1
|
Zhang D, Chen Y, Sun Y, Xu H, Wei R, Zhou Y, Li F, Li J, Wang J, Chen P, Xi L. Gambogic acid induces GSDME dependent pyroptotic signaling pathway via ROS/P53/Mitochondria/Caspase-3 in ovarian cancer cells. Biochem Pharmacol 2025; 232:116695. [PMID: 39643123 DOI: 10.1016/j.bcp.2024.116695] [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: 08/15/2024] [Revised: 10/27/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Gambogic acid (GA) is a naturally active compound extracted from the Garcinia hanburyi with various anticancer activities. However, whether GA induces pyroptosis (a newly discovered inflammation-mediated programmed cell death mechanism) in ovarian cancer (OC) has not yet been reported. This study revealed that GA treatment reduced cell viability by inducing pyroptosis in OC cell lines. Typical pyroptosis morphological manifestations such as cell swelling with large bubbles and loss of cell membrane integrity, were observed. Cleaved caspase-3 and GSDME-N levels increased after GA treatment, and knocking out GSDME or using a caspase-3 inhibitor could switch GA-induced cell death from pyroptosis to apoptosis, indicating GA induced caspase-3/GSDME-dependent pyroptosis. Furthermore, this research indicated that GA significantly increased reactive oxygen species (ROS) and p53 phosphorylation. OC cells pretreated with ROS inhibitor N-Acetylcysteine (NAC) and the specific p53 inhibitor pifithrin-μ could completely reverse the pyroptosis post-treatment. Elevated p53 and phosphorylated p53 reduced mitochondrial membrane potential (MMP) and Bcl-2, increase the expression of Bax, and damage mitochondria by releasing cytochrome c to activate the downstream pyroptosis pathway. Different doses of GA inhibited tumor growth in ID8 tumor-bearing mice, and high-dose GA increased in tumor-infiltrating lymphocytes CD3, CD4, and CD8 were detected in tumor tissues. Notably, the expressions of GSDME-N, cleaved caspase-3 and other proteins were increased in tumor tissues with high-dose GA groups. These findings demonstrate that GA-treated OC cells could induce GSDME-mediated pyroptosis through the ROS/p53/mitochondria signaling pathway and caspase-3/-9 activation. Thus, GA is a promising therapeutic agent for OC treatment.
Collapse
Affiliation(s)
- Danya Zhang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yuxin Chen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yue Sun
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hanjie Xu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Obstetrics and Gynecology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Rui Wei
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Fei Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jie Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Pingbo Chen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Ling Xi
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
2
|
Joha Z, Öztürk A, Yulak F, Karataş Ö, Ataseven H. Mechanism of anticancer effect of gambogic acid on gastric signet ring cell carcinoma. Med Oncol 2023; 40:269. [PMID: 37587317 DOI: 10.1007/s12032-023-02149-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
Gambogic acid has demonstrated inhibitory effects on the growth of various cancer cell types, such as breast cancer, pancreatic cancer, prostate cancer, lung cancer, and osteosarcoma. This study aims to investigate the antiproliferative activity of Gambogic acid on SNU-16 cells derived from gastric signet ring cell carcinoma and elucidate the underlying mechanisms. The cytotoxic effect of gambogic acid was evaluated in SNU-16 cells by treating them with different concentrations of the compound, and the XTT cell viability assay was employed to assess cell viability. ELISA was used to measure bax, BCL-2, caspase 3, PARP, and 8-oxo-dG levels. Additionally, immunofluorescence staining was applied to assess 8-oxo-dG and LC3β levels in SNU-16 cells. It was observed that gambogic acid exerted a dose-dependent and statistically significant antiproliferative effect on SNU-16 cells. The IC50 value of gambogic acid in SNU-16 cells was found to be 655.1 nM for 24 h. Subsequent investigations conducted using the IC50 dose revealed a significant upregulation of apoptotic proteins including cleaved caspase 3, Bax, and cleaved PARP (p < 0.001), along with a downregulation of BCL-2 (p < 0.001), an anti-apoptotic protein. Moreover, the administration of this drug led to an upregulation of 8-oxo-dG (p < 0.001), a widely acknowledged biomarker indicating oxidative damage in DNA, as well as an increase in LC3β levels (p < 0.05), a marker associated with autophagy. The antiproliferative effect of gambogic acid against gastric signet ring cell carcinoma is attributed to its ability to induce apoptosis and autophagy. This discovery highlights the promising potential of gambogic acid as a treatment option for gastric signet ring cell carcinoma.
Collapse
Affiliation(s)
- Ziad Joha
- Department of Pharmacology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey.
| | - Ayşegül Öztürk
- Departments of Medical Services and Techniques, Vocational School of Health Services, Sivas Cumhuriyet University, Sivas, Turkey
| | - Fatih Yulak
- Department of Physiology, School of Medicine, Cumhuriyet University, Sivas, Turkey
| | - Özhan Karataş
- Department of Veterinary Pathology, School of Veterinary Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Hilmi Ataseven
- Departments of Pharmacology, School of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
3
|
Ji Y, Li J, Xiao S, Kwan HY, Bian Z, Chu CC. Optimization of amino acid-based poly(ester urea urethane) nanoparticles for the systemic delivery of gambogic acid for treating triple negative breast cancer. Biomater Sci 2023. [PMID: 37144899 DOI: 10.1039/d3bm00128h] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Amino acid-based poly(ester urea urethane) (AA-PEUU) is developed from amino acid-based ester urea building blocks interconnected with urethane blocks functionalized with poly(ethylene glycol) (PEG). Each functional block consists of structural design features that could impact the properties and performances of AA-PEUU as a nanocarrier for the systemic delivery of gambogic acid (GA). The multifunctional AA-PEUU structure provides broad tunability to enable the optimization of nanocarriers. The study investigates the structure-property relationship by fine-tuning the structure of AA-PEUU, including the amino acid type, hydrocarbons, the ratio of functional building blocks, and PEGylation, to identify the nanoparticle candidate with optimized delivery performances. Compared to free GA, the optimized PEUU nanocarrier improves the intratumoral distribution of GA by more than 9-fold, which significantly enhances the bioavailability and persistence of GA after intravenous administration. In an MDA-MB-231 xenograft mouse model, GA delivered by the optimized AA-PEUU nanocarrier exhibits significant tumor inhibition, apoptosis induction, and the anti-angiogenesis effect. The study demonstrates the potency of engineering AA-PEUU nanocarriers with tailor-designed structures and versatile tunability for the systemic delivery of therapeutics in the treatment of triple negative breast tumor.
Collapse
Affiliation(s)
- Ying Ji
- Institute of Textiles and Clothing, School of Fashion and Textiles, Research Institute for Intelligent Wearable Systems, Hong Kong Polytechnic University, Hunghom, Kowloon, Hong Kong SAR.
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shilin Xiao
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR
| | - Chih-Chang Chu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
4
|
Wippel HH, Chavez JD, Keller AD, Bruce JE. Multiplexed Isobaric Quantitative Cross-Linking Reveals Drug-Induced Interactome Changes in Breast Cancer Cells. Anal Chem 2022; 94:2713-2722. [PMID: 35107270 PMCID: PMC8969885 DOI: 10.1021/acs.analchem.1c02208] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The study of protein structures and interactions is critical to understand their function. Chemical cross-linking of proteins with mass spectrometry (XL-MS) is a rapidly developing structural biology technique able to provide valuable insight into protein conformations and interactions, even as they exist within their native cellular environment. Quantitative analysis of cross-links can reveal protein conformational and interaction changes that occur as a result of altered biological states, environmental conditions, or pharmacological perturbations. Our laboratory recently developed an isobaric quantitative protein interaction reporter (iqPIR) cross-linking strategy for comparative interactome studies. This strategy relies on isotope encoded chemical cross-linkers that have the same molecular mass yet produce unique and specific isotope signatures upon fragmentation in the mass spectrometer which can be used for quantitative analysis of cross-linked peptides. The initial set of iqPIR molecules allowed for binary comparisons. Here, we describe the in vivo application of an extended set of six iqPIR reagents (6-plex iqPIR), allowing multiplexed quantitative interactome analysis of up to six biological samples in a single LC-MS acquisition. Multiplexed iqPIR is demonstrated on MCF-7 breast cancer cells treated with five different Hsp90 inhibitors revealing large scale protein conformational and interaction changes specific to the molecular class of the inhibitors.
Collapse
Affiliation(s)
| | | | - Andrew D. Keller
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - James E. Bruce
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Phang YL, Zheng C, Xu H. Structural diversity and biological activities of caged Garcinia xanthones: recent updates. ACTA MATERIA MEDICA 2022; 1. [DOI: 10.15212/amm-2022-0001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Caged xanthones are a class of natural compounds with approximately 200 members that are commonly isolated from the Garcinia genus in the Clusiaceae (formerly Guttiferae) family. They are often characterized by a notable 4-oxa-tricyclo[4.3.1.03,7]dec-2-one (caged) architecture with a common xanthone backbone. Because most caged xanthones have potent anticancer properties, they have become a target of interest in natural product chemistry. The unique chemical architectures and increasingly identified biological importance of these compounds have stimulated many studies and intense interest in their isolation, biological evaluation and mechanistic studies. This review summarizes recent progress and development in the chemistry and biological activity of caged Garcinia xanthones and of several compounds of non-Garcinia origin, from the years 2008 to 2021, providing an in-depth discussion of their structural diversity and medicinal potential. A preliminary discussion on structure-activity relationships is also provided.
Collapse
|
6
|
Sawanny R, Pramanik S, Agarwal U. Role of Phytochemicals in the Treatment of Breast Cancer: Natural Swords Battling Cancer Cells. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716666210106123255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Breast cancer is the most common type of malignancy among ladies (around 30% of
newly diagnosed patients every year). To date, various modern treatment modalities for breast cancer,
such as radiotherapy, surgical method, hormonal therapy, and chemotherapeutic drug utilisation,
are available. However, adverse drug reactions, therapeutic resistance, metastasis, or cancer reoccurrence
chances remain the primary causes of mortality for breast cancer patients. To overcome
all the potential drawbacks, we need to investigate novel techniques and strategies that are not considered
previously to treat breast cancer effectively with safety and efficacy. For centuries, we
utilise phytochemicals to treat various diseases because of their safety, low-cost, and least or no
side effects. Recently, naturally produced phytochemicals gain immense attention as potential
breast cancer therapeutics because of their ideal characteristics; for instance, they operate via modulating
molecular pathways associated with cancer growth and progression. The primary mechanism
involves inhibition of cell proliferation, angiogenesis, migration, invasion, increasing anti-oxidant
status, initiation of the arrest of the cell cycle, and apoptosis. Remedial viability gets effectively enhanced
when phytochemicals work as adjuvants with chemotherapeutic drugs. This comprehensive
review revolves around the latest chemopreventive, chemotherapeutic, and chemoprotective treatments
with their molecular mechanisms to treat breast cancer by utilising phytochemicals such as
vinca alkaloids, resveratrol, curcumin, paclitaxel, silibinin, quercetin, genistein, and epigallocatechin
gallate. The authors wish to extend the field of phytochemical study for its scientific validity
and its druggability.
Collapse
Affiliation(s)
- Rajni Sawanny
- Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Institutional Area, Greater Noida, Uttar Pradesh-201306, India
| | - Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu-600036, India
| | - Unnati Agarwal
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Delhi, Grand Trunk Road, Phagwara, Punjab-144001, India
| |
Collapse
|
7
|
Yao CL, Zhang JQ, Li JY, Wei WL, Wu SF, Guo DA. Traditional Chinese medicine (TCM) as a source of new anticancer drugs. Nat Prod Rep 2021; 38:1618-1633. [PMID: 33511969 DOI: 10.1039/d0np00057d] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Covering: up to July 2020Drugs derived from traditional Chinese medicine (TCM) include both single chemical entities and multi-component preparations. Drugs of both types play a significant role in the healthcare system in China, but are not well-known outside China. The research and development process, the molecular mechanisms of action, and the clinical evaluation associated with some exemplificative anticancer drugs based on TCM are discussed, along with their potential of integration in western medicine.
Collapse
Affiliation(s)
- Chang-Liang Yao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - Jian-Qing Zhang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - Jia-Yuan Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - Wen-Long Wei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - Shi-Fei Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China.
| |
Collapse
|
8
|
Liu Y, Chen Y, Lin L, Li H. Gambogic Acid as a Candidate for Cancer Therapy: A Review. Int J Nanomedicine 2020; 15:10385-10399. [PMID: 33376327 PMCID: PMC7764553 DOI: 10.2147/ijn.s277645] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Gambogic acid (GA), a kind of dry resin secreted by the Garcinia hanburyi tree, is a natural active ingredient with various biological activities, such as anti-cancer, anti-inflammatory, antioxidant, anti-bacterial effects, etc. An increasing amount of evidence indicates that GA has obvious anti-cancer effects via various molecular mechanisms, including the induction of apoptosis, autophagy, cell cycle arrest and the inhibition of invasion, metastasis, angiogenesis. In order to improve the efficacy in cancer treatment, nanometer drug delivery systems have been employed to load GA and form micelles, nanoparticles, nanofibers, and so on. In this review, we aim to offer a summary of chemical structure and properties, anti-cancer activities, drug delivery systems and combination therapy of GA, which might provide a reference to promote the development and clinical application of GA.
Collapse
Affiliation(s)
- Yuling Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yingchong Chen
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, People’s Republic of China
| | - Longfei Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Hui Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
9
|
Hatami E, Jaggi M, Chauhan SC, Yallapu MM. Gambogic acid: A shining natural compound to nanomedicine for cancer therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874:188381. [PMID: 32492470 DOI: 10.1016/j.bbcan.2020.188381] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023]
Abstract
The United States Food and Drug Administration has permitted number of therapeutic agents for cancer treatment. Most of them are expensive and have some degree of systemic toxicity which makes overbearing in clinical settings. Although advanced research continuously applied in cancer therapeutics, but drug resistance, metastasis, and recurrence remain unanswerable. These accounts to an urgent clinical need to discover natural compounds with precisely safe and highly efficient for the cancer prevention and cancer therapy. Gambogic acid (GA) is the principle bioactive and caged xanthone component, a brownish gamboge resin secreted from the of Garcinia hanburyi tree. This molecule showed a spectrum of biological and clinical benefits against various cancers. In this review, we document distinct biological characteristics of GA as a novel anti-cancer agent. This review also delineates specific molecular mechanism(s) of GA that are involved in anti-cancer, anti-metastasis, anti-angiogenesis, and chemo-/radiation sensitizer activities. Furthermore, recent evidence, development, and implementation of various nanoformulations of gambogic acid (nanomedicine) have been described.
Collapse
Affiliation(s)
- Elham Hatami
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.
| |
Collapse
|
10
|
Sang M, Han L, Luo R, Qu W, Zheng F, Zhang K, Liu F, Xue J, Liu W, Feng F. CD44 targeted redox-triggered self-assembly with magnetic enhanced EPR effects for effective amplification of gambogic acid to treat triple-negative breast cancer. Biomater Sci 2020; 8:212-223. [PMID: 31674634 DOI: 10.1039/c9bm01171d] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gambogic acid (GA) is a natural anti-tumor drug whose application is restricted by its poor aqueous solubility and inefficient bioavailability. Developing nanomaterials with excellent biocompatibility can amplify the therapeutic effects of GA. In this study, a tumor-targeted redox controllable self-assembled nano-system with magnetic enhanced EPR effects (mPEG-HA/CSO-SS-Hex/SPION/GA) was developed to improve the anticancer efficacy of GA. The nano-system is constituted by three layers: the outer layer is mono-aminated poly(ethylene glycol) grafted hyaluronic acid (mPEG-HA), which can target the CD44 receptor in breast cancer cells; the middle layer consists of disulfide linked hexadecanol (Hex) and chitosan oligosaccharide (CSO) to control the drug release by reduction response; the core layer is superparamagnetic iron oxide nanoparticles (SPION), which can enhance the EPR effect by magnetic guidance and contribute to GA entrapment. Different experiments were performed to characterize the complex self-assembly, and the cytotoxicity, pharmacokinetics, and in vivo antitumor activity of the self-assembly were investigated to evaluate its anti-tumor effects. The results revealed that mPEG-HA/CSO-SS-Hex/SPION/GA is an excellent nanosystem with appropriate size and sensitive responsiveness; it can accumulate in tumor sites and achieve excellent therapeutic effects on triple-negative breast cancer (TNBC). In summary, a CD44-targeted redox-triggered self-assembly nanosystem with magnetic enhanced EPR effects was developed for effective amplification of GA; it has potential to act as an effective carrier in drug delivery for chemotherapy of TNBC.
Collapse
Affiliation(s)
- Mangmang Sang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Stanković T, Dinić J, Podolski-Renić A, Musso L, Burić SS, Dallavalle S, Pešić M. Dual Inhibitors as a New Challenge for Cancer Multidrug Resistance Treatment. Curr Med Chem 2019; 26:6074-6106. [PMID: 29874992 DOI: 10.2174/0929867325666180607094856] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Dual-targeting in cancer treatment by a single drug is an unconventional approach in relation to drug combinations. The rationale for the development of dualtargeting agents is to overcome incomplete efficacy and drug resistance frequently present when applying individual targeting agents. Consequently, -a more favorable outcome of cancer treatment is expected with dual-targeting strategies. METHODS We reviewed the literature, concentrating on the association between clinically relevant and/or novel dual inhibitors with the potential to modulate multidrug resistant phenotype of cancer cells, particularly the activity of P-glycoprotein. A balanced analysis of content was performed to emphasize the most important findings and optimize the structure of this review. RESULTS Two-hundred and forty-five papers were included in the review. The introductory part was interpreted by 9 papers. Tyrosine kinase inhibitors' role in the inhibition of Pglycoprotein and chemosensitization was illustrated by 87 papers. The contribution of naturalbased compounds in overcoming multidrug resistance was reviewed using 92 papers, while specific dual inhibitors acting against microtubule assembling and/or topoisomerases were described with 55 papers. Eleven papers gave an insight into a novel and less explored approach with hybrid drugs. Their influence on P-glycoprotein and multidrug resistance was also evaluated. CONCLUSION These findings bring into focus rational anticancer strategies with dual-targeting agents. Most evaluated synthetic and natural drugs showed a great potential in chemosensitization. Further steps in this direction are needed for the optimization of anticancer treatment.
Collapse
Affiliation(s)
- Tijana Stanković
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Jelena Dinić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Loana Musso
- DeFENS, Department of Food, Environmental and Nutritional Sciences, Universita degli Studi di Milano, Milano, Italy
| | - Sonja Stojković Burić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| | - Sabrina Dallavalle
- DeFENS, Department of Food, Environmental and Nutritional Sciences, Universita degli Studi di Milano, Milano, Italy
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, Belgrade, Serbia
| |
Collapse
|
12
|
Sang MM, Liu FL, Wang Y, Luo RJ, Huan XX, Han LF, Zhang ZT, Feng F, Qu W, Liu W, Zheng F. A novel redox/pH dual-responsive and hyaluronic acid-decorated multifunctional magnetic complex micelle for targeted gambogic acid delivery for the treatment of triple negative breast cancer. Drug Deliv 2019; 25:1846-1857. [PMID: 30334478 PMCID: PMC6225507 DOI: 10.1080/10717544.2018.1486472] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gambogic acid (GA) is a naturally derived potent anticancer agent with extremely poor biocompatibility. In the present study, a novel of redox/pH dual-responsive multifunctional magnetic complex micelle (sPEG/HA/CSO-SS-Hex/Fe3O4/GA), which consisted of a reducible hexadecanol-modified chitosan oligosaccharide polymer micelle (CSO-SS-Hex) coated with hyaluronic acid (HA) and DCA grafted sheddable PEG-PLL (sPEG) copolymers and loaded with gambogic acid (GA) and Fe3O4 nanoparticles were developed for parenteral delivery for the treatment of triple negative breast cancer (TNBC). The ex vivo study showed that the sPEG shielded cationic HA/CSO-SS-Hex/Fe3O4/GA core at physiological pH but quickly shed off to re-expose the core due to its charge reversible property. The sPEG/HA/CSO-SS-Hex/Fe3O4/GA micelles effectively facilitated tumor-targeted GA delivery by HA, which is a targeting ligand for CD44 receptor of TNBC cells, meanwhile increase GA uptake at the acidic condition but diminished the drug uptake at neutral pH. The in vitro cellular uptake study and in vivo biodistribution and antitumor activity of the formulations were determined, all results showed that the complex micelle enhanced TNBC tumor cellular uptake and fast drug release due to the combined effect of magnet targeting, CD44 receptor-mediated internalization and redox/pH dual-responsive drug release. Hence, tumor-targeted delivery of GA with redox/pH dual-responsive multifunctional magnetic complex micelle sPEG/HA/CSO-SS-Hex/Fe3O4/GA might have potential implications for the chemotherapy of TNBC.
Collapse
Affiliation(s)
- Mang Mang Sang
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Fu Lei Liu
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yang Wang
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Ren Jie Luo
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Xiao Xian Huan
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Ling Fei Han
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Zhong Tao Zhang
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Feng Feng
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Wei Qu
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Wenyuan Liu
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Feng Zheng
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| |
Collapse
|
13
|
Wang Y, Wang X, Zhang J, Wang L, Ou C, Shu Y, Wu Q, Ma G, Gong C. Gambogic acid-encapsulated polymeric micelles improved therapeutic effects on pancreatic cancer. CHINESE CHEM LETT 2019; 30:885-888. [DOI: 10.1016/j.cclet.2019.02.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
14
|
Sang M, Han L, Luo R, Liu F, Wang Y, Qu W, Zheng F, Liu W, Feng F. WITHDRAWN: Magnetic and CD44 receptor dual targeting redox-responsive polymeric micelle for precise delivery of Gambogic acid to triple-negative breast cancer. Asian J Pharm Sci 2018. [DOI: 10.1016/j.ajps.2018.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
15
|
Huang R, Li J, Kebebe D, Wu Y, Zhang B, Liu Z. Cell penetrating peptides functionalized gambogic acid-nanostructured lipid carrier for cancer treatment. Drug Deliv 2018; 25:757-765. [PMID: 29528244 PMCID: PMC6058566 DOI: 10.1080/10717544.2018.1446474] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Tumor-targeted delivery is considered a crucial component of current anticancer drug development and is the best approach to increase the efficacy and reduce the toxicity. Nanomedicine, particularly ligand-based nanoparticles have shown a great potential for active targeting of tumor. Cell penetrating peptide is one of the promising ligands in a targeted cancer therapy. In this study, the gambogic acid-loaded nanostructured lipid carrier (GA-NLC) was modified with two kinds of cell penetrating peptides (cRGD and RGERPPR). The GA-NLC was prepared by emulsification and solvent evaporation method and coupled with cRGD, RGERPPR, and combination cRGD and RGERPPR to form GA-NLC-cRGD, GA-NLC-RGE, and GA-NLC-cRGD/RGE, respectively. The formulations were characterized by their particle size and morphology, zeta potential, encapsulation efficiency, and differential scanning calorimetry. In vitro cytotoxicity and cellular uptake study of the formulations were performed against breast cancer cell (MDA-MB-231). Furthermore, in vivo biodistribution and antitumor activity of the formulations were determined by in vivo imaging and in tumor-bearing nude mice, respectively. The result of in vitro cytotoxicity study showed that GA-NLC-RGE exhibited a significantly higher cytotoxicity on MDA-MB-231 as compared with GA-NLC and GA-Sol. Similarly, RGE-Cou-6-NLC showed remarkably higher uptake by the cells than other NLCs over the incubation period. The in vivo imaging study has demonstrated that among the formulations, the RGE-decorated DiR-NLC were more accumulated in the tumor site. The in vivo antitumor activity revealed that RGE-GA-NLC inhibits the tumor growth more efficiently than other formulations. In conclusion, RGERPPR has a potential as an effective carrier in targeting drug delivery of anticancer agents.
Collapse
MESH Headings
- Absorption, Physiological
- Animals
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/pharmacokinetics
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell-Penetrating Peptides/adverse effects
- Cell-Penetrating Peptides/chemistry
- Drug Carriers/administration & dosage
- Drug Carriers/pharmacokinetics
- Drug Carriers/pharmacology
- Drug Carriers/therapeutic use
- Drug Compounding
- Female
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Electron, Transmission
- Nanostructures/adverse effects
- Nanostructures/chemistry
- Nanostructures/ultrastructure
- Particle Size
- Random Allocation
- Surface Properties
- Tissue Distribution
- Tumor Burden/drug effects
- Xanthones/administration & dosage
- Xanthones/pharmacokinetics
- Xanthones/pharmacology
- Xanthones/therapeutic use
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Rui Huang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Jiawei Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Dereje Kebebe
- Tianjin State Key Laboratory of Modern Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- School of Pharmacy, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Yumei Wu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Bing Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Zhidong Liu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| |
Collapse
|
16
|
Abstract
Sirtuins (SIRT) are coenzyme NAD+-dependent histone deacetylases for the transfer of modified acetyl groups. Sirtuins are widely involved in various physiological processes and therefore associated with cardiovascular disease, diabetes, Parkinson's disease, cancer and beyond. Consequently, the development of modulators for sirtuins has considerable clinical value. To date, a variety of SIRT1/2 inhibitors have been reported and none has been approved for the market. This review summarizes the recent progress in the discovery and development of SIRT1/2 inhibitors including their inhibitory potency, structure–activity relationship and binding mode analysis as well as discusses the perspective for the future development of SIRT1/2 inhibitors.
Collapse
|
17
|
Banik K, Harsha C, Bordoloi D, Lalduhsaki Sailo B, Sethi G, Leong HC, Arfuso F, Mishra S, Wang L, Kumar AP, Kunnumakkara AB. Therapeutic potential of gambogic acid, a caged xanthone, to target cancer. Cancer Lett 2017; 416:75-86. [PMID: 29246645 DOI: 10.1016/j.canlet.2017.12.014] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/04/2017] [Accepted: 12/08/2017] [Indexed: 12/11/2022]
Abstract
Natural compounds have enormous biological and clinical activity against dreadful diseases such as cancer, as well as cardiovascular and neurodegenerative disorders. In spite of the widespread research carried out in the field of cancer therapeutics, cancer is one of the most prevalent diseases with no perfect treatment till date. Adverse side effects and the development of chemoresistance are the imperative limiting factors associated with conventional chemotherapeutics. For this reason, there is an urgent need to find compounds that are highly safe and efficacious for the prevention and treatment of cancer. Gambogic acid (GA) is a xanthone structure extracted from the dry, brownish gamboge resin secreted from the Garcinia hanburyi tree in Southeast Asia and has inherent anti-cancer properties. In this review, the molecular mechanisms underlying the targets of GA that are liable for its effective anti-cancer activity are discussed that reveal the potential of GA as a pertinent candidate that can be appropriately developed and designed into a capable anti-cancer drug.
Collapse
Affiliation(s)
- Kishore Banik
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India
| | - Choudhary Harsha
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India
| | - Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India
| | - Gautam Sethi
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, 700000, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 700000, Viet Nam; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Hin Chong Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia
| | - Srishti Mishra
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Alan P Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia; National University Cancer Institute, National University Health System, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam, 781039, India.
| |
Collapse
|
18
|
Ji Y, Shan S, He M, Chu CC. Inclusion complex from cyclodextrin-grafted hyaluronic acid and pseudo protein as biodegradable nano-delivery vehicle for gambogic acid. Acta Biomater 2017; 62:234-245. [PMID: 28859900 DOI: 10.1016/j.actbio.2017.08.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 01/26/2023]
Abstract
β-Cyclodextrin can form inclusion complex with a series of guest molecules including phenyl moieties, and has gained considerable popularity in the study of supramolecular nanostructure. In this study, a biodegradable nanocomplex (HA(CD)-4Phe4 nanocomplex) was developed from β-cyclodextrin grafted hyaluronic acid (HA) and phenylalanine based poly(ester amide). The phenylalanine based poly(ester amide) is a biodegradable pseudo protein which provides the encapsulation capacity for gambogic acid (GA), a naturally-derived chemotherapeutic which has been effectively employed to treat multidrug resistant tumor. The therapeutic potency of free GA is limited due to its poor solubility in water and the lack of tumor-selective toxicity. The nanocomplex carrier enhanced the solubility and availability of GA in aqueous media, and the HA component enabled the targeted delivery to tumor cells with overexpression of CD44 receptors. In the presence of hyaluronidase, the release of GA from the nanocomplex was significantly accelerated, due to the enzymatic biodegradation of the carrier. Compared to free GA, GA-loaded nanocomplex exhibited improved cytotoxicity in MDA-MB-435/MDR multidrug resistant melanoma cells, and induced enhanced level of apoptosis and mitochondrial depolarization, at low concentration of GA (1-2µM). The nanocomplex enhanced the therapeutic potency of GA, especially when diluted in physiological environment. In addition, suppressed matrix metalloproteinase activity was also detected in MDA-MB-435/MDR cells treated by GA-loaded nanocomplex, which demonstrated its potency in the inhibition of tumor metastasis. The in vitro data suggested that HA(CD)-4Phe4 nanocomplex could provide a promising alternative in the treatment of multidrug resistant tumor cells. STATEMENT OF SIGNIFICANCE Gambogic acid (GA), naturally derived from genus Garcinia trees, exhibited significant cytotoxic activity against multiple types of tumors with resistance to traditional chemotherapeutics. Unfortunately, the poor solubility of GA in conventional pharmaceutical solvents and non-targeted distribution in normal tissues greatly limited its therapeutic potency. To overcome the challenges, we develop a nanoplatform from the supramolecular assembly of β-cyclodextrin grafted hyaluronic acid (HA) and phenylalanine based pseudo protein. The pseudo protein in the nanocomplex provided the hydrophobic interaction and loading capacity for GA, while the HA component targeted the overexpressed CD44 receptor and improved the selective endocytosis in multidrug resistant melanoma cells. The supramolecular nanocomplex provide a promising platform for the delivery of hydrophobic chemotherapeutics to improve the bioavailability and efficiency.
Collapse
|
19
|
Gambogic Acid Induces Cell Apoptosis and Inhibits MAPK Pathway in PTEN -/-/p53 -/- Prostate Cancer Cells In Vitro and Ex Vivo. Chin J Integr Med 2017; 24:109-116. [PMID: 28578487 DOI: 10.1007/s11655-017-2410-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To investigate the effect of gambogic acid (GA) on the growth and cell death of castrate resistant prostate cancer (PC) with phosphate and tension homology (PTEN) and p53 genes deleted in vitro and ex vivo, and elucidate the underlying possible molecular mechanisms. METHODS PTEN-/-/p53-/- PC cells and Los Angeles prostate cancer-4 (LAPC-4) cells were treated with GA for 24 h and 48 h, then cell viability was determined by cell proliferation assay. PTEN-/-/p53-/- PC cells organoids number was calculated under GA treatment for 1 week. In addition, cell titer glo assay was performed to analyze 3 dimensional cell viability of patients derived xenografts (PDX) 170.2 organoids. Flow cytometry was used to detect apoptotic cells treated with GA. And confocal image was performed to detect the apoptotic mitochondrial morphological changes. Apoptotic cell death related protein levels were measured through Western blot (WB) in GA treated cells and organoids. The expression levels of mitogen-activated protein kinases (MAPKs) pathway related ribonucleic acid (RNAs) and proteins were analyzed by reverse transcription polymerase chain reaction (RT-PCR) and WB, respectively. RESULTS The treatment of GA significantly reduced cell viability of PTEN-/-/p53-/- PC cells and LAPC-4 in a time- and concentration-dependent manner. In organoids, GA showed strong inhibition towards organoids' numbers and diameters and continuously led to a complete organoids inhibition with GA 150 nmol/L. Ex vivo results validated that GA 1 μmol/L inhibited 44.6% PDX170.2 organoids growth. As for mechanism, flow cytometry detected continuously increased apoptotic portion under GA treatment from 1.98% to 11.78% (6 h) and 29.94% (8 h, P<0.05). In addition, mitochondrial fragmentation emerged in GA treated cells indicated the mitochondrial apoptotic pathway might be involved. Furthermore, WB detected caspases-3, -9 activation and light chain (LC)-3 conversion with GA treatment. WB revealed decreased activity of MAPK pathway and down-regulation of downstream c-fos oncogene RNA level was detected by RT-PCR before undergoing apoptosis (P<0.05). CONCLUSION GA was a potent anti-tumor compound as for PTEN-/-/p53-/- PC, which contributed to cell apoptosis via inhibition of the MAPK pathway and c-fos.
Collapse
|
20
|
Kavitha N, Ein Oon C, Chen Y, Kanwar JR, Sasidharan S. Phaleria macrocarpa (Boerl.) fruit induce G 0/G 1 and G 2/M cell cycle arrest and apoptosis through mitochondria-mediated pathway in MDA-MB-231 human breast cancer cell. JOURNAL OF ETHNOPHARMACOLOGY 2017; 201:42-55. [PMID: 28263848 DOI: 10.1016/j.jep.2017.02.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/22/2017] [Accepted: 02/25/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phaleria macrocarpa (Scheff) Boerl, is a well-known folk medicinal plant in Indonesia. Traditionally, P. macrocarpa has been used to control cancer, impotency, hemorrhoids, diabetes mellitus, allergies, liver and hearth disease, kidney disorders, blood diseases, acne, stroke, migraine, and various skin diseases. AIM OF THE STUDY The purpose of this study was to determine the in situ cytotoxicity effect P. macrocarpa fruit ethyl acetate fraction (PMEAF) and the underlying molecular mechanism of cell death. MATERIALS AND METHODS MDA-MB-231 cells were incubated with PMEAF for 24h. Cell cycle and viability were examined using flow cytometry analysis. Apoptosis was determined using the Annexin V assay and also by fluorescence microscopy. Apoptosis protein profiling was detected by RayBio® Human Apoptosis Array. RESULTS The AO/PI staining and flow cytometric analysis of MDA-MB-231 cells treated with PMEAF were showed apoptotic cell death. The cell cycle analysis by flow cytometry analysis revealed that the accumulation of PMEAF treated MDA-MB-231 cells in G0/G1 and G2/M-phase of the cell cycle. Moreover, the PMEAF exert cytotoxicity by increased the ROS production in MDA-MB-231 cells consistently stimulated the loss of mitochondrial membrane potential (∆Ψm) and induced apoptosis cell death by activation of numerous signalling proteins. The results from apoptosis protein profiling array evidenced that PMEAF stimulated the expression of 9 pro-apoptotic proteins (Bax, Bid, caspase 3, caspase 8, cytochrome c, p21, p27, p53 and SMAC) and suppressed the 4 anti-apoptotic proteins (Bcl-2, Bcl-w, XIAP and survivin) in MDA-MB-231 cells. CONCLUSION The results indicated that PMEAF treatment induced apoptosis in MDA-MB-231 cells through intrinsic mitochondrial related pathway with the participation of pro and anti-apoptotic proteins, caspases, G0/G1 and G2/M-phases cell cycle arrest by p53-mediated mechanism.
Collapse
Affiliation(s)
- Nowroji Kavitha
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800 Pulau Pinang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800 Pulau Pinang, Malaysia
| | - Yeng Chen
- Dental Research & Training Unit, and Oral Cancer Research and Coordinating Centre (OCRCC), Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Jagat R Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research (LIMBR), School of Medicine (SoM), Faculty of Health, Institute for Frontier Materials (IFM), Deakin University, Waurn Ponds, VIC 3217 Australia
| | - Sreenivasan Sasidharan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800 Pulau Pinang, Malaysia.
| |
Collapse
|
21
|
Foggetti G, Ottaggio L, Russo D, Monti P, Degan P, Fronza G, Menichini P. Gambogic acid counteracts mutant p53 stability by inducing autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:382-392. [PMID: 27899303 DOI: 10.1016/j.bbamcr.2016.11.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 11/21/2016] [Accepted: 11/24/2016] [Indexed: 12/19/2022]
Abstract
Mutant p53 (mutp53) proteins are frequently present at higher levels than the wild-type (wt) protein in tumors, and some of them can acquire oncogenic properties. Consistently, knockdown of mutp53 protein in human cancer cell lines leads to reduced cell proliferation and invasion as well as to an increased sensitivity to some anticancer drugs. Therefore, the exploitation of cellular pathways and/or molecules that promote mutp53 degradation may have a therapeutic interest. Recently, autophagy is emerging as an important pathway involved in the stability of mutp53. In this paper, we explored the autophagic potential of gambogic acid (GA), a molecule that stimulates the degradation of mutp53 and increases the sensitivity of cancer cells to chemotherapeutic agents. We demonstrated that GA may induce mutp53 degradation through autophagy in cancer cells expressing the p53-R280K (MDA-MB-231) and the p53-S241F (DLD1) proteins. The inhibition of autophagy with bafilomycin A1 or chloroquine counteracted mutp53 degradation by GA. However, the autophagy induction and mutp53 degradation affected cell survival and proliferation only at low GA concentrations. At higher GA concentrations, when cells undergo massive apoptosis, autophagy is no longer detectable by immuno-fluorescence analysis. We concluded that autophagy is a relevant pathway for mutp53 degradation in cancer cells but it contributes only partially to GA-induced cell death, in a time and dose-dependent manner.
Collapse
Affiliation(s)
- Giorgia Foggetti
- U.O.C. Mutagenesi, IRCCS AOU San Martino-IST, 16132 Genova, Italy
| | - Laura Ottaggio
- U.O.C. Mutagenesi, IRCCS AOU San Martino-IST, 16132 Genova, Italy
| | - Debora Russo
- U.O.C. Mutagenesi, IRCCS AOU San Martino-IST, 16132 Genova, Italy
| | - Paola Monti
- U.O.C. Mutagenesi, IRCCS AOU San Martino-IST, 16132 Genova, Italy
| | - Paolo Degan
- U.O.C. Mutagenesi, IRCCS AOU San Martino-IST, 16132 Genova, Italy
| | - Gilberto Fronza
- U.O.C. Mutagenesi, IRCCS AOU San Martino-IST, 16132 Genova, Italy
| | - Paola Menichini
- U.O.C. Mutagenesi, IRCCS AOU San Martino-IST, 16132 Genova, Italy.
| |
Collapse
|
22
|
Molecular targets of gambogic acid in cancer: recent trends and advancements. Tumour Biol 2016; 37:12915-12925. [PMID: 27448303 DOI: 10.1007/s13277-016-5194-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/13/2016] [Indexed: 12/11/2022] Open
|
23
|
Doddapaneni R, Patel K, Owaid IH, Singh M. Tumor neovasculature-targeted cationic PEGylated liposomes of gambogic acid for the treatment of triple-negative breast cancer. Drug Deliv 2016; 23:1232-41. [PMID: 26701717 PMCID: PMC5024788 DOI: 10.3109/10717544.2015.1124472] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/22/2015] [Indexed: 11/13/2022] Open
Abstract
Gambogic acid (GA) is a naturally derived potent anticancer agent with extremely poor aqueous solubility. In the present study, positively charged PEGylated liposomal formulation of GA (GAL) was developed for parenteral delivery for the treatment of triple-negative breast cancer (TNBC). The GAL was formulated with a particle size of 107.3 ± 10.6 nm with +32 mV zeta potential. GAL showed very minimal release of GA over 24 h period confirming the non-leakiness and stability of liposomes. In vitro cytotoxicity assays showed similar cell killing with GA and GAL against MDA-MB-231 cells but significantly higher inhibition of HUVEC growth was observed with GAL. Furthermore, GAL significantly (p < 0.05) inhibited the MDA-MB-231 orthotopic xenograft tumor growth with >50% reduction of tumor volume and reduction in tumor weight by 1.7-fold and 2.2-fold when compared to GA and controls, respectively. Results of western blot analysis indicated that GAL significantly suppressed the expression of apoptotic markers, bcl2, cyclinD1, survivin and microvessel density marker-CD31 and increased the expression of p53 and Bax compared to GA and control. Collectively, these data provide further support for the potential applications of cationic GAL in its intravenous delivery and its significant role in inhibiting angiogenesis against TNBC.
Collapse
Affiliation(s)
- Ravi Doddapaneni
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ketan Patel
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ibtisam Hasan Owaid
- John D. Dingell VA Medical Center, 4646 John R. Street, Detroit, MI 48201, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
24
|
Kumar A, Chauhan S. How much successful are the medicinal chemists in modulation of SIRT1: A critical review. Eur J Med Chem 2016; 119:45-69. [PMID: 27153347 DOI: 10.1016/j.ejmech.2016.04.063] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/14/2016] [Accepted: 04/25/2016] [Indexed: 12/27/2022]
Abstract
Silent information regulator two homologue one (SIRT1) is the most widely studied member of the sirtuin family related to histone deacetylases class III super-family using nicotinamide adenine dinucleotide (NAD(+)) as its cofactor. It is located in the nucleus but also modulates the targets in cytoplasm and mainly acts as transacetylase rather than deacetylase. SIRT1 specifically cleaves the nicotinamide ribosyl bond of NAD(+) and transfers the acetyl group from proteins to their co-substrate through an ADP- ribose-peptidyl imidate intermediate. It has been indicated that SIRT1 and its histone as well as non histone targets are involved in a wide range of biological courses including metabolic diseases, age related diseases, viral infection, inflammation, tumor-cell growth and metastasis. Modulation of SIRT1 expression may present a new insight in the discovery of a number of therapeutics. This review summarizes studies about SIRT1 and mainly focuses on the various modulators of SIRT1 evolved by natural as well as synthetic means.
Collapse
Affiliation(s)
- Ashwani Kumar
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India.
| | - Shilpi Chauhan
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India
| |
Collapse
|
25
|
Liu WY, Wu XU, Liao CQ, Shen J, Li J. Apoptotic effect of gambogic acid in esophageal squamous cell carcinoma cells via suppression of the NF-κB pathway. Oncol Lett 2016; 11:3681-3685. [PMID: 27284372 PMCID: PMC4887765 DOI: 10.3892/ol.2016.4437] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 03/09/2016] [Indexed: 02/06/2023] Open
Abstract
Despite extensive investigations of therapeutic improvements for surgical techniques, chemotherapy and chemoradiotherapy, esophageal squamous cell carcinoma (ESCC) remains one of the most aggressive forms of cancer, and the prognosis for patients with advanced ESCC remains poor. Therefore, effective therapies are urgently required in order to improve the prognosis of patients with ESCC. TE-1 cells were treated with gambogic acid (GA), and then subjected to western blot analysis, TUNEL assay and caspase activity analysis. GA significantly induced apoptosis in ESCC TE-1 cells. In addition, the antitumor activity of GA was accompanied by the decreased expression of phosphorylated-protein kinase B (p-AKT) and nuclear factor of κ light polypeptide gene enhancer in B-cells 1 (NF-κB). The inhibition of protein kinase B (AKT) and NF-κB activation by chemical inhibitors augmented the apoptotic effect responses to GA in the TE-1 cells. The pan-caspase inhibitor z-VAD-fmk (zVAD) decreased GA-induced apoptosis. Furthermore, zVAD attenuated GA-induced growth inhibition in TE-1 cells. GA induced apoptosis in ESCC TE-1 via suppression of NF-κB pathway. The findings of the present study may provide a novel insight into ESCC treatment.
Collapse
Affiliation(s)
- Wen-Yue Liu
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Department of Thoracic Surgery, Yuebei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - X U Wu
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Cheng-Quan Liao
- Department of Cardiovascular Surgery, Yuebei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Jie Shen
- Department of Medical Records, Yuebei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Jun Li
- Department of Thoracic Surgery, Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
26
|
Gambogic Acid and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 928:375-395. [DOI: 10.1007/978-3-319-41334-1_15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
27
|
Jia B, Li S, Hu X, Zhu G, Chen W. Recent research on bioactive xanthones from natural medicine: Garcinia hanburyi. AAPS PharmSciTech 2015; 16:742-58. [PMID: 26152816 PMCID: PMC4508296 DOI: 10.1208/s12249-015-0339-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/18/2015] [Indexed: 12/24/2022] Open
Abstract
Garcinia hanburyi, a tropical plant found in south Asia, has a special long history in the development of both medicine and art. This review mainly focuses on the pharmacy research of the bioactive compounds from the plant in recent years. Preparative and analysis separation methods were introduced. Moreover, the chemical structure of the isolated compounds was included. The studies of biological activities of the caged xanthones from the plant, including antitumor, anti-HIV-1, antibacterial, and neurotrophic activities, were reviewed in detail. Furthermore, the mechanisms of its antitumor activity were also reviewed. As mentioned above, some of the xanthones from G. hanburyi can be promising drug candidates, which is worth studying. However, we still need much evidence to prove their efficacy and safety. So, further research is critical for the future application of xanthones from G. hanburyi.
Collapse
Affiliation(s)
- Buyun Jia
- />School of Pharmacy, Anhui University of Chinese Medicine, 001 Qianjiang Road, Hefei, 230012 Anhui China
| | - Shanshan Li
- />School of Pharmacy, Anhui University of Chinese Medicine, 001 Qianjiang Road, Hefei, 230012 Anhui China
| | - Xuerui Hu
- />School of Pharmacy, Anhui University of Chinese Medicine, 001 Qianjiang Road, Hefei, 230012 Anhui China
| | - Guangyu Zhu
- />School of Pharmacy, Anhui University of Chinese Medicine, 001 Qianjiang Road, Hefei, 230012 Anhui China
- />Ma’anshan Central Hospital, 027 Hudong Road, Ma’anshan, 243000 Anhui China
| | - Weidong Chen
- />School of Pharmacy, Anhui University of Chinese Medicine, 001 Qianjiang Road, Hefei, 230012 Anhui China
| |
Collapse
|
28
|
Huang GM, Sun Y, Ge X, Wan X, Li CB. Gambogic acid induces apoptosis and inhibits colorectal tumor growth via mitochondrial pathways. World J Gastroenterol 2015; 21:6194-6205. [PMID: 26034354 PMCID: PMC4445096 DOI: 10.3748/wjg.v21.i20.6194] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/22/2014] [Accepted: 01/30/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of gambogic acid (GA) on apoptosis in the HT-29 human colon cancer cell line.
METHODS: H-29 cells were used for in vitro experiments in this study. Relative cell viability was assessed using MTT assays. Cell apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling and Hoechst 33342 staining, and quantified by flow cytometry. Cellular ultrastructure was observed by transmission electron microscopy. Real-time PCR and Western blot analyses were used to evaluate gene and protein expression levels. For in vivo experiments, BALB/c nude mice received subcutaneous injections of HT-29 cells in the right armpit. When well-established xenografts were palpable with a tumor size of 75 mm3, mice were randomly assigned to a vehicle (negative) control, positive control or GA treatment group (n = 6 each). The animals in the treatment group received one of three dosages of GA (in saline; 5, 10 or 20 mg/kg) via the caudal vein twice weekly, whereas animals in the negative and positive control groups were given equal volumes of 0.9% saline or 10 mg/kg docetaxel, respectively, via the caudal vein once weekly.
RESULTS: The cell viability assay showed that GA inhibited proliferation of HT-29 cells in a dose- and time-dependent manner after treatment with GA (0.00, 0.31, 0.62, 1.25, 2.50, 5.00 or 10.00 μmol/L) for 24, 48 or 72 h. After 48 h, the percentage of apoptotic cells in cells treated with 0.00, 1.25, 2.50 and 5.00 μmol/L GA was 1.4% ± 0.3%, 9.8% ± 1.2%, 25.7% ± 3.3% and 49.3% ± 5.8%, respectively. Ultrastructural analysis of HT-29 cells treated for 48 h with 2.5μmol/L GA revealed apoptotic bodies and condensed and fragmented nuclei. Levels of caspase-8, -9 and -3 mRNAs were significantly increased after treatment with GA (1.25, 2.50 or 5.00 μmol/L) for 48 h (P < 0.05 for all). Protein levels of apoptosis-related factors Fas, FasL, FADD, cytochrome c, and Apaf-1 were increased in GA-treated cells, whereas levels of pro-caspase-8, -9 and -3 were significantly decreased (P < 0.05 for all). Furthermore, GA significantly and dose-dependently inhibited the growth of HT-29 tumors in a mouse xenograft model (P < 0.05).
CONCLUSION: GA inhibits HT-29 proliferation via induction of apoptosis. The anti-cancer effects are likely mediated by death receptor (extrinsic) and mitochondrial (intrinsic) pathways.
Collapse
|
29
|
Wu Z, Liu B, E C, Liu J, Zhang Q, Liu J, Chen N, Chen R, Zhu R. Resveratrol inhibits the proliferation of human melanoma cells by inducing G1/S cell cycle arrest and apoptosis. Mol Med Rep 2014; 11:400-4. [PMID: 25333673 DOI: 10.3892/mmr.2014.2716] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 07/22/2014] [Indexed: 11/06/2022] Open
Abstract
Resveratrol (Res), a natural plant extract, is an effective inducer of cell apoptosis and cell cycle arrest in multiple carcinoma cell types, which has been demonstrated by its ability to inhibit the proliferation of multiple human tumor cells in vitro. Although Res possesses chemopreventive properties against several types of cancer, the molecular mechanism underlying its anticancer activity remains to be fully elucidated. The present study demonstrated that Res induced cell cycle arrest and inhibited the proliferation of human melanoma A375 (IC50=23 µM after 48 h; P<0.05) and SK-MEL-31 (IC50=15 µM after 48 h; P<0.05) cells. Western blot analysis demonstrated that Res induced the apoptosis of human melanoma A375 and SK-MEL-31 cells by upregulating the expression of Bcl-2-associated X protein and B-cell lymphoma 2, possibly via the p53 pathway and activation of caspase-9 and caspase-3.
Collapse
Affiliation(s)
- Zhiyuan Wu
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Bin Liu
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Cailing E
- Department of Dermatology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Jie Liu
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Qingyu Zhang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Juntao Liu
- The Second Department of Orthopedics, Ningde Hospital, Affiliated Hospital of Fujian Medical University, Ningde, Fujian 352100, P.R. China
| | - Nianping Chen
- Laboratory of Hepatobiliary Surgery, Guangdong Medical College, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| | - Rongyi Chen
- Department of Dermatology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Runzhi Zhu
- Laboratory of Hepatobiliary Surgery, Guangdong Medical College, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
30
|
Li H, Chen Z, Zhou S. Apoptosis in glioma-bearing rats after neural stem cell transplantation. Neural Regen Res 2014; 8:1793-802. [PMID: 25206476 PMCID: PMC4145955 DOI: 10.3969/j.issn.1673-5374.2013.19.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/03/2013] [Indexed: 12/22/2022] Open
Abstract
Abnormal activation of the Ras/Raf/Mek/Erk signaling cascade plays an important role in glioma. Inhibition of this aberrant activity could effectively hinder glioma cell proliferation and promote cell apoptosis. To investigate the mechanism of glioblastoma treatment by neural stem cell transplantation with respect to the Ras/Raf/Mek/Erk pathway, C6 glioma cells were prepared in suspension and then infused into the rat brain to establish a glioblastoma model. Neural stem cells isolated from fetal rats were then injected into the brain of this glioblastoma model. Results showed that Raf-1, Erk and Bcl-2 protein expression significantly increased, while Caspase-3 protein expression decreased. After transplantation of neural stem cells, Raf-1, Erk and Bcl-2 protein expression significantly decreased, while Caspase-3 protein expression significantly increased. Our findings indicate that transplantation of neural stem cells may promote apoptosis of glioma cells by inhibiting Ras/Raf/Mek/Erk signaling, and thus may represent a novel treatment approach for glioblastoma.
Collapse
Affiliation(s)
- Hua Li
- Department of Neurology, the 476 Hospital of Chinese PLA, Fuzhou 350002, Fujian Province, China
| | - Zhenjun Chen
- Department of Neurology, the 476 Hospital of Chinese PLA, Fuzhou 350002, Fujian Province, China
| | - Shaopeng Zhou
- Department of Anesthesiology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| |
Collapse
|
31
|
Kapoor S. Gambogic Acid and Inhibition of Tumor Growth: A Close Association. Cancer Biother Radiopharm 2014; 29:265. [DOI: 10.1089/cbr.2012.1432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
32
|
Pandey MK, Kale VP, Song C, Sung SS, Sharma AK, Talamo G, Dovat S, Amin SG. Gambogic acid inhibits multiple myeloma mediated osteoclastogenesis through suppression of chemokine receptor CXCR4 signaling pathways. Exp Hematol 2014; 42:883-96. [PMID: 25034231 DOI: 10.1016/j.exphem.2014.07.261] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 06/11/2014] [Accepted: 07/05/2014] [Indexed: 11/17/2022]
Abstract
Bone disease, characterized by the presence of lytic lesions and osteoporosis is the hallmark of multiple myeloma (MM). Stromal cell-derived factor 1α (SDF-1α) and its receptor, CXC chemokine receptor 4 (CXCR4), has been implicated as a regulator of bone resorption, suggesting that agents that can suppress SDF1α/CXCR4 signaling might inhibit osteoclastogenesis, a process closely linked to bone resorption. We, therefore, investigated whether gambogic acid (GA), a xanthone, could inhibit CXCR4 signaling and suppress osteoclastogenesis induced by MM cells. Through docking studies we predicted that GA directly interacts with CXCR4. This xanthone down-regulates the expression of CXCR4 on MM cells in a dose- and time-dependent manner. The down-regulation of CXCR4 was not due to proteolytic degradation, but rather GA suppresses CXCR4 mRNA expression by inhibiting nuclear factor-kappa B (NF-κB) DNA binding. This was further confirmed by quantitative chromatin immunoprecipitation assay, as GA inhibits p65 binding at the CXCR4 promoter. GA suppressed SDF-1α-induced chemotaxis of MM cells and downstream signaling of CXCR4 by inhibiting phosphorylation of Akt, p38, and Erk1/2 in MM cells. GA abrogated the RANKL-induced differentiation of macrophages to osteoclasts in a dose- and time-dependent manner. In addition, we found that MM cells induced differentiation of macrophages to osteoclasts, and that GA suppressed this process. Importantly, suppression of osteoclastogenesis by GA was mediated through IL-6 inhibition. Overall, our results show that GA is a novel inhibitor of CXCR4 expression and has a strong potential to suppress osteoclastogenesis mediated by MM cells.
Collapse
Affiliation(s)
- Manoj K Pandey
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA.
| | - Vijay P Kale
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Chunhua Song
- Division of Pediatric Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Shen-shu Sung
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Arun K Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Giampaolo Talamo
- Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA
| | - Sinisa Dovat
- Division of Pediatric Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA
| | - Shantu G Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA
| |
Collapse
|
33
|
Capilliposide Isolated from Lysimachia capillipes Hemsl. Induces ROS Generation, Cell Cycle Arrest, and Apoptosis in Human Nonsmall Cell Lung Cancer Cell Lines. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:497456. [PMID: 24523821 PMCID: PMC3910464 DOI: 10.1155/2014/497456] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/10/2013] [Indexed: 12/24/2022]
Abstract
Several data has reported that capilliposide, extracted from a traditional Chinese medicine, Lysimachia capillipes Hemsl. (LC) could exhibit inhibitory effect on cell proliferation in various cancers. The current study investigated the antitumor efficacy of Capilliposide and elucidated its potential molecular mechanism involved in vivo and vitro. Our results indicated that LC capilliposide inhibited proliferation of lung cancer cells in a dose-dependent manner. LC capilliposide induced cell cycle arrest at the S stage and enhanced apoptosis in NSCLC cells. Treatment with LC capilliposide increased the intracellular level of ROS, which activated the mitochondrial apoptotic pathway. Blockage of ROS by NAC highly reversed the effect of LC capilliposide on apoptosis. Xenograft tumor growth was significantly lower in the LC-treated group compared with the untreated control group (P < 0.05). The results also show that LC treatment does not produce any overt signs of acute toxicity in vivo. These findings demonstrate that LC capilliposide could exert an anti-tumor effect on NSCLC through mitochondrial-mediated apoptotic pathway and the activation of ROS is involved.
Collapse
|
34
|
Wu S, Liu B, Zhang Q, Liu J, Zhou W, Wang C, Li M, Bao S, Zhu R. Dihydromyricetin reduced Bcl-2 expression via p53 in human hepatoma HepG2 cells. PLoS One 2013; 8:e76886. [PMID: 24223706 PMCID: PMC3817187 DOI: 10.1371/journal.pone.0076886] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/27/2013] [Indexed: 12/16/2022] Open
Abstract
Dihydromyricetin (DHM) is a major active ingredient of flavonoids compounds. It exhibited anticancer activity and induced apoptosis in human hepatocellular carcinoma HepG2 cells according to our previous data. In this study, we investigated whether p53 is involved in DHM-triggered viability inhibition and apoptosis induction in cancer cells. MTT [3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide] assay was employed to evaluate the viability of HepG2 cells after DHM treatment. Meanwhile, p53 small interfering RNA (siRNA) was adopted to silence p53 expression. Protein level of p53 and Bax/Bcl-2 were evaluated by western blot analysis. Cell counting assay showed that DHM inhibited HepG2 cell growth effectively in a time- and dose-dependent manner. P53 expression was significantly increased after DHM treatment, whereas Bcl-2 was reduced potently. Furthermore, after co-treatment with Pifithrin-α (PFT-α, p53 inhibitor), Bcl-2 expression was reversed. The expression of Bax was no significant change, which was also observed after p53 silence. These findings defined and supported a novel function that DHM could induce human hepatocellular carcinoma HepG2 cells apoptosis by up-regulating Bax/Bcl-2 expression via p53 signal pathway.
Collapse
Affiliation(s)
- Shixing Wu
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Bin Liu
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Qingyu Zhang
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Jie Liu
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Wei Zhou
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Chang Wang
- Emergency Department, Ningde Hospital, Affiliated Hospital of Fujian Medical University, Ningde, China
| | - Mingyi Li
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
- * E-mail: (ML); (SB); (RZ)
| | - Shiting Bao
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
- * E-mail: (ML); (SB); (RZ)
| | - Runzhi Zhu
- Laboratory of Regenerative Medicine, Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
- * E-mail: (ML); (SB); (RZ)
| |
Collapse
|
35
|
Xu P, Li J, Shi L, Selke M, Chen B, Wang X. Synergetic effect of functional cadmium-tellurium quantum dots conjugated with gambogic acid for HepG2 cell-labeling and proliferation inhibition. Int J Nanomedicine 2013; 8:3729-36. [PMID: 24109183 PMCID: PMC3792847 DOI: 10.2147/ijn.s51622] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We prepared and studied novel fluorescent nanocomposites based on gambogic acid (GA) and cadmium–tellurium (CdTe) quantum dots (CdTe QDs) modified with cysteamine for purpose of cancer cell labeling and combined treatment. The nanocomposites were denoted as GA-CdTe. Characterization results indicated that the CdTe QDs can readily bind onto cell plasma membranes and then be internalized into cancer cells for real-time labeling and tracing of human liver hepatocellular carcinoma cell line (HepG2) cells. GA-CdTe significantly enhanced drug accumulation in HepG2 cells and inhibited cancer cell proliferation. GA-CdTe nanocomposites also improved the drug action of GA molecules in HepG2 cells and induced the G2/M phase arrest of the cancer cell cycle, promoting cell apoptosis. Given the sensitive, pH-triggered release of GA-CdTe, the side effects of GA anticancer agents on normal cells/tissues in the blood circulation markedly decreased. Efficient drug release and accumulation in target tumor cells were also facilitated. Thus, the fluorescent GA-CdTe offered a new strategy for potential multimode cancer therapy and provided new channels for research into naturally-active compounds extracted from traditional Chinese medicinal plants.
Collapse
Affiliation(s)
- Peipei Xu
- Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
36
|
Yu F, He C, Waddad AY, Munyendo WLL, Lv H, Zhou J, Zhang Q. N-octyl-N-arginine-chitosan (OACS) micelles for gambogic acid oral delivery: preparation, characterization and its study on in situ intestinal perfusion. Drug Dev Ind Pharm 2013; 40:774-82. [PMID: 23679668 DOI: 10.3109/03639045.2013.786723] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CONTEXT Gambogic acid (GA) can inhibit the growth of various cancer cells. However, the low bioavailability caused by insolubility, limits its clinical application. L-arginine is always used with GA to form a complex to obtain the higher solubility. Moreover, guanidyl group from arginine, which can facilitate the cellular uptake, was identified. OBJECTIVE In this study, L-arginine and chitosan (CS) were used for the first time to prepare N-octyl-N-arginine CS (OACS), a novel amphiphilic carrier for GA with solubility- and absorption-enhancing functions; the characterization of the GA loaded OACS micelles (GA-OACS) and its absorption-enhancing effect were also investigated. MATERIALS AND METHODS GA-OACS were prepared by the dialysis method. The formed micelles were characterized and evaluated by atomic force microscope (AFM), dynamic light scattering, differential scanning calorimeter (DSC), solubility test, in vitro release and in situ intestinal perfusion. RESULTS The GA-OACS micelles were successfully prepared attaining a 35.3% drug loading and 82.2% entrapment efficiency. GA-OACS had a homogeneous particle size of 160.3 nm; +21.8 mv zeta potential with smooth continuous surface was observed by using AFM. DSC diagram suggested that GA was encapsulated in the micelles. Meanwhile, GA encapsulated in micelles exhibited a desirable slow release in vitro experiment. The solubility of GA in OACS micelles was increased up to 3.16 ± 0.13 mg/mL, 2320 times than that of free GA. The single pass perfusion showed that the absorption of GA-OACS micelles was enhanced 3.6-fold, 2.1-fold and 2.2-fold for jejunum, ileum and colon, respectively. DISCUSSION AND CONCLUSION OACS provided excellent ability of drug loading, increasing solubility and enhanced absorption for GA, which indicated that OACS micelles as an oral drug delivery carrier may have potential research and application values.
Collapse
Affiliation(s)
- Fan Yu
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University , Nanjing , China
| | | | | | | | | | | | | |
Collapse
|
37
|
Xu J, Zhou M, Ouyang J, Wang J, Zhang Q, Xu Y, Xu Y, Zhang Q, Xu X, Zeng H. Gambogic acid induces mitochondria-dependent apoptosis by modulation of Bcl-2 and Bax in mantle cell lymphoma JeKo-1 cells. Chin J Cancer Res 2013; 25:183-91. [PMID: 23592899 DOI: 10.3978/j.issn.1000-9604.2013.02.06] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 02/21/2013] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To study the mechanisms in gambogic acid (GA) -induced JeKo-1 human Mantle Cell Lymphoma cell apoptosis in vitro. METHODS The proliferation of GA-treated JeKo-1 cells was measured by CCK-8 assay and Ki-67 immunocytochemical detection. Apoptosis, cell cycle and mitochondrial membrane potential were measured by flow cytometric analysis. Caspase-3, -8 and -9 were detected by colorimetric assay. Bcl-2 and Bax were analyzed by Western blotting. RESULTS GA inhibited cell growth in a time- and dose- dependent manner. GA induces apoptosis in JeKo-1 cells but not in normal bone marrow cells, which was involved in reducing the membrane potential of mitochondria, activating caspases-3, -8 and -9 and decreasing the ratio of Bcl-2 and Bax without cell cycle arresting. CONCLUSIONS GA induced apoptosis in human MCL JeKo-1 cells by regulating Bcl-2/Bax and activating caspase-3, -8 and -9 via mitochondrial pathway without affecting cell cycle.
Collapse
Affiliation(s)
- Jingyan Xu
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210038, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhang ZH, Wang XP, Ayman WY, Munyendo WLL, Lv HX, Zhou JP. Studies on lactoferrin nanoparticles of gambogic acid for oral delivery. Drug Deliv 2013; 20:86-93. [PMID: 23495734 DOI: 10.3109/10717544.2013.766781] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Lactoferrin (Lf), a mammalian cationic iron-binding glycoprotein belonging to the transferrin (Tf) family, has been widely used in a variety of fields ranging from treating infant diarrhea and supporting newborn growth to food and pharmaceutical applications. In this study, Lf nanoparticles were firstly used as carriers of gambogic acid (GA) to enhance oral absorption and anti-cancer activity, hence reducing the related toxic effect. METHODS Gambogic acid-lactoferrin nanoparticles (GL-NPs) were prepared by the nanoparticle albumin-bound (NAB) technology. The formed nanoparticles were characterized by DSC, TEM, etc. In situ intestinal perfusion experiment was performed to clarify the absorption mechanism of GL-NPs. Furthermore in vivo and in vitro anti-tumor activities of GL-NPs were also investigated. RESULTS GL-NPs was successfully prepared with about 150 nm mean size, +20 mV ζ potential, 92.3 ± 7.2% encapsulation efficiency and 9.04 ± 0.7% DL; GL-NPs also exhibited a better stability and a desirable slow release in vitro experiment. The results of in situ intestinal perfusion showed a transformation of GA absorption from passive diffusion into active transport or facilitated diffusion by GL-NPs. MTT assay of GL-NPs showed almost an equal anti-proliferative effect with arginine solution of GA (Arg-GA) in HepG2 cell. The inhibitory rate against S180 tumor mice after oral administration of GL-NPs was up to 86.01% which was 1.39-folds of intravenous injection of Arg-GA. CONCLUSION The in vitro results showed that the NAB technology was feasible for industrial production of Lf nanoparticles and the in vivo results proved that the effective GL-NPs is a promising approach for the oral delivery of GA. These obtained research works have also paved the preliminary way for the study of Lf as an oral drug delivery carrier.
Collapse
Affiliation(s)
- Zhen-Hai Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | |
Collapse
|
39
|
Gambogic Acid Sensitizes Ovarian Cancer Cells to Doxorubicin Through ROS-Mediated Apoptosis. Cell Biochem Biophys 2013; 67:199-206. [DOI: 10.1007/s12013-013-9534-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
40
|
Jiang XL, Zhang Y, Luo CL, Wu XH. Targeting Renal Cell Carcinoma with Gambogic Acid in Combination with Sunitinib in Vitro and in Vivo. Asian Pac J Cancer Prev 2012; 13:6463-8. [DOI: 10.7314/apjcp.2012.13.12.6463] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
41
|
Li C, Qi Q, Lu N, Dai Q, Li F, Wang X, You Q, Guo Q. Gambogic acid promotes apoptosis and resistance to metastatic potential in MDA-MB-231 human breast carcinoma cells. Biochem Cell Biol 2012. [PMID: 23194187 DOI: 10.1139/o2012-030] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gambogic acid (GA) is considered a potent anti-tumor agent for its multiple effects on cancer cells in vitro and in vivo. Low concentrations of GA (0.3-1.2 µmol/L) can suppress invasion of human breast carcinoma cells without affecting cell viability. To get a whole profile of the inhibition on breast cancers, higher concentrations of GA and spontaneous metastatic animal models were employed. Treatment with GA (3 and 6 µmol/L) induced apoptosis in MDA-MB-231 cells and the accumulation of reactive oxygen species (ROS). Furthermore, GA induced PARP cleavage, activation of caspase-3, caspase-8, and caspase-9, as well as an increased ratio of Bax/Bcl-2. Moreover, the translocation of apoptotic inducing factor (AIF) and the release of cytochrome c (Cyt c) from mitochondria were observed, indicating that GA induced apoptosis through accumulation of ROS and mitochondrial apoptotic pathway. GA also inhibited cell survival via blocking Akt/mTOR signaling. In vivo, GA significantly inhibited the xenograft tumor growth and lung metastases in athymic BALB/c nude mice bearing MDA-MB-231 cells. Collectively, these data provide further support for the multiple effects of GA on human breast cancer cells, as well as for its potential application to inhibit tumor growth and prevent metastasis in human cancers.
Collapse
Affiliation(s)
- Chenglin Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Fang L, Chen B, Liu S, Wang R, Hu S, Xia G, Tian Y, Cai X. Synergistic effect of a combination of nanoparticulate Fe3O4 and gambogic acid on phosphatidylinositol 3-kinase/Akt/Bad pathway of LOVO cells. Int J Nanomedicine 2012; 7:4109-18. [PMID: 22888247 PMCID: PMC3415324 DOI: 10.2147/ijn.s32475] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Indexed: 11/23/2022] Open
Abstract
Background: The present study evaluated whether magnetic nanoparticles containing Fe3O4 could enhance the activity of gambogic acid in human colon cancer cells, and explored the potential mechanisms involved. Methods: Cytotoxicity was evaluated by MTT assay. The percentage of cells undergoing apoptosis was analyzed by flow cytometry, and cell morphology was observed under both an optical microscope and a fluorescence microscope. Reverse transcriptase polymerase chain reaction and Western blot assay were performed to determine the transcription of genes and expression of proteins, respectively. Results: Gambogic acid could inhibit proliferation of LOVO cells in a dose-dependent and time-dependent manner and induce apoptosis, which was dramatically enhanced by magnetic nanoparticles containing Fe3O4. The typical morphological features of apoptosis in LOVO cells were observed after treatment comprising gambogic acid with and without magnetic nanoparticles containing Fe3O4. Transcription of cytochrome c, caspase 9, and caspase 3 genes was higher in the group treated with magnetic nanoparticles containing Fe3O4 and gambogic acid than in the groups that received gambogic acid or magnetic nanoparticles containing Fe3O4, but transcription of phosphatidylinositol 3-kinase, Akt, and Bad genes decreased. Notably, expression of cytochrome c, caspase 9, and caspase 3 proteins in the group treated with gambogic acid and magnetic nanoparticles containing Fe3O4 was higher than in the groups receiving magnetic nanoparticles containing Fe3O4 or gambogic acid, while expression of p-PI3K, p-Akt, p-Bad, pro-caspase 9, and pro-caspase 3 degraded. Conclusion: Magnetic nanoparticles containing Fe3O4 can enhance apoptosis induced by gambogic acid which may be closely related to regulation of the PI3K/Akt/Bad pathway in the treatment of human colon cancer.
Collapse
Affiliation(s)
- Lianghua Fang
- No 1 Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Zou ZY, Wei J, Li XL, Yu LX, Wang TT, Qian XP, Liu BR. Enhancement of Anticancer Efficacy of Chemotherapeutics by Gambogic Acid Against Gastric Cancer Cells. Cancer Biother Radiopharm 2012; 27:299-306. [PMID: 22444164 DOI: 10.1089/cbr.2010.0943] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Zheng-Yun Zou
- Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Jia Wei
- Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xiao-Lin Li
- Jiangsu Province Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Li-Xia Yu
- Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | | | - Xiao-Ping Qian
- Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Bao-Rui Liu
- Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, China
| |
Collapse
|
44
|
Yang LJ, Chen Y, He J, Yi S, Wen L, Zhao S, Cui GH. Effects of gambogic acid on the activation of caspase-3 and downregulation of SIRT1 in RPMI-8226 multiple myeloma cells via the accumulation of ROS. Oncol Lett 2012; 3:1159-1165. [PMID: 22783411 DOI: 10.3892/ol.2012.634] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Accepted: 02/24/2012] [Indexed: 12/23/2022] Open
Abstract
Multiple myeloma (MM) is the second most commonly diagnosed hematologic malignancy. Although new drugs, including bortezomib and lenalidomide, have improved the treatment landscape for MM patients, MM remains incurable. Therefore, screening for novel anti-myeloma drugs is necessary. Gambogic acid (GA), the main active ingredient of gamboges secreted from the Garcinia hanburryi tree, has been reported to exhibit potent anticancer activity in certain solid tumors and hematological malignancies, while there are few studies that are available concerning its effects on MM cells. In the present study, we investigated the anticancer activity of GA on the MM RPMI-8226 cells and further studied the underlying mechanisms by which GA affected the cells. RPMI-8226 cells were cultured and the effect of GA on cell proliferation was analyzed using MTT assay. Hoechst 33258 staining was used to visualize nuclear fragmentation, and reactive oxygen species (ROS) levels were detected. GA was found to have a significant, dose-dependent effect on growth inhibition and apoptosis induction in RPMI-8226 cells. This activity is associated with the accumulation of ROS, which contributes to the activation of caspase-3 and the cleavage of poly (ADP-ribose) polymerase (PARP), accompanied with apoptosis in RPMI-8226 cells treated with GA. Mammalian SIRT1, as the closest homolog of the yeast Sir2, was extensively involved in regulating cell processes, including cell senescence, aging and neuronal protection, as well as having anti-apoptotic properties. Moreover, SIRT1 overexpression has been shown to protect cancer cells from chemotherapy and ionizing radiation. In the present study, we demonstrated that GA has the potential to downregulate the expression of SIRT1 via ROS accumulation. In conclusion, our study found that GA is able to induce apoptosis in RPMI-8226 cells via ROS accumulation followed by caspase-3 activation, PARP cleavage and SIRT1 downregulation. These results suggest that GA may have the potential to not only induce apoptosis in MM cells, but also to decrease the relapse rate of MM.
Collapse
Affiliation(s)
- Li-Jing Yang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | | | | | | | | | | | | |
Collapse
|
45
|
Saxena V, Hussain MD. Poloxamer 407/TPGS mixed micelles for delivery of gambogic acid to breast and multidrug-resistant cancer. Int J Nanomedicine 2012; 7:713-21. [PMID: 22359450 PMCID: PMC3284221 DOI: 10.2147/ijn.s28745] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background Delivery of a high concentration of anticancer drugs specifically to cancer cells remains the biggest challenge for the treatment of multidrug-resistant cancer. Poloxamers and D-α-Tocopheryl polyethylene glycol 1000 succinate (TPGS) are known inhibitors of P-glycoprotein (P-gp). Mixed micelles prepared from Poloxamer 407 and TPGS may increase the therapeutic efficacy of the drug by delivering high concentrations inside the cells and inhibiting P-gp. Gambogic acid (GA) is a naturally derived novel anticancer agent, but poor solubility and toxic side effects limit its use. In this study, we have developed Poloxamer 407 and TPGS mixed micelle-encapsulating GA for the treatment of breast and multidrug-resistant cancer. Methods GA-loaded Poloxamer 407/TPGS mixed micelles were prepared using a thin film hydration method, and their physicochemical properties were characterized. Cellular accumulation and cytotoxicity of the GA-loaded Poloxamer 407/TPGS mixed micelles were studied in breast cancer cells, MCF-7 cells, and multidrug-resistant NCI/ADR-RES cells. Results The diameter of GA-loaded Poloxamer 407/TPGS mixed micelles was about 17.4 ± 0.5 nm and the zeta potential −13.57 mV. The entrapment efficiency of GA was 93.1% ± 0.5% and drug loading was about 9.38% ± 0.29%. Differential scanning calorimetry and X-ray powder diffraction studies confirmed that GA is encapsulated by the polymers. The in vitro release studies showed that mixed micelles sustained the release of GA for more than 4 days. Results from cellular uptake studies indicated that GA-loaded Poloxamer 407/TPGS mixed micelles had increased cellular uptake of GA in NCI/ADR-RES cells. Cytotoxicity of GA-loaded Poloxamer 407/TPGS mixed micelles was found to be 2.9 times higher in multidrug-resistant NCI/ADR-RES cells, and 1.6 times higher in MCF-7 cells, as compared with unencapsulated GA. Conclusion This study suggests that Poloxamer 407/TPGS mixed micelles can be used as a delivery system for GA to treat breast and multidrug-resistant cancer.
Collapse
Affiliation(s)
- Vipin Saxena
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX, USA
| | | |
Collapse
|
46
|
Yin H, Guo R, Xu Y, Zheng Y, Hou Z, Dai X, Zhang Z, Zheng D, Xu H. Synergistic antitumor efficiency of docetaxel and curcumin against lung cancer. Acta Biochim Biophys Sin (Shanghai) 2012; 44:147-53. [PMID: 22126905 DOI: 10.1093/abbs/gmr106] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Curcumin (Cum), the principal polyphenolic curcuminoid, obtained from the turmeric rhizome Curcuma longa, is recently reported to have potential antitumor effects in vitro and in vivo. Docetaxel (Doc) is considered as first-line chemotherapy for the treatment of non-small cell lung cancer. Here we report for the first time that Cum could synergistically enhance the in vitro and in vivo antitumor efficacy of Doc against lung cancer. In the current study, combination index (CI) is calculated in both in vitro and in vivo studies to determine the interaction between Cum and Doc. In the in vitro cytotoxicity test, media-effect analysis clearly indicated a synergistic interaction between Cum and Doc in certain concentrations. Moreover, in vivo evaluation further demonstrated the superior anticancer efficacy of Cum + Doc compared with Doc alone by intravenous delivery in an established A549 transplanted xenograft model. Results showed that Cum synergistically increased the efficacy of Doc immediately after 4 days of the initial treatment. Additionally, simultaneous administration of Cum and Doc showed little toxicity to normal tissues including bone marrow and liver at the therapeutic doses. Therefore, in vitro and in vivo evaluations demonstrated the satisfying synergistic antitumor efficacy of Cum and Doc against lung cancer and the introduction of Cum in traditional chemotherapy is a most promising way to counter the spread of non-small cell lung cancer.
Collapse
Affiliation(s)
- Haitao Yin
- Department of Oncology, the Affiliated Zhongda Hospital of Southeastern University, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cheng H, Su JJ, Peng JY, Wang M, Wang XC, Yan FG, Wang XS, Li QL. Gambogenic acid inhibits proliferation of A549 cells through apoptosis inducing through up-regulation of the p38 MAPK cascade. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2011; 13:993-1002. [PMID: 22007630 DOI: 10.1080/10286020.2011.605062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Gamboge is a dry resin secreted from Garcinia hanburryi, and gambogenic acid (GNA) is one of the main active compounds of gamboge. We have previously demonstrated the anticancer activity of GNA in A549 cells and pointed out its potential effects in anticancer therapies. Previous studies reported that GNA induced apoptosis in many cancer cell lines and inhibited A549 tumor growth in xenograft of nude mice in vivo. However, the anticancer mechanism of GNA has still not been well studied. In this paper, we have investigated whether GNA-induced apoptosis is critically mediated by the p38 mitogen-activated protein kinase (MAPK) pathway. Our findings revealed that GNA could induce apoptosis, inhibit proliferation, down-regulate the expression of p38 and MAPK, increase the activations of caspase-9, caspase-3, and cytochrome c release. Furthermore, using SB203580, an adenosine triphosphate-competitive inhibitor of p38 MAPK, inhibit the expression of p-p38 and the experimental results show that it may promote the occurrence of apoptosis induced by GNA. Taken together, these results suggested that up-regulation of the p38 MAPK cascade may account for the activation of GNA-induced apoptosis.
Collapse
Affiliation(s)
- Hui Cheng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Traditional Chinese Medicine, Anhui University of Traditional Chinese Medicine, Hefei, 230038, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Wang J, Zhao Q, Qi Q, Gu HY, Rong JJ, Mu R, Zou MJ, Tao L, You QD, Guo QL. Gambogic acid-induced degradation of mutant p53 is mediated by proteasome and related to CHIP. J Cell Biochem 2011; 112:509-19. [PMID: 21268072 DOI: 10.1002/jcb.22941] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
As an oncoprotein, mutant p53 is a potential tumor-specific target for cancer therapy. Most mutated forms of the protein are largely accumulated in cancer cells due to their increased stability. In the present study, we demonstrate that mutant p53 protein stability is regulated by gambogic acid (GA). Following GA exposure, protein levels of mutant p53 decreased while the mRNA levels were not affected in MDA-MB-435 cells, which indicate that GA down-regulates mutant p53 at post-transcription level. Co-treatment with GA and cycloheximide, a protein synthesis inhibitor, induced a decrease of half-life of mutant p53 protein. These findings indicated that the reduction of mutant p53 by GA was due to the destabilization and degradation of the protein. Furthermore, inhibition of proteasome activity by MG132 blocked GA-induced down-regulation of mutant p53, causing mutant p53 accumulation in detergent-insoluble cellular fractions. Further studies revealed that mutant p53 was ubiquitinated and it was chaperones related ubiquitin ligase carboxy terminus of Hsp70-interacting protein (CHIP) rather than MDM2 involved in the degradation of mutant p53. In addition, GA prevented Hsp90/mutant p53 complex formation and enhanced interaction of mutant p53 with Hsp70. Depletion of CHIP stabilized mutant p53 in GA treated cells. In conclusion, mutant p53 may be down-regulated by GA through chaperones-assisted ubiquitin/proteasome degradation pathway in cancer cells.
Collapse
Affiliation(s)
- Jia Wang
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Jiangsu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Prasad S, Pandey MK, Yadav VR, Aggarwal BB. Gambogic acid inhibits STAT3 phosphorylation through activation of protein tyrosine phosphatase SHP-1: potential role in proliferation and apoptosis. Cancer Prev Res (Phila) 2011; 4:1084-94. [PMID: 21490133 DOI: 10.1158/1940-6207.capr-10-0340] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The transcription factor, STAT3, is associated with proliferation, survival, and metastasis of cancer cells. We investigated whether gambogic acid (GA), a xanthone derived from the resin of traditional Chinese medicine, Garcinia hanburyi (mangosteen), can regulate the STAT3 pathway, leading to suppression of growth and sensitization of cancer cells. We found that GA induced apoptosis in human multiple myeloma cells that correlated with the inhibition of both constitutive and inducible STAT3 activation. STAT3 phosphorylation at both tyrosine residue 705 and serine residue 727 was inhibited by GA. STAT3 suppression was mediated through the inhibition of activation of the protein tyrosine kinases Janus-activated kinase 1 (JAK1) and JAK2. Treatment with the protein tyrosine phosphatase (PTP) inhibitor pervanadate reversed the GA-induced downregulation of STAT3, suggesting the involvement of a PTP. We also found that GA induced the expression of the PTP SHP-1. Deletion of the SHP-1 gene by siRNA suppressed the ability of GA to inhibit STAT3 activation and to induce apoptosis, suggesting the critical role of SHP-1 in its action. Moreover, GA downregulated the expression of STAT3-regulated antiapoptotic (Bcl-2, Bcl-xL, and Mcl-1), proliferative (cyclin D1), and angiogenic (VEGF) proteins, and this correlated with suppression of proliferation and induction of apoptosis. Overall, these results suggest that GA blocks STAT3 activation, leading to suppression of tumor cell proliferation and induction of apoptosis.
Collapse
Affiliation(s)
- Sahdeo Prasad
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
50
|
Gupta SC, Kim JH, Prasad S, Aggarwal BB. Regulation of survival, proliferation, invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev 2010; 29:405-34. [PMID: 20737283 DOI: 10.1007/s10555-010-9235-2] [Citation(s) in RCA: 544] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Almost 25 centuries ago, Hippocrates, the father of medicine, proclaimed "Let food be thy medicine and medicine be thy food." Exploring the association between diet and health continues today. For example, we now know that as many as 35% of all cancers can be prevented by dietary changes. Carcinogenesis is a multistep process involving the transformation, survival, proliferation, invasion, angiogenesis, and metastasis of the tumor and may take up to 30 years. The pathways associated with this process have been linked to chronic inflammation, a major mediator of tumor progression. The human body consists of about 13 trillion cells, almost all of which are turned over within 100 days, indicating that 70,000 cells undergo apoptosis every minute. Thus, apoptosis/cell death is a normal physiological process, and it is rare that a lack of apoptosis kills the patient. Almost 90% of all deaths due to cancer are linked to metastasis of the tumor. How our diet can prevent cancer is the focus of this review. Specifically, we will discuss how nutraceuticals, such as allicin, apigenin, berberine, butein, caffeic acid, capsaicin, catechin gallate, celastrol, curcumin, epigallocatechin gallate, fisetin, flavopiridol, gambogic acid, genistein, plumbagin, quercetin, resveratrol, sanguinarine, silibinin, sulforaphane, taxol, gamma-tocotrienol, and zerumbone, derived from spices, legumes, fruits, nuts, and vegetables, can modulate inflammatory pathways and thus affect the survival, proliferation, invasion, angiogenesis, and metastasis of the tumor. Various cell signaling pathways that are modulated by these agents will also be discussed.
Collapse
Affiliation(s)
- Subash C Gupta
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|