First, we screened the potential antitumor activity of TCS using two types of SCC cell lines. Subsequently, a subcutaneous squamous cell carcinoma xenograft model in nude mice was established. These model mice were randomly divided into four groups and treated as follows: control group, TCS treatment group, GrzB treatment group, and TCS/GrzB combination treatment group. Various tumorigenesis parameters, such as Ki67, PCNA, caspase-3, Bcl-2 and VEGFA, et al., were performed to determine the effects of these treatments on tumor development. RESULTS Screening confirmed that the SCC25 line exhibited greater sensitivity than the SCC15 line to TCS in vitro studies. TCS or GrzB treatment significantly inhibited tumor growth compared with the inhibition seen in the control group. The TCS/GrzB combination inhibited tumor growth more than either drug alone. TCS treatment inhibited tumor proliferation by downregulating Ki67 and Bcl2 protein expression while accelerating tumor apoptosis. In the TCS/GrzB-treated group, expression of Ki67 was further downregulated, while the level of activated caspase-3 was increased, compared with their expression in either of the single drug treatment groups. CONCLUSION These results suggest that the TCS/GrzB combination could represent an effective immunotherapy for TSCC.
Collapse
Affiliation(s)
- Zeyao Zhu
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhenguang Ying
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Meiqi Zeng
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
| | - Qiang Zhang
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
- The Shenzhen Stomatology Hospital, Shenzhen, China
| | - Guiqing Liao
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Yunliu Liang
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Chunman Li
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Chengfei Zhang
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
- Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - Xia Wang
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Weipeng Jiang
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
| | - Ping Luan
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China.
| | - Ou Sha
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China.
- School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China.
| |
Collapse
|
12
|
Wani SS, Dar PA, Zargar SM, Dar TA. Therapeutic Potential of Medicinal Plant Proteins: Present Status and Future Perspectives. Curr Protein Pept Sci 2021; 21:443-487. [PMID: 31746291 DOI: 10.2174/1389203720666191119095624] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/10/2019] [Accepted: 10/27/2019] [Indexed: 02/07/2023]
Abstract
Biologically active molecules obtained from plant sources, mostly including secondary metabolites, have been considered to be of immense value with respect to the treatment of various human diseases. However, some inevitable limitations associated with these secondary metabolites like high cytotoxicity, low bioavailability, poor absorption, low abundance, improper metabolism, etc., have forced the scientific community to explore medicinal plants for alternate biologically active molecules. In this context, therapeutically active proteins/peptides from medicinal plants have been promoted as a promising therapeutic intervention for various human diseases. A large number of proteins isolated from the medicinal plants have been shown to exhibit anti-microbial, anti-oxidant, anti-HIV, anticancerous, ribosome-inactivating and neuro-modulatory activities. Moreover, with advanced technological developments in the medicinal plant research, medicinal plant proteins such as Bowman-Birk protease inhibitor and Mistletoe Lectin-I are presently under clinical trials against prostate cancer, oral carcinomas and malignant melanoma. Despite these developments and proteins being potential drug candidates, to date, not a single systematic review article has documented the therapeutical potential of the available biologically active medicinal plant proteome. The present article was therefore designed to describe the current status of the therapeutically active medicinal plant proteins/peptides vis-à-vis their potential as future protein-based drugs for various human diseases. Future insights in this direction have also been highlighted.
Collapse
Affiliation(s)
- Snober Shabeer Wani
- Department of Clinical Biochemistry, University of Kashmir, Srinagar-190006, Jammu and Kashmir, India
| | - Parvaiz A Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar-190006, Jammu and Kashmir, India
| | - Sajad M Zargar
- Division of Plant Biotechnology, S. K. University of Agricultural Sciences and Technology of Srinagar, Shalimar-190025, Srinagar, Jammu and Kashmir, India
| | - Tanveer A Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar-190006, Jammu and Kashmir, India
| |
Collapse
|
13
|
Yu G, Ali Z, Sajjad Khan A, Ullah K, Jamshaid H, Zeb A, Imran M, Sarwar S, Choi HG, Ud Din F. Preparation, Pharmacokinetics, and Antitumor Potential of Miltefosine-Loaded Nanostructured Lipid Carriers. Int J Nanomedicine 2021; 16:3255-3273. [PMID: 34012260 PMCID: PMC8127833 DOI: 10.2147/ijn.s299443] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/07/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The purpose of this study was to investigate the suitability of nanostructured lipid carriers (NLCs) loaded with miltefosine (HePC) as an anticancer drug for the treatment of breast cancer. METHODS HePC-NLCs were prepared using a microemulsion technique and then evaluated for particle size, polydispersity index (PDI), incorporation efficiency, in vitro release of entrapped drug, and hemolytic potential. Furthermore, pharmacokinetic, biodistribution, and liver toxicity analyses were performed in Sprague-Dawley rats, and antitumor efficacy was evaluated in Michigan Cancer Foundation-7 (MCF-7) and squamous cell carcinoma-7 (SCC-7) cells in vitro and in tumour-bearing BALB/c mice in vivo. Advanced analyses including survival rate, immunohistopathology, and terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assays were performed to evaluate apoptosis in vivo. RESULTS The average particle size of the HePC-NLCs was 143 ± 16 nm, with a narrow PDI (0.104 ± 0.002), and the incorporation efficiency was found to be 91 ± 7%. The NLCs released HePC in a sustained manner, and this release was significantly lower than that of free drug. The in vitro hemolytic assay demonstrated a significantly reduced hemolytic potential (~9%) of the NLCs compared to that of the test formulations. The HePC-NLCs demonstrated enhanced pharmacokinetic behaviour over free drug, including extended blood circulation and an abridged clearance rate in rats. Furthermore, the HePC-NLCs exhibited higher cytotoxicity than the free drug in MCF-7 and SCC-7 cells. Moreover, the HePC-NLCs showed significantly enhanced (P < 0.005) antitumor activity compared to that of the control and free drug-treated mouse groups. Tumour cell apoptosis was also confirmed, indicating the antitumor potential of the HePC-NLCs. CONCLUSION These findings demonstrate the ability of NLCs as a drug delivery system for enhanced pharmacokinetic, antitumor, and apoptotic effects, most importantly when loaded with HePC.
Collapse
Affiliation(s)
- Guo Yu
- Department of Head and Neck Breast, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang City, Henan Province, 453000, People's Republic of China
| | - Zakir Ali
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Anam Sajjad Khan
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Kalim Ullah
- Department of Zoology, Kohat University of Science & Technology, Kohat, Khyber Pakhtunkhwa, Pakistan
| | - Humzah Jamshaid
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Alam Zeb
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Muhammad Imran
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Sadia Sarwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Han-Gon Choi
- College of Pharmacy & Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| | - Fakhar Ud Din
- Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| |
Collapse
|
14
|
Hassanin I, Elzoghby A. Albumin-based nanoparticles: a promising strategy to overcome cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:930-946. [PMID: 35582218 PMCID: PMC8992568 DOI: 10.20517/cdr.2020.68] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/28/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022]
Abstract
Circumvention of cancer drug resistance is one of the major investigations in nanomedicine. In this regard, nanotechnology-based drug delivery has offered various implications. However, protein-based nanocarriers have been a versatile choice compared to other nanomaterials, provided by their favorable characteristics and safety profiles. Specifically, albumin-based nanoparticles have been demonstrated to be an effective drug delivery system, owing to the inherent targeting modalities of albumin, through gp60- and SPARC-mediated receptor endocytosis. Furthermore, surface functionalization was exploited for active targeting, due to albumin’s abundance of carboxylic and amino groups. Stimuli-responsive drug release has also been pertained to albumin nano-systems. Therefore, albumin-based nanocarriers could potentially overcome cancer drug resistance through bypassing drug efflux, enhancing drug uptake, and improving tumor accumulation. Moreover, albumin nanocarriers improve the stability of various therapeutic cargos, for instance, nucleic acids, which allows their systemic administration. This review highlights the recent applications of albumin nanoparticles to overcome cancer drug resistance, the nano-fabrication techniques, as well as future perspectives and challenges.
Collapse
Affiliation(s)
- Islam Hassanin
- Department of Biotechnology, Institute of Graduate studies and Research, Alexandria University, Alexandria 21526, Egypt.,Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
15
|
Ajji PK, Binder MJ, Walder K, Puri M. Recombinant Balsamin induces apoptosis in liver and breast cancer cells via cell cycle arrest and regulation of apoptotic pathways. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.05.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
16
|
Britikov VV, Britikova EV, Urban AS, Lesovoy DM, Le TBT, Van Phan C, Usanov SA, Arseniev AS, Bocharov EV. Backbone and side-chain chemical shift assignments for the ribosome-inactivating protein trichobakin (TBK). BIOMOLECULAR NMR ASSIGNMENTS 2020; 14:55-61. [PMID: 31734904 DOI: 10.1007/s12104-019-09920-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/11/2019] [Indexed: 06/10/2023]
Abstract
Trichobakin (TBK) is a type-I ribosome-inactivating protein (RIP-I), acting as an extremely potent inhibitor of protein synthesis in the cell-free translation system of rabbit reticulocyte lysate (IC50: 3.5 pM). In this respect, TBK surpasses the well-studied highly homologous RIP-I trichosanthin (IC50: 20-27 pM), therefore creation of recombinant toxins based on it is of great interest. TBK needs to penetrate into cytosol through the cell membrane and specifically bind to α-sarcin/ricin loop of 28S ribosome RNA to perform the function of specific RNA depurination. At the moment, there is no detailed structural-dynamic information in solution about diverse states RIP-I can adopt at different stages on the way to protein synthesis inhibition. In this work, we report a near-complete assignment of 1H, 13C, and 15N TBK (27.3 kDa) resonances and analysis of the secondary structure based on the experimental chemical shifts data. This work will serve as a basis for further investigations of the structure, dynamics and interactions of the TBK with its molecular partners using NMR techniques.
Collapse
Affiliation(s)
- Vladimir V Britikov
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Minsk, Belarus.
| | - Elena V Britikova
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Minsk, Belarus
| | - Anatoly S Urban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology (MIPT), Dolgoprudny, Russia
| | - Dmitry M Lesovoy
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Thi Bich Thao Le
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Chi Van Phan
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Sergey A Usanov
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Minsk, Belarus
| | - Alexander S Arseniev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology (MIPT), Dolgoprudny, Russia
| | - Eduard V Bocharov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology (MIPT), Dolgoprudny, Russia
| |
Collapse
|
17
|
Lu JQ, Zhu ZN, Zheng YT, Shaw PC. Engineering of Ribosome-inactivating Proteins for Improving Pharmacological Properties. Toxins (Basel) 2020; 12:toxins12030167. [PMID: 32182799 PMCID: PMC7150887 DOI: 10.3390/toxins12030167] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/23/2022] Open
Abstract
Ribosome-inactivating proteins (RIPs) are N-glycosidases, which depurinate a specific adenine residue in the conserved α-sarcin/ricin loop (α-SRL) of rRNA. This loop is important for anchoring elongation factor (EF-G for prokaryote or eEF2 for eukaryote) in mRNA translocation. Translation is inhibited after the attack. RIPs therefore may have been applied for anti-cancer, and anti-virus and other therapeutic applications. The main obstacles of treatment with RIPs include short plasma half-life, non-selective cytotoxicity and antigenicity. This review focuses on the strategies used to improve the pharmacological properties of RIPs on human immunodeficiency virus (HIV) and cancers. Coupling with polyethylene glycol (PEG) increases plasma time and reduces antigenicity. RIPs conjugated with antibodies to form immunotoxins increase the selective toxicity to target cells. The prospects for future development on the engineering of RIPs for improving their pharmacological properties are also discussed.
Collapse
Affiliation(s)
- Jia-Qi Lu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 99077, China; (J.-Q.L.); (Z.-N.Z.)
| | - Zhen-Ning Zhu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 99077, China; (J.-Q.L.); (Z.-N.Z.)
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms, National Kunming High level Biosafety Research Center for Non-human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China;
| | - Pang-Chui Shaw
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong 99077, China; (J.-Q.L.); (Z.-N.Z.)
- Correspondence:
| |
Collapse
|
18
|
Multi-responsive albumin-lonidamine conjugated hybridized gold nanoparticle as a combined photothermal-chemotherapy for synergistic tumor ablation. Acta Biomater 2020; 101:531-543. [PMID: 31706039 DOI: 10.1016/j.actbio.2019.11.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/28/2019] [Accepted: 11/01/2019] [Indexed: 12/22/2022]
Abstract
Herein, we developed a multifunctional nanoplatform based on the nanoassembly of gold nanoparticles (GNP) conjugated with lonidamine (LND) and aptamer AS1411 (AS-LAGN) as an effective cancer treatment. Conjugating AS1411 aptamer on the surface of the nanoparticle significantly improved particle accumulation in cancer cells via specific affinity toward the nucleolin receptors. In vitro study clearly revealed that laser irradiation-based hyperthermia effect enhanced the chemotherapeutic effects of LND. Combinational treatment modalities revealed significant apoptosis with higher cell killing effect due to increased ROS production and inhibition of cell migration. GNP's ability to convert the excited state photon energy into thermal heat enabled synergistic photothermal/chemotherapy with improved therapeutic efficacy in animal models. Moreover, immunohistochemistry staining assays confirmed the ability of AS-LAGN to induce cellular apoptosis/necrosis and ablation in tumor tissues, without causing evident damages to the surrounding healthy tissues. Altogether, this AS-LAGN nanoplatform could be a promising strategy for mitochondria-based cancer treatment. STATEMENT OF SIGNIFICANCE: We have designed a facile biodegradable multifunctional nanocarrier system to target the mitochondria, the major "power house" of the cancer cells. We have constructed a multifunctional nanoassembly of protein coronated gold nanoparticles (GNP) conjugated with lonidamine (LND) and aptamer AS1411 (AS-LAGN) as an effective combination of phototherapy with chemotherapy for cancer treatment. The LND was conjugated with albumin which was in turn conjugated to GNP via redox-liable disulfide linkage to generate oxidative stress and ROS to kill cancer cells. GNP's ability to convert the excited state photon energy into thermal heat enabled synergistic photothermal/chemotherapy with improved therapeutic efficacy in animal models. Consistently, AS-LAGN showed enhanced antitumor efficacy in xenograft tumor model with remarkable tumor regression property.
Collapse
|
19
|
Asrorov AM, Gu Z, Min KA, Shin MC, Huang Y. Advances on Tumor-Targeting Delivery of Cytotoxic Proteins. ACS Pharmacol Transl Sci 2019; 3:107-118. [PMID: 32259092 DOI: 10.1021/acsptsci.9b00087] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Indexed: 12/11/2022]
Abstract
Great attention has been paid to cytotoxic proteins (e.g., ribosome-inactivating proteins, RIPs) possessing high anticancer activities; unlike small drugs, cytotoxic proteins can effectively retain inside the cells and avoid drug efflux mediated by multidrug resistance transporters due to the large-size effect. However, the clinical translation of these proteins is severely limited because of various biobarriers that hamper their effective delivery to tumor cells. Hence, in order to overcome these barriers, many smart drug delivery systems (DDS) have been developed. In this review, we will introduce two representative type I RIPs, trichosanthin (TCS) and gelonin (Gel), and overview the major biobarriers for protein-based cancer therapy. Finally, we outline advances on the development of smart DDS for effective delivery of these cytotoxic proteins for various applications in cancer treatment.
Collapse
Affiliation(s)
- Akmal M Asrorov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.,Institute of Bioorganic Chemistry, Academy of Sciences of Uzbekistan, 83, M. Ulughbek Street, Tashkent 100125, Uzbekistan
| | - Zeyun Gu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Kyoung Ah Min
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, 197 Injero, Gimhae, Gyeongnam 50834, Korea
| | - Meong Cheol Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, 501 Jinju Daero, Jinju, Gyeongnam 52828, Korea
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| |
Collapse
|
20
|
Ruiz-Torres V, Rodríguez-Pérez C, Herranz-López M, Martín-García B, Gómez-Caravaca AM, Arráez-Román D, Segura-Carretero A, Barrajón-Catalán E, Micol V. Marine Invertebrate Extracts Induce Colon Cancer Cell Death via ROS-Mediated DNA Oxidative Damage and Mitochondrial Impairment. Biomolecules 2019; 9:biom9120771. [PMID: 31771155 PMCID: PMC6995635 DOI: 10.3390/biom9120771] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 12/29/2022] Open
Abstract
Marine compounds are a potential source of new anticancer drugs. In this study, the antiproliferative effects of 20 invertebrate marine extracts on three colon cancer cell models (HGUE-C-1, HT-29, and SW-480) were evaluated. Extracts from two nudibranchs (Phyllidia varicosa, NA and Dolabella auricularia, NB), a holothurian (Pseudocol ochirus violaceus, PS), and a soft coral (Carotalcyon sp., CR) were selected due to their potent cytotoxic capacities. The four marine extracts exhibited strong antiproliferative effects and induced cell cycle arrest at the G2/M transition, which evolved into early apoptosis in the case of the CR, NA, and NB extracts and necrotic cell death in the case of the PS extract. All the extracts induced, to some extent, intracellular ROS accumulation, mitochondrial depolarization, caspase activation, and DNA damage. The compositions of the four extracts were fully characterized via HPLC-ESI-TOF-MS analysis, which identified up to 98 compounds. We propose that, among the most abundant compounds identified in each extract, diterpenes, steroids, and sesqui- and seterterpenes (CR); cembranolides (PS); diterpenes, polyketides, and indole terpenes (NA); and porphyrin, drimenyl cyclohexanone, and polar steroids (NB) might be candidates for the observed activity. We postulate that reactive oxygen species (ROS) accumulation is responsible for the subsequent DNA damage, mitochondrial depolarization, and cell cycle arrest, ultimately inducing cell death by either apoptosis or necrosis.
Collapse
Affiliation(s)
- Verónica Ruiz-Torres
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández (UMH), 03202 Elche, Spain; (V.R.-T.); (M.H.-L.); (V.M.)
| | - Celia Rodríguez-Pérez
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, 18071 Granada, Spain (D.A.-R.); (A.S.-C.)
- Research and Development of Functional Food Centre (CIDAF), PTS Granada, Edificio BioRegion, 18016 Granada, Spain
| | - María Herranz-López
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández (UMH), 03202 Elche, Spain; (V.R.-T.); (M.H.-L.); (V.M.)
| | - Beatriz Martín-García
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, 18071 Granada, Spain (D.A.-R.); (A.S.-C.)
- Research and Development of Functional Food Centre (CIDAF), PTS Granada, Edificio BioRegion, 18016 Granada, Spain
| | - Ana-María Gómez-Caravaca
- Research and Development of Functional Food Centre (CIDAF), PTS Granada, Edificio BioRegion, 18016 Granada, Spain
| | - David Arráez-Román
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, 18071 Granada, Spain (D.A.-R.); (A.S.-C.)
- Research and Development of Functional Food Centre (CIDAF), PTS Granada, Edificio BioRegion, 18016 Granada, Spain
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, 18071 Granada, Spain (D.A.-R.); (A.S.-C.)
- Research and Development of Functional Food Centre (CIDAF), PTS Granada, Edificio BioRegion, 18016 Granada, Spain
| | - Enrique Barrajón-Catalán
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández (UMH), 03202 Elche, Spain; (V.R.-T.); (M.H.-L.); (V.M.)
- Correspondence: ; Tel.: +34-965-222-586
| | - Vicente Micol
- Instituto de Biología Molecular y Celular (IBMC) and Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández (UMH), 03202 Elche, Spain; (V.R.-T.); (M.H.-L.); (V.M.)
- CIBER, Fisiopatología de la Obesidad y la Nutrición, CIBERobn, Instituto de Salud Carlos III., Palma de Mallorca 07122, Spain
| |
Collapse
|
21
|
Ramasamy T, Ruttala HB, Kaliraj K, Poudel K, Jin SG, Choi HG, Ku SK, Yong CS, Kim JO. Polypeptide Derivative of Metformin with the Combined Advantage of a Gene Carrier and Anticancer Activity. ACS Biomater Sci Eng 2019; 5:5159-5168. [DOI: 10.1021/acsbiomaterials.9b00982] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Thiruganesh Ramasamy
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
- Center for Ultrasound Molecular Imaging and Therapeutics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Hima Bindu Ruttala
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Kaliappan Kaliraj
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, P.R. China
| | - Kishwor Poudel
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 712-715, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| |
Collapse
|
22
|
Wong JH, Ng TB, Wang H, Cheung RCF, Ng CCW, Ye X, Yang J, Liu F, Ling C, Chan K, Ye X, Chan WY. Antifungal Proteins with Antiproliferative Activity on Cancer Cells and HIV-1 Enzyme Inhibitory Activity from Medicinal Plants and Medicinal Fungi. Curr Protein Pept Sci 2019; 20:265-276. [PMID: 29895244 DOI: 10.2174/1389203719666180613085704] [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: 01/01/2018] [Revised: 04/10/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Abstract
A variety of fungi, plants, and their different tissues are used in Traditional Chinese Medicine to improve health, and some of them are recommended for dietary therapy. Many of these plants and fungi contain antifungal proteins and peptides which suppress spore germination and hyphal growth in phytopathogenic fungi. The aim of this article is to review antifungal proteins produced by medicinal plants and fungi used in Chinese medicine which also possess anticancer and human immunodeficiency virus-1 (HIV-1) enzyme inhibitory activities.
Collapse
Affiliation(s)
- Jack Ho Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hexiang Wang
- State Key Laboratory for Agrobiotechnology and Department of Microbiology, China Agricultural University, Beijing, China
| | - Randy Chi Fai Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Charlene Cheuk Wing Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiuyun Ye
- National Engineering Laboratory for High-Efficiency Enzyme Expression and College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian, China
| | - Jie Yang
- National Engineering Laboratory for High-Efficiency Enzyme Expression and College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian, China
| | - Fang Liu
- Department of Microbiology, Nankai University, Tianjin, China
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, FL, United States
| | - Ki Chan
- Biomedical and Tissue Engineering Research Group, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, China
| | - Xiujuan Ye
- Key Laboratory of Plant Virology of Fujian Province, Institute of Plant Virology, and Key Laboratory of Biopesticide and Chemical Biology, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Wai Yee Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
23
|
Dai X, Jiang P, Ji Y, Zhu X, Sun X. Suppression of murine B-cell lymphoma growth by trichosanthin through anti-angiogenesis. Am J Transl Res 2019; 11:3567-3577. [PMID: 31312367 PMCID: PMC6614629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/03/2019] [Indexed: 06/10/2023]
Abstract
Studies have suggested trichosanthin (TCS) exerts antitumor activity mainly through direct cytotoxicity toward cancer cells and immune regulation. In this study, we conducted the proliferation and apoptosis assay on A20 cells and endothelial cells (ECs) with different concentrations of TCS and investigated the levels of gene expression linked to angiogenesis. Herein, a new mechanism that TCS inhibits murine B-cell lymphoma growth by anti-angiogenesis was reported. First, TCS inhibit tumor growth and prolonged survival significantly in vivo, and TCS depressed the formation of new blood vessels around the tumor in a dose-dependent manner. Further studies showed that the platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31)-positive endothelial cell numbers, and the serum levels of MMP-2 and MMP-9 were also lower in the study group than controls. However, TCS did neither change the ratio of T cells and NK cells in the spleen of treated mice nor affect the proliferation and apoptosis of A20 cells in vitro. Additionally, the newly formed blood vessels in chorioallantoic membranes treated with TCS were significantly reduced. Last, TCS may suppress the proliferation, induce apoptosis and decrease tube formation and migration of endothelial cells (ECs). And, the mRNA and protein levels of VEGF in ECs treated with TCS were lower than that in the control group. These findings confirm that inhibitory effect of TCS on A20 murine B-cell lymphoma growth is mediated via anti-angiogenesis, and which may be associated with the down-regulation of VEGF and MMPs expression. This is an indication that TCS may represent a natural anti-angiogenic drug for lymphoma therapy.
Collapse
Affiliation(s)
- Xingbin Dai
- Department of Hematology, Affiliated Hospital of Nanjing University of Chinese MedicineNanjing 210029, China
- First Clinical Medical College, Nanjing University of Chinese MedicineNanjing 210029, China
| | - Pengjun Jiang
- First Clinical Medical College, Nanjing University of Chinese MedicineNanjing 210029, China
| | - Yanhua Ji
- School of Life Science & Technology, China Pharmaceutical UniversityNanjing 210009, China
| | - Xuejun Zhu
- First Clinical Medical College, Nanjing University of Chinese MedicineNanjing 210029, China
| | - Xuemei Sun
- First Clinical Medical College, Nanjing University of Chinese MedicineNanjing 210029, China
| |
Collapse
|
24
|
Wang W, Tan B, Zhang J, Qiu R, Liu X, Liu D, Li X, Wang J. Human nasopharyngeal carcinoma can be radiosensitized by trichosanthin via inhibition of the PI3K pathway. Exp Ther Med 2018; 16:4181-4186. [PMID: 30402158 DOI: 10.3892/etm.2018.6719] [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: 04/21/2017] [Accepted: 03/16/2018] [Indexed: 11/05/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a prevalent tumor that affects the head and neck. Radiation therapy is typically used to treat NPC; however, poor prognoses and distant metastases are common due to radiation resistance. The antitumor activities of trichosanthin (TCS) have been reported in several types of tumors. The aim of the present study was to investigate whether TCS may serve as a potential radiosensitizer in the treatment of NPC tumors. In the present study, NPC cells were treated with radiation alone or together with TCS and radiosensitivity was compared. Clonogenic assay, flow cytometry and an animal study were performed to assess cell death in NPC. The clonogenic assay demonstrated that TCS had a significant radiosensitizing effect on NPC cells. Western blotting indicated that phosphorylated protein kinase B and signal-regulated kinase [phosphoinositide 3-kinase (PI3K) pathway] were downregulated, and that cleaved caspase-3 was upregulated by combined treatment with TCS and radiation. Furthermore, TCS potently radiosensitized NPC xenografts in vivo. In conclusion, TCS radiosensitized NPC in vitro and in vivo via downregulation of PI3K pathways and the upregulation of cleaved caspase-3.
Collapse
Affiliation(s)
- Wen Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Bo Tan
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Jingwei Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Rongliang Qiu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Xinju Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Dongmei Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Xue Li
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| | - Jianhua Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, P.R. China
| |
Collapse
|
25
|
Shi WW, Wong KB, Shaw PC. Structural and Functional Investigation and Pharmacological Mechanism of Trichosanthin, a Type 1 Ribosome-Inactivating Protein. Toxins (Basel) 2018; 10:toxins10080335. [PMID: 30127254 PMCID: PMC6115768 DOI: 10.3390/toxins10080335] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 01/11/2023] Open
Abstract
Trichosanthin (TCS) is an RNA N-glycosidase that depurinates adenine-4324 in the conserved α-sarcin/ricin loop (α-SRL) of rat 28 S ribosomal RNA (rRNA). TCS has only one chain, and is classified as type 1 ribosome-inactivating protein (RIP). Our structural studies revealed that TCS consists of two domains, with five conserved catalytic residues Tyr70, Tyr111, Glu160, Arg163 and Phe192 at the active cleft formed between them. We also found that the structural requirements of TCS to interact with the ribosomal stalk protein P2 C-terminal tail. The structural analyses suggest TCS attacks ribosomes by first binding to the C-terminal domain of ribosomal P protein. TCS exhibits a broad spectrum of biological and pharmacological activities including anti-tumor, anti-virus, and immune regulatory activities. This review summarizes an updated knowledge in the structural and functional studies and the mechanism of its multiple pharmacological effects.
Collapse
Affiliation(s)
- Wei-Wei Shi
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Sha Tin New Town, Hong Kong, China.
| | - Kam-Bo Wong
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Sha Tin New Town, Hong Kong, China.
| | - Pang-Chui Shaw
- Centre for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Sha Tin New Town, Hong Kong, China.
| |
Collapse
|
26
|
Li C, Zeng M, Chi H, Shen J, Ng TB, Jin G, Lu D, Fan X, Xiong B, Xiao Z, Sha O. Trichosanthin increases Granzyme B penetration into tumor cells by upregulation of CI-MPR on the cell surface. Oncotarget 2018; 8:26460-26470. [PMID: 28460437 PMCID: PMC5432272 DOI: 10.18632/oncotarget.15518] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/08/2017] [Indexed: 12/21/2022] Open
Abstract
Trichosanthin is a plant toxin belonging to the family of ribosome-inactivating proteins. It has various biological and pharmacological activities, including anti-tumor and immunoregulatory effects. In this study, we explored the potential medicinal applications of trichosanthin in cancer immunotherapy. We found that trichosanthin and cation-independent mannose-6-phosphate receptor competitively bind to the Golgi-localized, γ-ear containing and Arf-binding proteins. It in turn promotes the translocation of cation-independent mannose-6-phosphate receptor from the cytosol to the plasma membrane, which is a receptor of Granzyme B. The upregulation of this receptor on the tumor cell surface increased the cell permeability to Granzyme B, and the latter is one of the major factors of cytotoxic T lymphocyte-mediated tumor cell apoptosis. These results suggest a novel potential application of trichosanthin and shed light on its anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Chunman Li
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| | - Meiqi Zeng
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| | - Huju Chi
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Tzi-Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Guangyi Jin
- School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| | - Desheng Lu
- School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| | - Xinmin Fan
- School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| | - Bilian Xiong
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ou Sha
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, Guangdong, China
| |
Collapse
|
27
|
Khandaker M, Akter S, Imam MZ. Trichosanthes dioica Roxb.: A vegetable with diverse pharmacological properties. FOOD SCIENCE AND HUMAN WELLNESS 2018. [DOI: 10.1016/j.fshw.2017.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
28
|
Choi HS, Kim MK, Lee K, Lee KM, Choi YK, Shin YC, Cho SG, Ko SG. SH003 represses tumor angiogenesis by blocking VEGF binding to VEGFR2. Oncotarget 2018; 7:32969-79. [PMID: 27105528 PMCID: PMC5078067 DOI: 10.18632/oncotarget.8808] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/31/2016] [Indexed: 11/25/2022] Open
Abstract
Tumor angiogenesis is a key feature of cancer progression, because a tumor requires abundant oxygen and nutrition to grow. Here, we demonstrate that SH003, a mixed herbal extract containing Astragalus membranaceus (Am), Angelica gigas (Ag) and Trichosanthes Kirilowii Maximowicz (Tk), represses VEGF-induced tumor angiogenesis both in vitro and in vivo. SH003 inhibited VEGF-induced migration, invasion and tube formation in human umbilical vein endothelial cells (HUVEC) with no effect on the proliferation. SH003 reduced CD31-positive vessel numbers in tumor tissues and retarded tumor growth in our xenograft mouse tumor model, while SH003 did not affect pancreatic tumor cell viability. Consistently, SH003 inhibited VEGF-stimulated vascular permeability in ears and back skins. Moreover, SH003 inhibited VEGF-induced VEGFR2-dependent signaling by blocking VEGF binding to VEGFR2. Therefore, our data conclude that SH003 represses tumor angiogenesis by inhibiting VEGF-induced VEGFR2 activation, and suggest that SH003 may be useful for treating cancer.
Collapse
Affiliation(s)
- Hyeong Sim Choi
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Min Kyoung Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Kangwook Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Kang Min Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Youn Kyung Choi
- Jeju International Marine Science Center for Research and Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju, Korea
| | - Yong Cheol Shin
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Sung-Gook Cho
- Department of Biotechnology, Korea National University of Transportation, Jeungpyeong, Chungbuk, Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
29
|
You C, Sun Y, Zhang S, Tang G, Zhang N, Li C, Tian X, Ma S, Luo Y, Sun W, Wang F, Liu X, Xiao Y, Gong Y, Zhang J, Xie C. Trichosanthin enhances sensitivity of non-small cell lung cancer (NSCLC) TRAIL-resistance cells. Int J Biol Sci 2018; 14:217-227. [PMID: 29483839 PMCID: PMC5821042 DOI: 10.7150/ijbs.22811] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 01/31/2018] [Indexed: 01/27/2023] Open
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) has a specific antitumour activity against many malignant tumours. However, more than half of lung cancer cells are resistant to TRAIL-relevant drugs. Trichosanthin (TCS) is a traditional Chinese medicine with strong inhibitive effects on various malignancies. Nevertheless, its function on TRAIL resistance has not been revealed in non-small cell lung cancer (NSCLC). To examine the molecular mechanisms of TCS-induced TRAIL sensitivity, we administrated TCS to TRAIL-resistance NSCLC cells, and found that the combination treatment of TCS and TRAIL inhibited cancer cell proliferation and invasion, and induced cell apoptosis and S-phase arrest. This combined therapeutic method regulated the expression levels of extrinsic apoptosis-associated proteins Caspase 3/8 and PARP; intrinsic apoptosis-associated proteins BCL-2 and BAX; invasion-associated proteins E-cadherin, N-cadherin, Vimentin, ICAM-1, MMP-2 and MMP-9; and cell cycle-associated proteins P27, CCNE1 and CDK2. Up-expression and redistribution of death receptors (DRs) on the cell surface were also observed in combined treatment. In conclusion, our results indicated that TCS rendered NSCLC cells sensitivity to TRAIL via upregulating and redistributing DR4 and DR5, inducing apoptosis, and regulating invasion and cell cycle related proteins. Our results provided a potential therapeutic method to enhance TRAIL-sensitivity.
Collapse
Affiliation(s)
- Chengcheng You
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Pathology, China Three Gorges University Medical College, Yichang, China
| | - Yingming Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shiyu Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guiliang Tang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Nannan Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chunyang Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaoli Tian
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shijing Ma
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenjie Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Feng Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington DC, USA
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junhong Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumour Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
30
|
Song L, Xu X, Li Z. A serine protease extracted from Trichosanthes kirilowii inhibits epithelial-mesenchymal transition via antagonizing PKM2-mediated STAT3/Snail1 pathway in human colorectal adenocarcinoma cells. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
31
|
Ruttala HB, Chitrapriya N, Kaliraj K, Ramasamy T, Shin WH, Jeong JH, Kim JR, Ku SK, Choi HG, Yong CS, Kim JO. Facile construction of bioreducible crosslinked polypeptide micelles for enhanced cancer combination therapy. Acta Biomater 2017; 63:135-149. [PMID: 28890258 DOI: 10.1016/j.actbio.2017.09.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 08/14/2017] [Accepted: 09/02/2017] [Indexed: 12/27/2022]
Abstract
In this study, we developed pH and redox-responsive crosslinked polypeptide-based combination micelles for enhanced chemotherapeutic efficacy and minimized side effects. The stability and drug release properties of the polypeptide micelles were efficiency balanced by the corona-crosslinking of the triblock copolymer, poly(ethylene glycol)-b-poly(aspartic acid)-b-poly(tyrosine) (PEG-b-pAsp-b-pTyr) with coordinated redox and pH dual-sensitivity by introducing disulfide crosslinkages. Because of the crosslinking of the middle shell of the triblock polypeptide micelles, their robust structure was maintained in strong destabilization conditions and exhibited excellent stability. GSH concentrations were significantly higher in tumor tissue than in normal tissue, which formed the basis for our design. Drug release was elevated under redox and low acidic conditions. Furthermore, crosslinked micelles showed a superior anticancer effect compared to that of non-crosslinked micelles. Incorporation of docetaxel (DTX) and lonidamine (LND) in crosslinked polypeptide micelles increased the intracellular reactive oxygen species (ROS) level and oxidative stress and caused damage to intracellular components that resulted in greater apoptosis of cancer cells than when DTX or LND was used alone. The combination of DTX and LND in crosslinked micelles exhibited efficacious inhibition of tumor growth with an excellent safety profile compared to that reported for drug cocktail combinations and non-crosslinked micelles. Overall, redox/pH-responsive polypeptide micelles could be an interesting platform for efficient chemotherapy. STATEMENT OF SIGNIFICANCE We have synthesized a biodegradable polypeptide block copolymer to construct a facile pH and redox-responsive polymeric micelle asan advanced therapeutic system for cancer therapy. We have designed a corona-crosslinked triblock copolymer (poly (ethylene glycol)-b-poly(aspartic acid)-b-poly(tyrosine) (PEG-b-pAsp-b-pTyr)) micelles co-loaded with docetaxel and lonidamine (cl-M/DL). The corona of triblock polymer was crosslinked to maintain its structural integrity in the physiological environment. The mitochondrial targeting LND is expected to generate ROS, oxidative stress and thereby synergize the chemotherapeutic efficacy of DTX in killing cancer cells. Consistently, cl-M/DL exhibited excellent antitumor efficacy in xenograft tumor model with remarkable tumor regression. Overall, we demonstrated the construction of bioreducible nanosystem for the effective synergistic delivery of DTX/LND in tumor tissues towards cancer treatment.
Collapse
|
32
|
Lee KM, Lee K, Choi YK, Choi YJ, Seo HS, Ko SG. SH003‑induced G1 phase cell cycle arrest induces apoptosis in HeLa cervical cancer cells. Mol Med Rep 2017; 16:8237-8244. [PMID: 28944910 DOI: 10.3892/mmr.2017.7597] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 05/03/2017] [Indexed: 11/05/2022] Open
Abstract
Cervical cancer is a prevalent disease that may lead to mortality in women. In spite of the development of common therapeutic agents to treat cancer, there are several limitations of their use owing to side effects and drug resistance, which may induce cancer recurrence. The anticancer effects of the new herbal mixture SH003 (comprising Astragalus membranaceus, Angelica gigas and Trichosanthes kirilowii Maximowicz) have been examined in various types of cancer. Thus, the present study hypothesized that SH003 may be an effective treatment for cervical cancer. SH003 treatment inhibited the growth of HeLa cells, whereas it did not affect the growth of rat intestinal epithelial cells. In addition, SH003 treatment increased the expression of apoptosis‑related proteins and promoted apoptotic cell death in HeLa cells. SH003 treatment also led to G1 phase arrest in HeLa cells. Furthermore, SH003 treatment induced the production of reactive oxygen species (ROS); however, ROS production did not appear to be related to SH003‑mediated apoptosis. Results from the present study indicated that the SH003‑induced inhibition of HeLa cell growth may be mediated through G1 phase arrest and extrinsic apoptosis, suggested that SH003 may be a potential treatment for cervical cancer.
Collapse
Affiliation(s)
- Kang Min Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02247, Republic of Korea
| | - Kangwook Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02247, Republic of Korea
| | - Youn Kyung Choi
- Jeju International Marine Science Center for Research & Education, Korea Institute of Ocean Science and Technology, Jeju 63349, Republic of Korea
| | - Yu-Jeong Choi
- Department of Cancer Preventive Material Development, Graduate School, Kyung Hee University, Seoul 02247, Republic of Korea
| | - Hye-Sook Seo
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02247, Republic of Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02247, Republic of Korea
| |
Collapse
|
33
|
Tang Y, Liang J, Wu A, Chen Y, Zhao P, Lin T, Zhang M, Xu Q, Wang J, Huang Y. Co-Delivery of Trichosanthin and Albendazole by Nano-Self-Assembly for Overcoming Tumor Multidrug-Resistance and Metastasis. ACS APPLIED MATERIALS & INTERFACES 2017; 9:26648-26664. [PMID: 28741923 DOI: 10.1021/acsami.7b05292] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Multidrug resistance (MDR) and metastasis are the major obstacles in cancer chemotherapy. Nanotechnology-based combination therapy is a useful strategy. Recently, the combination of biologics and small drugs has attracted much attention in cancer therapy. Yet, the treatment outcomes are often compromised by the different pharmacokinetic profiles of the co-administered drugs thus leading to inconsistent drug uptake and suboptimal drug combination at the tumor sites. Nanotechnology-based co-delivery offers a promising method to address this problem, which is well demonstrated in the use of small drug combinations. However, co-delivery of the drugs bearing different physicochemical properties (e.g., proteins and small drugs) remains a formidable challenge. Herein, we developed a self-assembled nanosystem for co-delivery of trichosanthin (TCS) protein and albendazole (ABZ) as a combination therapy for overcoming MDR and metastasis. TCS is a ribosome-inactivating protein with high antitumor activity. However, the druggability of TCS is poor due to its short half-life, lack of tumor-specific action, and low cell uptake. ABZ is a clinically used antihelmintic drug, which can also inhibit tubulin polymerization and thus serve as a potential antitumor drug. In our work, ABZ was encapsulated in the albumin-coated silver nanoparticles (termed ABZ@BSA/Ag NP). The thus-formed NPs were negatively charged and could tightly bind with the cationic TCS that was modified with a cell-penetrating peptide (CPP) low-molecular-weight protamine (termed rTL). Via the stable charge interaction, the nanosystem (rTL/ABZ@BSA/Ag NP) was self-assembled, and featured by the TCS corona. The co-delivery system efficiently inhibited the proliferation of the drug-resistant tumor cells (A549/T and HCT8/ADR) by impairing the cytoskeleton, arresting the cell cycle, and enhancing apoptosis. In addition, the migration and invasion of tumor cells were inhibited presumably due to the impeded cytoskeleton functions. The anti-MDR effect was further confirmed by the in vivo studies with the subcutaneous A549/T tumor mouse model. More importantly, the co-delivery system was demonstrated to be able to inhibit metastasis. The co-delivery system of TCS/ABZ provided a potential strategy for both overcoming drug resistance and inhibiting tumor metastasis.
Collapse
Affiliation(s)
- Yisi Tang
- Guangzhou University of Chinese Medicine , 12 Ji-chang Road, Guangzhou 510450, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Jianming Liang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Aihua Wu
- Guangzhou University of Chinese Medicine , 12 Ji-chang Road, Guangzhou 510450, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Yingzhi Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Pengfei Zhao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Tingting Lin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
- Department of Pharmacy, Binzhou Medical University Hospital , Binzhou 256603, China
| | - Meng Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| | - Qin Xu
- Guangzhou University of Chinese Medicine , 12 Ji-chang Road, Guangzhou 510450, China
| | - Jianxin Wang
- Guangzhou University of Chinese Medicine , 12 Ji-chang Road, Guangzhou 510450, China
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, School of Pharmacy, Fudan University , Shanghai 201203, China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 501 Haike Road, Shanghai 201203, China
| |
Collapse
|
34
|
Chen Y, Zhang M, Jin H, Li D, Xu F, Wu A, Wang J, Huang Y. Glioma Dual-Targeting Nanohybrid Protein Toxin Constructed by Intein-Mediated Site-Specific Ligation for Multistage Booster Delivery. Am J Cancer Res 2017; 7:3489-3503. [PMID: 28912890 PMCID: PMC5596438 DOI: 10.7150/thno.20578] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/13/2017] [Indexed: 01/29/2023] Open
Abstract
Malignant glioma is one of the most untreatable cancers because of the formidable blood-brain barrier (BBB), through which few therapeutics can penetrate and reach the tumors. Biologics have been booming in cancer therapy in the past two decades, but their application in brain tumor has long been ignored due to the impermeable nature of BBB against effective delivery of biologics. Indeed, it is a long unsolved problem for brain delivery of macromolecular drugs, which becomes the Holy Grail in medical and pharmaceutical sciences. Even assisting by targeting ligands, protein brain delivery still remains challenging because of the synthesis difficulties of ligand-modified proteins. Herein, we propose a rocket-like, multistage booster delivery system of a protein toxin, trichosanthin (TCS), for antiglioma treatment. TCS is a ribosome-inactivating protein with the potent activity against various solid tumors but lack of specific action and cell penetration ability. To overcome the challenge of its poor druggability and site-specific modification, intein-mediated ligation was applied, by which a gelatinase-cleavable peptide and cell-penetrating peptide (CPP)-fused recombinant TCS toxin can be site-specifically conjugated to lactoferrin (LF), thus constructing a BBB-penetrating, gelatinase-activatable cell-penetrating nanohybrid TCS toxin. This nanohybrid TCS system is featured by the multistage booster strategy for glioma dual-targeting delivery. First, LF can target to the BBB-overexpressing low-density lipoprotein receptor-related protein-1 (LRP-1), and assist with BBB penetration. Second, once reaching the tumor site, the gelatinase-cleavable peptide acts as a separator responsive to the glioma-associated matrix metalloproteinases (MMPs), thus releasing to the CPP-fused toxin. Third, CPP mediates intratumoral and intracellular penetration of TCS toxin, thereby enhancing its antitumor activity. The BBB penetration and MMP-2-activability of this delivery system were demonstrated. The antiglioma activity was evaluated in the subcutaneous and orthotopic animal models. Our work provides a useful protocol for improving the druggability of such class of protein toxins and promoting their in-vivo application for targeted cancer therapy.
Collapse
|
35
|
Ruttala HB, Ramasamy T, Poudal BK, Choi Y, Choi JY, Kim J, Ku SK, Choi HG, Yong CS, Kim JO. Molecularly targeted co-delivery of a histone deacetylase inhibitor and paclitaxel by lipid-protein hybrid nanoparticles for synergistic combinational chemotherapy. Oncotarget 2017; 8:14925-14940. [PMID: 28122339 PMCID: PMC5362455 DOI: 10.18632/oncotarget.14742] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/10/2017] [Indexed: 01/05/2023] Open
Abstract
In this study, a transferrin-anchored albumin nanoplatform with PEGylated lipid bilayers (Tf-L-APVN) was developed for the targeted co-delivery of paclitaxel and vorinostat in solid tumors. Tf-L-APVN exhibited a sequential and controlled release profile of paclitaxel and vorinostat, with an accelerated release pattern at acidic pH. At cellular levels, Tf-L-APVN significantly enhanced the synergistic effects of paclitaxel and vorinostat on the proliferation of MCF-7, MDA-MB-231, and HepG2 cancer cells. Vorinostat could significantly enhance the cytotoxic potential of paclitaxel, induce marked cell apoptosis, alter cell cycle patterns, and inhibit the migratory capacity of cancer cells. In addition, Tf-L-APVN showed prolonged circulation in the blood and maintained an effective ratio of 1:1 (for paclitaxel and vorinostat) throughout the study period. In HepG2 tumor-bearing mice, Tf-L-APVN displayed excellent antitumor efficacy and the combination of paclitaxel and vorinostat significantly inhibited the tumor growth. Taken together, dual drug-loaded Tf receptor-targeted nanomedicine holds great potential in chemotherapy of solid tumors.
Collapse
Affiliation(s)
- Hima Bindu Ruttala
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| | - Thiruganesh Ramasamy
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| | - Bijay Kumar Poudal
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| | - Yongjoo Choi
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| | - Ju Yeon Choi
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| | - Jeonghwan Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 712-715, South Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Sangnok-gu, Ansan 426-791, South Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, South Korea
| |
Collapse
|
36
|
Lo HY, Li TC, Yang TY, Li CC, Chiang JH, Hsiang CY, Ho TY. Hypoglycemic effects of Trichosanthes kirilowii and its protein constituent in diabetic mice: the involvement of insulin receptor pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:53. [PMID: 28100206 PMCID: PMC5242006 DOI: 10.1186/s12906-017-1578-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/12/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Diabetes is a serious chronic metabolic disorder. Trichosanthes kirilowii Maxim. (TK) is traditionally used for the treatment of diabetes in traditional Chinese medicine (TCM). However, the clinical application of TK on diabetic patients and the hypoglycemic efficacies of TK are still unclear. METHODS A retrospective cohort study was conducted to analyze the usage of Chinese herbs in patients with type 2 diabetes in Taiwan. Glucose tolerance test was performed to analyze the hypoglycemic effect of TK. Proteomic approach was performed to identify the protein constituents of TK. Insulin receptor (IR) kinase activity assay and glucose tolerance tests in diabetic mice were further used to elucidate the hypoglycemic mechanisms and efficacies of TK. RESULTS By a retrospective cohort study, we found that TK was the most frequently used Chinese medicinal herb in type 2 diabetic patients in Taiwan. Oral administration of aqueous extract of TK displayed hypoglycemic effects in a dose-dependent manner in mice. An abundant novel TK protein (TKP) was further identified by proteomic approach. TKP interacted with IR by docking analysis and activated the kinase activity of IR. In addition, TKP enhanced the clearance of glucose in diabetic mice in a dose-dependent manner. CONCLUSIONS In conclusion, this study applied a bed-to-bench approach to elucidate the hypoglycemic efficacies and mechanisms of TK on clinical usage. In addition, we newly identified a hypoglycemic protein TKP from TK. Our findings might provide a reasonable explanation of TK on the treatment of diabetes in TCM.
Collapse
Affiliation(s)
- Hsin-Yi Lo
- Graduate Institute of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Tsai-Chung Li
- Graduate Institute of Biostatistics, China Medical University, Taichung, 40402, Taiwan
| | - Tse-Yen Yang
- Molecular and Genomic Epidemiology Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Chia-Cheng Li
- Graduate Institute of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Jen-Huai Chiang
- Management Office for Health Data, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Chien-Yun Hsiang
- Department of Microbiology, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
| | - Tin-Yun Ho
- Graduate Institute of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
37
|
Ramasamy T, Ruttala HB, Chitrapriya N, Poudal BK, Choi JY, Kim ST, Youn YS, Ku SK, Choi HG, Yong CS, Kim JO. Engineering of cell microenvironment-responsive polypeptide nanovehicle co-encapsulating a synergistic combination of small molecules for effective chemotherapy in solid tumors. Acta Biomater 2017; 48:131-143. [PMID: 27794477 DOI: 10.1016/j.actbio.2016.10.034] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 11/17/2022]
Abstract
In this study, we report a facile method to construct a bioactive (poly(phenylalanine)-b-poly(l-histidine)-b-poly(ethylene glycol) polypeptide nanoconstruct to co-load doxorubicin (DOX) and quercetin (QUR) (DQ-NV). The smart pH-sensitive nanovehicle was fabricated with precisely tailored drug-to-carrier ratio that resulted in accelerated, sequential drug release. As a result of ratiometric loading, QUR could significantly enhance the cytotoxic potential of DOX, induced marked cell apoptosis; change cell cycle patterns, inhibit the migratory capacity of sensitive and resistant cancer cells. In particular, pro-oxidant QUR from DQ-NV remarkably reduced the GSH/GSSG ratio, indicating high oxidative stress and damage to cellular components. DQ-NV induced tumor shrinkage more effectively than the single drugs in mice carrying subcutaneous SCC-7 xenografts. DQ-NV consistently induced high expression of caspase-3 and PARP and low expression of Ki67 and CD31 immunomarkers. In summary, we demonstrate the development of a robust polypeptide-based intracellular nanovehicle for synergistic delivery of DOX/QUR in cancer chemotherapy. STATEMENT OF SIGNIFICANCE In this study, we report a facile method to construct bioactive and biodegradable polypeptide nanovehicles as an advanced platform technology for application in cancer therapy. We designed a robust (poly(phenylalanine)-b-poly(l-histidine)-b-poly(ethylene glycol) nanoconstruct to co-load doxorubicin (DOX) and quercetin (QUR) (DQ-NV). The conformational changes of the histidine block at tumor pH resulted in accelerated, sequential drug release. QUR could significantly enhance the cytotoxic potential of DOX, induce marked cell apoptosis, change cell cycle patterns, and inhibit the migratory capacity of sensitive and resistant cancer cells. DQ-NV induced tumor shrinkage more effectively than the single drugs and the 2-drug cocktail in tumor xenografts. In summary, we demonstrate the development of an intracellular nanovehicle for synergistic delivery of DOX/QUR in cancer chemotherapy.
Collapse
Affiliation(s)
- Thiruganesh Ramasamy
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Hima Bindu Ruttala
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Nataraj Chitrapriya
- Biophysical Chemistry Lab, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Bijay Kumar Poudal
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Ju Yeon Choi
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Ssang Tae Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, SungKyunKwan University, 300 Cheoncheon-dong, Jangan-gu, Suwon, 440-746, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan 712-715, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea.
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea.
| |
Collapse
|
38
|
Choi YJ, Choi YK, Lee KM, Cho SG, Kang SY, Ko SG. SH003 induces apoptosis of DU145 prostate cancer cells by inhibiting ERK-involved pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:507. [PMID: 27927199 PMCID: PMC5142381 DOI: 10.1186/s12906-016-1490-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/17/2016] [Indexed: 01/22/2023]
Abstract
Background Herbal medicines have been used in cancer treatment, with many exhibiting favorable side effect and toxicity profiles compared with conventional chemotherapeutic agents. SH003 is a novel extract from Astragalus membranaceus, Angelica gigas, and Trichosanthes Kirilowii Maximowicz combined at a 1:1:1 ratio that impairs the growth of breast cancer cells. This study investigates anti-cancer effects of SH003 in prostate cancer cells. Methods SH003 extract in 30% ethanol was used to treat the prostate cancer cell lines DU145, LNCaP, and PC-3. Cell viability was determined by MTT and BrdU incorporation assays. Next, apoptotic cell death was determined by Annexin V and 7-AAD double staining methods. Western blotting was conducted to measure protein expression levels of components of cell death and signaling pathways. Intracellular reactive oxygen species (ROS) levels were measured using H2DCF-DA. Plasmid-mediated ERK2 overexpression in DU145 cells was used to examine the effect of rescuing ERK2 function. Results were analyzed using the Student’s t-test and P-values < 0.05 were considered to indicate statistically-significant differences. Results Our data demonstrate that SH003 induced apoptosis in DU145 prostate cancer cells by inhibiting ERK signaling. SH003 induced apoptosis of prostate cancer cells in dose-dependent manner, which was independent of androgen dependency. SH003 also increased intracellular ROS levels but this is not associated with its pro-apoptotic effects. SH003 inhibited phosphorylation of Ras/Raf1/MEK/ERK/p90RSK in androgen-independent DU145 cells, but not androgen-dependent LNCaP and PC-3 cells. Moreover, ERK2 overexpression rescued SH003-induced apoptosis in DU145 cells. Conclusions SH003 induces apoptotic cell death of DU145 prostate cancer cells by inhibiting ERK2-mediated signaling. Electronic supplementary material The online version of this article (doi:10.1186/s12906-016-1490-5) contains supplementary material, which is available to authorized users.
Collapse
|
39
|
Chen Y, Zhang M, Jin H, Tang Y, Wang H, Xu Q, Li Y, Li F, Huang Y. Intein-mediated site-specific synthesis of tumor-targeting protein delivery system: Turning PEG dilemma into prodrug-like feature. Biomaterials 2016; 116:57-68. [PMID: 27914267 DOI: 10.1016/j.biomaterials.2016.11.033] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/23/2016] [Accepted: 11/23/2016] [Indexed: 12/30/2022]
Abstract
Poor tumor-targeted and cytoplasmic delivery is a bottleneck for protein toxin-based cancer therapy. Ideally, a protein toxin drug should remain stealthy in circulation for prolonged half-life and reduced side toxicity, but turn activated at tumor. PEGylation is a solution to achieve the first goal, but creates a hurdle for the second because PEG rejects interaction between the drugs and tumor cells therein. Such PEG dilemma is an unsolved problem in protein delivery. Herein proposed is a concept of turning PEG dilemma into prodrug-like feature. A site-selectively PEGylated, gelatinase-triggered cell-penetrating trichosanthin protein delivery system is developed with three specific aims. The first is to develop an intein-based ligation method for achieving site-specific modification of protein toxins. The second is to develop a prodrug feature that renders protein toxins remaining stealthy in blood for reduced side toxicity and improved EPR effect. The third is to develop a gelatinase activatable cell-penetration strategy for enhanced tumor targeting and cytoplasmic delivery. Of note, site-specific modification is a big challenge in protein drug research, especially for such a complicated, multifunctional protein delivery system. We successfully develop a protocol for constructing a macromolecular prodrug system with intein-mediated ligation synthesis. With an on-column process of purification and intein-mediated cleavage, the site-specific PEGylation then can be readily achieved by conjugation with the activated C-terminus, thus constructing a PEG-capped, cell-penetrating trichosanthin system with a gelatinase-cleavable linker that enables tumor-specific activation of cytoplasmic delivery. It provides a promising method to address the PEG dilemma for enhanced protein drug delivery, and importantly, a facile protocol for site-specific modification of such a class of protein drugs for improving their druggability and industrial translation.
Collapse
Affiliation(s)
- Yingzhi Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai 201203, China; University of Chinese Academy of Sciences, 19A Yuquan Rd, Beijing 100049, China
| | - Meng Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai 201203, China; University of Chinese Academy of Sciences, 19A Yuquan Rd, Beijing 100049, China
| | - Hongyue Jin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai 201203, China
| | - Yisi Tang
- Guangzhou University of Chinese Medicine Tropical Medical Institute, 12 Ji-chang Rd, Guangzhou 510405, China
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai 201203, China
| | - Qin Xu
- Guangzhou University of Chinese Medicine Tropical Medical Institute, 12 Ji-chang Rd, Guangzhou 510405, China
| | - Yaping Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai 201203, China
| | - Feng Li
- Hampton University School of Pharmacy, Kittrell Hall RM216, Hampton, VA 23668, USA
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Hai-ke Rd, Shanghai 201203, China.
| |
Collapse
|
40
|
Zhang L, Zhang J, Qi B, Jiang G, Liu J, Zhang P, Ma Y, Li W. The anti-tumor effect and bioactive phytochemicals of Hedyotis diffusa willd on ovarian cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:132-139. [PMID: 27426510 DOI: 10.1016/j.jep.2016.07.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa willd (HDW) is a widely used medicinal herb in China. It processed various medicinal properties including antioxidative, anti-inflamatory and anti-cancer effects. This study aimed to investigate the anti-tumor effects of HDW on ovarian cancer cells and the underlying mechanisms as well as identify the bioactive compounds. MATERIALS AND METHODS Effects of HDW on the viability of ovarian cancer A2780 cells were detected by MTT assay. Apoptosis was detected by cell morphologic observation through DAPI staining and flow cytometry analysis. The migration of ovarian cancer cells which exposed to HDW were detected by wound healing and transwell assays. The protein levels of caspase 3/9, Bcl-2 and MMP-2/9 in human ovarian cancer cells treated with HDW were assessed by western blotting analysis. The potential bioactive compounds were characterized by HPLC-Q-TOF-MS. RESULTS HDW significantly inhibited the growth of A2780 ovarian cancer cells and induced apoptosis. The induction of apoptosis by HDW was associated with down-regulation of anti-apoptotic protein Bcl-2 and the activation of caspase 3/9. Wound healing and transwell chamber assays indicated HDW suppressed the migration of ovarian cancer cells. HDW dramatically decreased MMP-2/9 expression. A HPLC-Q-TOF-MS analysis of HDW indicated the presence of 13 flavonoids compounds and one anthraquinone compound, which may contribute to the anticancer activity of the HDW. CONCLUSIONS HDW effectively restricted the growth of ovarian cancer cells and induced apoptosis through the mitochondria-associated apoptotic pathway. Furthermore, HDW suppressed the migration of ovarian cancer cells through down-regulation of MMP-2 and MMP-9 expression. These results showed that HDW hold potential therapeutic effect for ovarian cancer patients.
Collapse
Affiliation(s)
- Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Jing Zhang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Bing Qi
- Acute Abdomen Department of the First Affiliated Hospital, Dalian medical University, Dalian 116000, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Pei Zhang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Yuan Ma
- Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
41
|
Lu YZ, Li PF, Li YZ, Luo F, Guo C, Lin B, Cao XW, Zhao J, Wang FJ. Enhanced anti-tumor activity of trichosanthin after combination with a human-derived cell-penetrating peptide, and a possible mechanism of activity. Fitoterapia 2016; 112:183-90. [DOI: 10.1016/j.fitote.2016.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/27/2016] [Accepted: 03/28/2016] [Indexed: 12/13/2022]
|
42
|
Li J, Li H, Zhang Z, Wang N, Zhang Y. The anti-cancerous activity of recombinant trichosanthin on prostate cancer cell PC3. Biol Res 2016; 49:21. [PMID: 27015938 PMCID: PMC4807558 DOI: 10.1186/s40659-016-0081-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/18/2016] [Indexed: 01/07/2023] Open
Abstract
CONTEXT Trichosanthin produced in the root tube of Trichosanthes kirilowii shows anti-tumor activity on a series of cancer cells including Hela, MCF-7, HL-60. But there is little information about its effect on the carcinogenesis of prostate cancer. OBJECTIVE This work was designed to study the role of trichosanthin on prostate cancer cells PC3. MATERIALS AND METHODS Trichosanthin was expressed in BL21 strain and purified by affinity chromatography. MTT assay was designed to determine the effect of trichosanthin on growth of PC3 cells at doses of 10, 20, 40, 60, 80, and 120 μg/ml. Then the effect of 50 μg/ml rTCS alone or combined with 2 μM IL-2 on PC3 cell proliferation was analyzed. And the mechanism of rTCS was studied by western blot. After that the in vivo effect of rTCS combined with IL-2 was explored in mice bearing PC3 xenograft tumor. RESULTS Trichosanthin was successfully expressed in BL21 and purified by 100 mM imidazole. It was shown to inhibit proliferation of PC3 cells in a dose-dependent manner with IC50 50.6 μg/ml. When combined with cytokine IL-2, a significant synergic effect was obtained. The inhibition rate on PC3 was around 50 % in combination group while only 35.5 % in single rTCS group at 50 μg/ml. Further, the expression of full length caspase-8 and Bcl-2 decreased significantly while cleaved caspase-8 and Bax were up-regulated, which suggest that caspase-8-mediated apoptosis pathway may be activated by rTCS in PC3 cells. Moreover, our data demonstrated that tumor volume and tumor weight were significantly reduced in rTCS-treated or rTCS/IL-2-treated nude mice bearing PC3 xenograft tumor compared with control. And significant difference was also found between rTCS and rTCS/IL-2 group. CONCLUSIONS This study demonstrates that rTCS is a potential agent with high in vitro and in vivo anti-tumor activity on PC3 cells. And rTCS combined with IL-2 is a promising strategy in treating patients with prostate cancer in future.
Collapse
Affiliation(s)
- JinLong Li
- />Department of Laboratory Medicine, The Second Affiliated Hospital of Southeast University, Zhongfu Road 1-1, Nanjing, 210003 China
- />Department of Biochemistry and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210093 China
| | - Hui Li
- />Department of Neonatology, The Taizhou People’s Hospital, Taizhou, 225300 China
| | - ZhaoLi Zhang
- />Department of Pharmacy, The Second Affiliated Hospital of Southeast University, Nanjing, 210003 China
| | - NianYue Wang
- />Department of Laboratory Medicine, The Second Affiliated Hospital of Southeast University, Zhongfu Road 1-1, Nanjing, 210003 China
| | - YongChen Zhang
- />Department of Laboratory Medicine, The Second Affiliated Hospital of Southeast University, Zhongfu Road 1-1, Nanjing, 210003 China
| |
Collapse
|
43
|
Trichosanthin-induced autophagy in gastric cancer cell MKN-45 is dependent on reactive oxygen species (ROS) and NF-κB/p53 pathway. J Pharmacol Sci 2016; 131:77-83. [PMID: 27032906 DOI: 10.1016/j.jphs.2016.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 02/29/2016] [Accepted: 03/01/2016] [Indexed: 12/29/2022] Open
Abstract
Trichosanthin (TCS), isolated from the root tuber of Trichosanthes kirilowii tubers in the Cucurbitaceae family, owns a great deal of biological and pharmacological activities including anti-virus and anti-tumor. TCS has been reported to induce cell apoptosis of a diversity of cancers, including cervical cancer, choriocarcinoma, and gastric cancer, etc. However, whether TCS would induce autophagy in gastric cancer cells was seldom investigated. In current study, human gastric cancer MKN-45 cell growth was significantly inhibited by TCS. The anti-proliferation effect of TCS was due to an increased autophagy, which was confirmed by monodansylcadervarine (MDC) staining, up-regulation of Autophagy protein 5 (Atg5), and conversion of LC3 I to LC3 II (autophagosome marker). In addition, TCS induced reactive oxygen species (ROS) in MKN-45 cells and ROS scavenger N-acetylcysteine (NAC) significantly reversed TCS-induced autophagy. Furthermore, NF-κB/p53 pathway was activated during the process of autophagy induced by TCS and the ROS generation was mediated by it in MKN-45 cells. In vivo results showed that TCS exerted significantly anti-tumor effect on MKN-45 bearing mice. Considering the clinical usage of TCS on other human diseases, these research progresses provided a new insight into cancer research and new therapeutic avenues for patients with gastric cancer.
Collapse
|
44
|
Song L, Chang J, Li Z. A serine protease extracted from Trichosanthes kirilowii induces apoptosis via the PI3K/AKT-mediated mitochondrial pathway in human colorectal adenocarcinoma cells. Food Funct 2016; 7:843-54. [DOI: 10.1039/c5fo00760g] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A novel protein TKP extracted from T. kirilowii fruit exerted potential anti-colorectal cancer activity by inducing apoptosis, which was regulated by the PI3K/AKT-mediated mitochondria-dependent pathway.
Collapse
Affiliation(s)
- Li Song
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| | - Jiao Chang
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| | - Zhuoyu Li
- Institute of Biotechnology
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education
- Shanxi University
- Taiyuan 030006
- China
| |
Collapse
|
45
|
ZHU YINGJIE, SUN YUELI, CAI YUCHEN, SHA OU, JIANG WENQI. Trichosanthin reduces the viability of SU-DHL-2 cells via the activation of the extrinsic and intrinsic apoptotic pathways. Mol Med Rep 2015; 13:403-11. [DOI: 10.3892/mmr.2015.4531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 10/06/2015] [Indexed: 11/06/2022] Open
|
46
|
Akkouh O, Ng TB, Cheung RCF, Wong JH, Pan W, Ng CCW, Sha O, Shaw PC, Chan WY. Biological activities of ribosome-inactivating proteins and their possible applications as antimicrobial, anticancer, and anti-pest agents and in neuroscience research. Appl Microbiol Biotechnol 2015; 99:9847-63. [PMID: 26394859 DOI: 10.1007/s00253-015-6941-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/10/2015] [Accepted: 08/13/2015] [Indexed: 02/06/2023]
Abstract
Ribosome-inactivating proteins (RIPs) are enzymes which depurinate ribosomal RNA (rRNA), thus impeding the process of translation resulting in inhibition of protein synthesis. They are produced by various organisms including plants, fungi and bacteria. RIPs from plants are linked to plant defense due to their antiviral, antifungal, antibacterial, and insecticidal activities in which they can be applied in agriculture to combat microbial pathogens and pests. Their anticancer, antiviral, embryotoxic, and abortifacient properties may find medicinal applications. Besides, conjugation of RIPs with antibodies or other carriers to form immunotoxins has been found useful to research in neuroscience and anticancer therapy.
Collapse
Affiliation(s)
- Ouafae Akkouh
- Department of Biology and Medical Laboratory Research, Faculty of Technology, University of Applied Sciences Leiden, Zernikdreef 11, 2333 CK, Leiden, The Netherlands.
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Randy Chi Fai Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Jack Ho Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Wenliang Pan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Charlene Cheuk Wing Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Ou Sha
- School of Medicine, Shenzhen University Health Science Centre, Shenzhen University, Shenzhen, China.
| | - Pang Chui Shaw
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Wai Yee Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| |
Collapse
|
47
|
Miao J, Jiang Y, Wang D, Zhou J, Fan C, Jiao F, Liu B, Zhang J, Wang Y, Zhang Q. Trichosanthin suppresses the proliferation of glioma cells by inhibiting LGR5 expression and the Wnt/β-catenin signaling pathway. Oncol Rep 2015; 34:2845-52. [PMID: 26397053 PMCID: PMC4722885 DOI: 10.3892/or.2015.4290] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/20/2015] [Indexed: 01/16/2023] Open
Abstract
Studies have indicated that trichosanthin (TCS), a bioactive protein extracted and purified from the tuberous root of Trichosanthes kirilowii (a well-known traditional Chinese medicinal plant), produces antitumor effects on various types of cancer cells. However, the effects of TCS on glioma cells are poorly understood. The objective of this study was to investigate the antitumor effects of TCS on the U87 and U251 cell lines. The in vitro effects of TCS on these two cell lines were determined using a Cell Counting Kit-8 (CCK-8) assay, Annexin V-FITC staining, DAPI staining, Transwell assays, terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) assays, 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethyl-imidacar-bocyanine iodide (JC-1) staining and western blotting, which was utilized to assess the expression of leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) and key proteins in the Wnt/β-catenin signaling pathway. Our data indicated that TCS inhibited the proliferation of glioma cells in a dose- and time-dependent manner and played a role in inhibiting glioma cell invasion and migration. Additional investigation revealed that the expression levels of LGR5 and of key proteins in the Wnt/β-catenin signaling pathway were markedly decreased after TCS treatment. The results suggest that TCS may induce apoptosis in glioma cells by targeting LGR5 and repressing the Wnt/β-catenin signaling pathway. In the future, in vivo experiments should be conducted to examine the potential use of this compound as a novel therapeutic agent for gliomas.
Collapse
Affiliation(s)
- Junjie Miao
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Yilin Jiang
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Dongliang Wang
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jingru Zhou
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Cungang Fan
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Feng Jiao
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Bo Liu
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jun Zhang
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Yangshuo Wang
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Qingjun Zhang
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| |
Collapse
|
48
|
Zeng M, Zheng M, Lu D, Wang J, Jiang W, Sha O. Anti-tumor activities and apoptotic mechanism of ribosome-inactivating proteins. CHINESE JOURNAL OF CANCER 2015; 34:325-34. [PMID: 26184404 PMCID: PMC4593346 DOI: 10.1186/s40880-015-0030-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/14/2015] [Indexed: 01/22/2023]
Abstract
Ribosome-inactivating proteins (RIPs) belong to a family of enzymes that attack eukaryotic ribosomes and potently inhibit cellular protein synthesis. RIPs possess several biomedical properties, including anti-viral and anti-tumor activities. Multiple RIPs are known to inhibit tumor cell proliferation through inducing apoptosis in a variety of cancers, such as breast cancer, leukemia/lymphoma, and hepatoma. This review focuses on the anti-tumor activities of RIPs and their apoptotic effects through three closely related pathways: mitochondrial, death receptor, and endoplasmic reticulum pathways.
Collapse
Affiliation(s)
- Meiqi Zeng
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Manyin Zheng
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Desheng Lu
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Jun Wang
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| | - Wenqi Jiang
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
- School of Medicine, Shenzhen University, Shenzhen, 518060, Guangdong, People's Republic of China.
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, 510060, People's Republic of China.
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Ou Sha
- School of Medicine, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, People's Republic of China.
| |
Collapse
|
49
|
Riproximin modulates multiple signaling cascades leading to cytostatic and apoptotic effects in human breast cancer cells. J Cancer Res Clin Oncol 2015; 142:135-47. [DOI: 10.1007/s00432-015-2013-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
|
50
|
Dan X, Wong JH, Fang EF, Chan FCW, Ng TB. Purification and Characterization of a Novel Hemagglutinin with Inhibitory Activity toward Osteocarcinoma Cells from Northeast China Black Beans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:3903-3914. [PMID: 25816710 DOI: 10.1021/acs.jafc.5b00106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In the present study, we isolated a novel hemagglutinin from an edible legume and explored its growth-inhibitory effect on osteocarcinoma and liver cancer cells. The protein was purified by liquid chromatography techniques which entailed affinity chromatography on Affi-gel blue gel, ion-exchange chromatography on Mono Q, and gel filtration on Superdex 75 with an FPLC system. The hemagglutinating activity of this hemagglutinin was demonstrated to be ion dependent and stable over a wide range of temperature and pH values. Antiproliferative activity was observed in the tumor cell lines MG-63 and HepG2 but not in the normal cell line WRL 68. Osteocarcinoma cells treated with the hemagglutinin underwent obvious cell shrinkage, chromatin condensation, mitochondrial membrane depolarization, and apoptosis. The mRNA expression level of interleukin-2 (IL-2), interleukin-6 (IL-6), interleukin-1 beta (IL-1β), interferon-gamma (IFN-γ), and tumor necrosis factor alpha (TNF-α) were found to be up-regulated to different extents after treatment of this hemagglutinin.
Collapse
Affiliation(s)
- Xiuli Dan
- †School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jack Ho Wong
- †School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Evandro Fei Fang
- ‡National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States
| | - Francis Chun Wai Chan
- §School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Tzi Bun Ng
- †School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|