1
|
Xiong Q, Li Z, Yang D, Liu X, Pu W, Yue X, Jia K, Wan X, Zou Y. Progress in the study of bioactivity, chemical composition and pharmacological mechanism of action in Wolfiporia cocos (F.A. Wolf) Ryvarden & Gilb. Front Pharmacol 2025; 16:1521235. [PMID: 40098611 PMCID: PMC11911342 DOI: 10.3389/fphar.2025.1521235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
The Latin name of Wolfiporia cocos is Wolfiporia cocos (F.A. Wolf) Ryvarden & Gilb, it a medicinal and edible mushroom belonging to the family Polyporaceae. Traditional Chinese medicine believes that it can strengthen the spleen, diuretic, tranquillise the mind and dispel dampness. So far, the chemical and active metabolites isolated and extracted from Wolfiporia cocos are mainly polysaccharides, triterpenoids, and sterols. Modern pharmacology has found that these chemical and active metabolites have a wide range of pharmacological effects, including antitumour, antioxidation, anti-inflammatory, immunomodulation, regulation of intestinal flora, regulation of glycolipid metabolism, and improvement of organ function. By applying Poria cocos, Poria, Wolfiporia cocos, Wolfiporia cocos (F.A. Wolf) Ryvarden & Gilb as search terms, we searched all the relevant studies on Poria cocos from Web of Science and PubMed databases and classified these categories of chemical and active metabolites according to the main research content of each literature and summarized its mechanism of action, updated its latest research results, and discussed the direction of further research in the future to provide a better reference for future clinical applications with better therapeutic effects and potential medicinal value.
Collapse
Affiliation(s)
- Qi Xiong
- School of Medical Information, Changchun University of Chinese Medicine, Changchun, China
| | - Zhuoran Li
- School of Medical Information, Changchun University of Chinese Medicine, Changchun, China
| | - Defeng Yang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Xinze Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Wanxu Pu
- School of Medical Information, Changchun University of Chinese Medicine, Changchun, China
| | - Xitao Yue
- School of Medical Information, Changchun University of Chinese Medicine, Changchun, China
| | - Kaikai Jia
- School of Medical Information, Changchun University of Chinese Medicine, Changchun, China
| | - Xilin Wan
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
- Jilin Aodong Pharmaceutial Group Co., Ltd., Post-Doctoral Research Center, Yanji, China
| | - Yuanjun Zou
- School of Medical Information, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
2
|
Nie J, Zhang H, Li X, Qin J, Zhou J, Lu Y, Yang N, Li Y, Li H, Li C. Pachymic acid promotes ferroptosis and inhibits gastric cancer progression by suppressing the PDGFRB-mediated PI3K/Akt pathway. Heliyon 2024; 10:e38800. [PMID: 39512319 PMCID: PMC11541463 DOI: 10.1016/j.heliyon.2024.e38800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
Gastric cancer (GC) is a common malignant tumour with high incidence and mortality rates worldwide. Despite current treatment modalities, including surgical resection and chemotherapy, challenges such as postoperative recurrence, metastasis and drug resistance persist. Therefore, investigating the feasibility and mechanism of traditional Chinese medicine in treating gastric cancer is crucial for discovering new anti-gastric cancer drugs or adjuvant therapies. Pachymic acid (PA) is a natural triterpenoid found in the traditional Chinese medicinal herb Poria cocos (PC) (Schw. Wolf). Recent studies have reported its inhibitory effects on various cancer cells, including liver, cervical, breast and gastric cancer. Our in vitro and in vivo experiments confirmed that PA inhibits the proliferation, migration and invasion of gastric cancer cells. The treatment of gastric cancer cells with various death inhibitors revealed that PA may suppress gastric cancer progression by inducing ferroptosis. Malondialdehyde, Fe2+, reactive oxygen species and glutathione assays were performed to validate the effects of PA on ferroptosis in gastric cancer. High-throughput sequencing combined with analysis of the TCGA database identified PDGFRB as a potential downstream target of PA. In vivo experiments indicated that the PDGFRB overexpression could counteract the antitumour effects of PA, while ferroptosis induced by the PI3K/Akt signalling pathway may play a key role in this process. This study provides initial evidence that PA, through its interaction with PDGFRB, alters the PI3K/Akt signalling pathway, leading to ferroptosis in gastric cancer cells, thus manifesting its antitumour properties. This discovery holds promise for the development of novel therapeutic strategies for gastric cancer patients.
Collapse
Affiliation(s)
- Jinlin Nie
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People's Republic of China
| | - Haoran Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People's Republic of China
| | - Xiaofeng Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| | - Jiawei Qin
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| | - Jiawei Zhou
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| | - Yuhui Lu
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| | - Nengjia Yang
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| | - Yanan Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| | - Hailiang Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| | - Cheng Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, People's Republic of China
| |
Collapse
|
3
|
Xiao Y, Hu Z, Liu H, Jiang X, Zhou T, Wang H, Long H, Li M. A review on antitumor effect of pachymic acid. Medicine (Baltimore) 2024; 103:e39752. [PMID: 39312302 PMCID: PMC11419566 DOI: 10.1097/md.0000000000039752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Poria cocos, also known as Jade Ling and Songbai taro, is a dry fungus core for Wolfiporia cocos, which is parasitic on the roots of pine trees. The ancients called it "medicine of four seasons" because of its extensive effect and ability to be combined with many medicines. Pachymic acid (PA) is one of the main biological compounds of Poria cocos. Research has shown that PA has various pharmacological properties, including anti-inflammatory and antioxidant. PA has recently attracted much attention due to its anticancer properties. Researchers have found that PA showed anticancer activity by regulating apoptosis and the cell cycle in vitro and in vivo. Using PA with anticancer drugs, radiotherapy, and biomaterials could also improve the sensitivity of cancer cells and delay the progression of cancer. The purpose of this review was to summarize the anticancer mechanism of PA by referencing the published documents. A review of the collected data indicated that PA had the potential to be developed into an effective anticancer agent.
Collapse
Affiliation(s)
- Yubo Xiao
- School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Zhaotun Hu
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province College of Biological and Food Engineering, Huaihua University, Huaihua, China
| | - Hang Liu
- School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Xinglin Jiang
- School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Taimei Zhou
- School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Haiying Wang
- School of Public Health and Laboratory Medicine, Hunan University of Medicine, Huaihua, China
| | - Heng Long
- Department of Breast and Thyroid Surgery, First People’s Hospital of Huaihua City, Huaihua, China
| | - Ming Li
- Department of Histology and Embryology, Hunan University of Medicine, Huaihua, China
| |
Collapse
|
4
|
Li D, Wang J, Tuo Z, Yoo KH, Yu Q, Miyamoto A, Zhang C, Ye X, Wei W, Wu R, Feng D. Natural products and derivatives in renal, urothelial and testicular cancers: Targeting signaling pathways and therapeutic potential. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 127:155503. [PMID: 38490077 DOI: 10.1016/j.phymed.2024.155503] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Natural products have demonstrated significant potential in cancer drug discovery, particularly in renal cancer (RCa), urothelial carcinoma (UC), and testicular cancer (TC). PURPOSE This review aims to examine the effects of natural products on RCa, UC and TC. STUDY DESIGN systematic review METHODS: PubMed and Web of Science databases were retrieved to search studies about the effects of natural products and derivatives on these cancers. Relevant publications in the reference list of enrolled studies were also checked. RESULTS This review highlighted their diverse impacts on key aspects such as cell growth, apoptosis, metastasis, therapy response, and the immune microenvironment. Natural products not only hold promise for novel drug development but also enhance the efficacy of existing chemotherapy and immunotherapy. Importantly, we exert their effects through modulation of critical pathways and target genes, including the PI3K/AKT pathway, NF-κB pathway, STAT pathway and MAPK pathway, among others in RCa, UC, and TC. CONCLUSION These mechanistic insights provide valuable guidance for researchers, facilitating the selection of promising natural products for cancer management and offering potential avenues for further gene regulation studies in the context of cancer treatment.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - Qingxin Yu
- Department of pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province, 315211, China
| | - Akira Miyamoto
- Department of Rehabilitation, West Kyushu University, Japan
| | - Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Xing Ye
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| |
Collapse
|
5
|
Li X, He A, Liu C, Li Y, Luo Y, Xiong W, Nian W, Zuo D. Pachymic acid activates TP53INP2/TRAF6/caspase-8 pathway to promote apoptosis in renal cell carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2024. [PMID: 38560766 DOI: 10.1002/tox.24195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/26/2024] [Accepted: 02/10/2024] [Indexed: 04/04/2024]
Abstract
While pachymic acid (PA), a key component of Poria cocos (Schw.), has demonstrated anti-tumor effects in lung, breast, and pancreatic cancers, its impact on renal cell carcinoma (RCC) is unclear. This study evaluated the effect of PA on proliferation, migration, and apoptosis in human renal cancer A498 and ACHN cells as well as in cancer xenograft mice using wound scratch test, Western blotting, and co-immunoprecipitation assays. In a dose- and time-dependent manner, PA exhibited significant inhibition of RCC cell proliferation, migration, and invasion, accompanied by the induction of apoptosis. Additionally, PA upregulated the expression of tumor protein p53-inducible nuclear protein 2 (TP53INP2) and tumor necrosis factor receptor-associated factor 6 (TRAF6), which were downregulated in renal papillary and chromophobe carcinoma, resulting in inhibited tumor growth in mice. PA treatment elevated cleaved-caspase 3 and 8, and PARP levels, and facilitated TP53INP2 and TRAF6 binding to caspase 8, promoting its ubiquitination. Molecular docking revealed interactions between PA and TP53INP2, TRAF6. In summary, PA inhibits RCC development by upregulating TP53INP2 and promoting TRAF6-induced caspase 8 ubiquitination, activating apoptotic pathways.
Collapse
Affiliation(s)
- Xunjia Li
- Department of Nephrology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
- Department of Research and Development, Chongqing Precision Medical Industry Technology Research Institute, Chongqing, China
| | - An He
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chengxuan Liu
- Department of Nephrology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Ying Li
- Department of Nephrology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Yan Luo
- Department of Nephrology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Weijian Xiong
- Department of Nephrology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Weiqi Nian
- Department of Oncology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Deyu Zuo
- Department of Rehabilitation Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
6
|
Farooqi AA, Venera R, Kapanova G, Tanbayeva G, Akhmetova G, Kudabayev Y, Turgambayeva A. TRAIL-mediated signaling in bladder cancer: realization of clinical efficacy of TRAIL-based therapeutics in medical oncology. Med Oncol 2023; 40:236. [PMID: 37432489 DOI: 10.1007/s12032-023-02078-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/12/2023] [Indexed: 07/12/2023]
Abstract
Bladder cancer is a therapeutically challenging disease and wealth of knowledge has enabled researchers to develop a clear understanding of mechanisms which underlie carcinogenesis and metastasis. Excitingly, research over decades has unveiled wide-ranging mechanisms which serve as central engine in progression of bladder cancer. Loss of apoptosis, drug resistance, and pro-survival signaling are some of the highly studied cellular mechanisms. Therefore, restoration of apoptosis in resistant cancers is a valuable and attractive strategy. Discovery of TRAIL-mediated signaling cascade is an intriguing facet of molecular oncology. In this review, we have provided an overview of the translational and foundational advancements in dissecting the genomic and proteomic cartography of TRAIL signaling exclusively in the context of bladder cancer. We have also summarized how different natural products sensitized drug-resistant bladder cancer cells to TRAIL-mediated apoptosis. Interestingly, different death receptors that activate agonistic antibodies have been tested in various phases of clinical trials against different cancers. Certain clues of scientific evidence have provided encouraging results about efficacy of these agonistic antibodies (lexatumumab and mapatumumab) against bladder cancer cell lines. Therefore, multipronged approaches consisting of natural products, chemotherapeutics, and agonistic antibodies will realistically and mechanistically provide proof-of-concept for the translational potential of these combinatorial strategies in well-designed clinical trials.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan.
| | | | - Gulnara Kapanova
- Al-Farabi Kazakh National University, Almaty, 71 al-Farabi Ave, 050040, Almaty, Kazakhstan
- Scientific Center of Anti-infectious Drugs, Kazakhstan, 75 a al-Faraby Ave, 050040, Almaty, Kazakhstan
| | - Gulnur Tanbayeva
- Al-Farabi Kazakh National University, Almaty, 71 al-Farabi Ave, 050040, Almaty, Kazakhstan
| | - Gulshara Akhmetova
- Scientific Center of Anti-infectious Drugs, Kazakhstan, 75 a al-Faraby Ave, 050040, Almaty, Kazakhstan
| | | | - Assiya Turgambayeva
- Department Public Health and Management, NJSC, Astana Medical University, Astana, Kazakhstan
| |
Collapse
|
7
|
Yang M, Yan Q, Luo Y, Wang B, Deng S, Luo H, Ye B, Wang X. Molecular mechanism of Ganji Fang in the treatment of hepatocellular carcinoma based on network pharmacology, molecular docking and experimental verification technology. Front Pharmacol 2023; 14:1016967. [PMID: 36744264 PMCID: PMC9892186 DOI: 10.3389/fphar.2023.1016967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a malignant tumor harmful to human health. Ganji Fang (GJF) has good clinical efficacy in the treatment of HCC, but its mechanism is still unclear. Objective: The aim of this study was to investigate the mechanism of action of GJF in the treatment of HCC through network pharmacology, molecular docking and in vitro experiments. Methods: A series of network pharmacology methods were used to identify the potential targets and key pathways of GJF in the treatment of HCC. Then, molecular docking technology was used to explore the binding ability of key active ingredients and targets in GJF. Multiple external databases were used to validate the key targets. In in vitro experiments, we performed MTT assays, wound-healing assays, cell cycle assays, apoptosis assays and RT‒qPCR to verify the inhibitory effect of GJF on the Human hepatoma G2 (HepG2) cells. Result: A total of 162 bioactive components and 826 protein targets of GJF were screened, and 611 potential targets of HCC were identified. Finally, 63 possible targets of GJF acting on HCC were obtained. KEGG enrichment analyses showed that the top five pathways were the cell cycle, cellular senescence, p53 signaling pathway, PI3K/Akt signaling pathway, and progesterone-mediated oocyte maturation. Among them, we verified the PI3K/Akt signaling pathway. CCNE1, PKN1, CCND2, CDK4, EPHA2, FGFR3, CDK6, CDK2 and HSP90AAI were enriched in the PI3K/Akt pathway. The molecular docking results showed that the docking scores of eight active components of GJF with the two targets were all less than -5.0, indicating that they had certain binding activity. In vitro cell experiments showed that GJF could inhibit the proliferation and migration of HepG2 cells, block the cell cycle and induce apoptosis of HepG2 cells, which may be related to the PI3K/Akt signaling pathway. In summary, EPHA2 may be an important target of GJF in HCC, and pachymic acid may be an important critical active compound of GJF that exerts anticancer activity. Conclusion: In general, we demonstrated, for the first time, that the molecular mechanism of GJF in HCC may involve induction of G0/G1 phase cycle arrest through inhibition of the PI3K/Akt signaling pathway and promote apoptosis of hepatoma cell lines. This study provides a scientific basis for the subsequent clinical application of GJF and the in-depth study of its mechanism.
Collapse
Affiliation(s)
- Miaolun Yang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Yan
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuehua Luo
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Boqing Wang
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shicong Deng
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiyan Luo
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baoqian Ye
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiongwen Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Xiongwen Wang,
| |
Collapse
|
8
|
Wang H, Sun X, Wei C, Wang J, Xu Y, Bai G, Yao Q, Zhang L. Synthesis and bioactivity evaluation of pachymic acid derivatives as potential cytotoxic agents. Med Chem Res 2023; 32:342-354. [PMID: 36593868 PMCID: PMC9797892 DOI: 10.1007/s00044-022-03009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022]
Abstract
Pachymic acid, a well-known natural lanostane-type triterpenoid, exhibits various pharmacological properties. In this study, 18 derivatives of pachymic acid were synthesized by modifying their molecular structures and evaluated for their anticancer activity against two human cancer cell lines using the CCK-8 assay. Structure-activity relationship studies according to the in vitro cytotoxicity unexpectedly found one promising derivative A17 (namely tumulosic acid, also found in Poria cocos), which had stronger anti-proliferative activity than the positive drug cisplatin against HepG2 and HSC-2 cell lines with IC50 values of 7.36 ± 0.98 and 2.50 ± 0.15 μM, respectively. Further pharmacological analysis demonstrated that A17 induced HSC-2 cell cycle arrest at the S phase, cell apoptosis, and autophagy. Western blotting confirmed the regulatory effects of A17 on cell cycle arrest-, apoptosis-, and autophagy-related proteins expression. In addition, A17 regulated the AKT and AMPK pathways in HSC-2 cells. These results demonstrated that A17 possesses great potential as an anticancer agent. Graphical Abstract
Collapse
Affiliation(s)
- Hezhen Wang
- grid.417409.f0000 0001 0240 6969Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 563000 Zunyi, China
| | - Xun Sun
- grid.417409.f0000 0001 0240 6969Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 563000 Zunyi, China
| | - Chunyong Wei
- grid.417409.f0000 0001 0240 6969Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 563000 Zunyi, China
| | - Jing Wang
- grid.417409.f0000 0001 0240 6969Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 563000 Zunyi, China
| | - Yingshu Xu
- grid.417409.f0000 0001 0240 6969Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 563000 Zunyi, China
| | - Guohui Bai
- grid.417409.f0000 0001 0240 6969Key Laboratory of Oral Disease Research, School of Stomatology, Zunyi Medical University, 563000 Zunyi, China
| | - Qizheng Yao
- grid.254147.10000 0000 9776 7793Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, 210009 Nanjing, China
| | - Lei Zhang
- grid.417409.f0000 0001 0240 6969Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, 563000 Zunyi, China
| |
Collapse
|
9
|
Jiang F, Zhu T, Yang C, Chen Y, Fu Z, Jiang L, Liu Y. Pachymic Acid Inhibits Growth and Metastatic Potential in Liver Cancer HepG2 and Huh7 Cells. Biol Pharm Bull 2023; 46:35-41. [PMID: 36273899 DOI: 10.1248/bpb.b22-00440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Pachymic acid (PA), exacted from Polyporaceae, has been known for its biological activities including diuretic, dormitive, anti-oxidant, anti-aging, anti-inflammatory and anticancer properties in several types of diseases. Recently, studies have demonstrated that PA could suppress cell growth and induce cell apoptosis in different kinds of cancer cells. But the underlying mechanisms remain poorly elucidated. In the current study, we investigated the effect of pachymic acid on liver cancer cells and its underlying mechanisms. Our results evidenced that pachymic acid effectively inhibited the cell growth and metastatic potential in HepG2 and Huh7 cells. Mechanistically, we revealed that pachymic acid triggered cell apoptosis by increasing caspase 3 and caspase 9 cleavage, upregulating Bax and cytochrome c expression, while reducing the expression of Bcl2. Besides, pachymic acid could markedly inhibit the cell invasion and migration and cell metastatic potential by mediating epithelial-to-mesenchymal transition (EMT) markers and metastasis-associated genes in HepG2 and Huh7 cells. In addition, we demonstrated that FAK-Src-Jun N-terminal kinase (JNK)-matrix metalloproteinase 2 (MMP2) axis was involved in PA-inhibited liver cell EMT. Together, these results contribute to our deeper understanding of the anti-cancer effects of pachymic acid on liver cancer cells. This study also provided compelling evidence that PA might be a potential therapeutic agent for liver cancer treatment.
Collapse
Affiliation(s)
- Feng Jiang
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University
| | - Tieming Zhu
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University
| | - Chunfeng Yang
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University
| | - Yang Chen
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University
| | - Zhidong Fu
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University
| | - Lihui Jiang
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University
| | - Yongzhi Liu
- Department of General Surgery, Affiliated Xiaoshan Hospital, Hangzhou Normal University
| |
Collapse
|
10
|
Yang T, Tian S, Wang Y, Ji J, Zhao J. Antitumor activity of pachymic acid in cervical cancer through inducing endoplasmic reticulum stress, mitochondrial dysfunction, and activating the AMPK pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2121-2132. [PMID: 35524978 DOI: 10.1002/tox.23555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 04/17/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
Pachymic acid has various pharmacological effects, including anti-inflammatory, antioxidant, immunomodulatory, and antitumor. However, the role of pachymic acid in cervical cancer remains unclear. So, we investigated the effects of pachymic acid in cervical cancer and elucidated the underlying mechanisms. We treated HeLa cells and normal cervical epithelial cells (HUCECs) with pachymic acid (0, 10, 20, 40, 80, or 160 μM) for 72 h, and found the cell activity was decreased in cells treated with 160 μM pachymic acid for 48 h or 80 μM pachymic acid for 72 h, while HUCECs viability without effect. Next, we observed that endoplasmic reticulum (ER) related gene expression, mitochondrial membrane potential (MMP) changes, ATP depletion, reactive oxygen species (ROS) generation and apoptosis were increased. Moreover, we observed that cytochrome C (Cytc) expression was increased and apoptosis-inducing factor (AIF) was decreased in the cytoplasm of pachymic acid-treated HeLa cells. Tauroursodeoxycholic acid (TUDCA) of ER stress inhibitor reversed the effects of pachymic acid on HeLa cells. Phosphorylation of AMPK and acetyl-CoA carboxylase (ACC) of the AMPK pathway key protein was upregulated in pachymic acid-induced HeLa cells. Finally, we subcutaneously implanted HeLa cells into female nude mice and treated them with pachymic acid (50 mg/kg) for 3 weeks (5 days/week), and observed in pachymic acid induced xenograft mice, tumor growth was suppressed, cell apoptosis, ER-related gene expression, and ROS levels in tumor tissues were increased. Therefore, these findings demonstrated that pachymic acid plays an anti-tumor activity in cervical cancer through inducing ER stress, mitochondrial dysfunction, and activating the AMPK pathway.
Collapse
Affiliation(s)
- Ting Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Sijuan Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yaohui Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Jing Ji
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Juan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
11
|
Wei C, Wang H, Sun X, Bai Z, Wang J, Bai G, Yao Q, Xu Y, Zhang L. Pharmacological profiles and therapeutic applications of pachymic acid (Review). Exp Ther Med 2022; 24:547. [PMID: 35978941 PMCID: PMC9366251 DOI: 10.3892/etm.2022.11484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/17/2022] [Indexed: 01/10/2023] Open
Abstract
Poria cocos is a saprophytic fungus that grows in diverse species of Pinus. Its sclerotium, called fu-ling or hoelen, has been used in various traditional Chinese medicines and health foods for thousands of years, and in several modern proprietary traditional Chinese medicinal products. It has extensive clinical indications, including sedative, diuretic, and tonic effects. Pachymic acid (PA) is the main lanostane-type triterpenoid in Poria cocos. Evidence suggests that PA has various biological properties such as cytotoxic, anti-inflammatory, antihyperglycemic, antiviral, antibacterial, sedative-hypnotic, and anti-ischemia/reperfusion activities. Although considerable advancements have been made, some fundamental and intricate issues remain unclear, such as the underlying mechanisms of PA. The present study aimed to summarize the biological properties and therapeutic potential of PA. The biosynthetic, pharmacokinetic, and metabolic pathways of PA, and its underlying mechanisms were also comprehensively summarized.
Collapse
Affiliation(s)
- Chunyong Wei
- Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hezhen Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xun Sun
- Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhixun Bai
- Department of Internal Medicine, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jing Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Guohui Bai
- Key Laboratory of Oral Disease Research, School of Stomatology, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Qizheng Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Yingshu Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Lei Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
12
|
Chen HY, Lei JY, Li SL, Guo LQ, Lin JF, Wu GH, Lu J, Ye ZW. Progress in biological activities and biosynthesis of edible fungi terpenoids. Crit Rev Food Sci Nutr 2022; 63:7288-7310. [PMID: 35238261 DOI: 10.1080/10408398.2022.2045559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The edible fungi have both edible and medicinal functions, in which terpenoids are one of the most important active ingredients. Terpenoids possess a wide range of biological activities and show great potential in the pharmaceutical and healthcare industries. In this review, the diverse biological activities of edible fungi terpenoids were summarized with emphasis on the mechanism of anti-cancer and anti-inflammation. Subsequently, this review focuses on advances in knowledge and understanding of the biosynthesis of terpenoids in edible fungi, especially in the generation of sesquiterpenes, diterpenes, and triterpenes. This paper is aim to provide an overview of biological functions and biosynthesis developed for utilizing the terpenoids in edible fungi.
Collapse
Affiliation(s)
- Hai-Ying Chen
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jin-Yu Lei
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Shu-Li Li
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Li-Qiong Guo
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Jun-Fang Lin
- College of Food Science, South China Agricultural University, Guangzhou, China
| | - Guang-Hong Wu
- College of Food Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Jun Lu
- Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Zhi-Wei Ye
- College of Food Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, China
| |
Collapse
|
13
|
Panda SK, Sahoo G, Swain SS, Luyten W. Anticancer Activities of Mushrooms: A Neglected Source for Drug Discovery. Pharmaceuticals (Basel) 2022; 15:176. [PMID: 35215289 PMCID: PMC8876642 DOI: 10.3390/ph15020176] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/08/2023] Open
Abstract
Approximately 270 species of mushrooms have been reported as potentially useful for human health. However, few mushrooms have been studied for bioactive compounds that can be helpful in treating various diseases. Like other natural regimens, the mushroom treatment appears safe, as could be expected from their long culinary and medicinal use. This review aims to provide a critical discussion on clinical trial evidence for mushrooms to treat patients with diverse types of cancer. In addition, the review also highlights the identified bioactive compounds and corresponding mechanisms of action among the explored mushrooms. Furthermore, it also discusses mushrooms with anticancer properties, demonstrated either in vitro and/or in vivo models, which have never been tested in clinical studies. Several mushrooms have been tested in phase I or II clinical trials, mostly for treating breast cancer (18.6%), followed by colorectal (14%) and prostate cancer (11.6%). The majority of clinical studies were carried out with just 3 species: Lentinula edodes (22.2%), Coriolus versicolor, and Ganoderma lucidum (both 13.9%); followed by two other species: Agaricus bisporus and Grifola frondosa (both 11.1%). Most in vitro cell studies use breast cancer cell lines (43.9%), followed by lung (14%) and colorectal cancer cell lines (13.1%), while most in vivo animal studies are performed in mice tumor models (58.7%). Although 32 species of mushrooms at least show some promise for the treatment of cancer, only 11 species have been tested clinically thus far. Moreover, most clinical studies have investigated fewer numbers of patients, and have been limited to phase III or IV. Therefore, despite the promising preclinical and clinical data publication, more solid scientific efforts are required to clarify the therapeutic value of mushrooms in oncology.
Collapse
Affiliation(s)
- Sujogya Kumar Panda
- Center of Environment, Climate Change and Public Health, RUSA 2.0, Utkal University, Bhubaneswar 751004, India
- Department of Zoology, Utkal University, Bhubaneswar 751004, India;
- Department of Biology, KU Leuven, 3000 Leuven, Belgium;
| | - Gunanidhi Sahoo
- Department of Zoology, Utkal University, Bhubaneswar 751004, India;
| | - Shasank S. Swain
- Division of Microbiology and NCDs, ICMR-Regional Medical Research Centre, Bhubaneswar 751023, India;
| | - Walter Luyten
- Department of Biology, KU Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
14
|
Tan W, Pan T, Wang S, Li P, Men Y, Tan R, Zhong Z, Wang Y. Immunometabolism modulation, a new trick of edible and medicinal plants in cancer treatment. Food Chem 2021; 376:131860. [PMID: 34971892 DOI: 10.1016/j.foodchem.2021.131860] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/04/2021] [Accepted: 12/10/2021] [Indexed: 12/23/2022]
Abstract
The edible and medicinal plants (EMPs) are becoming an abundant source for cancer prevention and treatment since the natural and healthy trend for modern human beings. Currently, there are more than one hundred species of EMPs widely used and listed by the national health commission of China, and most of them indicate immune or metabolic regulation potential in cancer treatment with numerous studies over the past two decades. In the present review, we focused on the metabolic influence in immunocytes and tumor microenvironment, including immune response, immunosuppressive factors and cancer cells, discussing the immunometabolic potential of EMPs in cancer treatment. There are more than five hundred references collected and analyzed through retrieving pharmacological studies deposited in PubMed by medical subject headings and the corresponding names derived from pharmacopoeia of China as a sole criterion. Finally, the immunometabolism modulation of EMPs was sketch out implying an immunometabolic control in cancer treatment.
Collapse
Affiliation(s)
- Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Tingrui Pan
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu 215123, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| | - Yongfan Men
- Research Laboratory of Biomedical Optics and Molecular Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Rui Tan
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China.
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China.
| |
Collapse
|
15
|
Ma R, Zhang Z, Xu J, Liang X, Zhao Q. Poricoic acid A induces apoptosis and autophagy in ovarian cancer via modulating the mTOR/p70s6k signaling axis. Braz J Med Biol Res 2021; 54:e11183. [PMID: 34669780 PMCID: PMC8521541 DOI: 10.1590/1414-431x2021e11183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Due to the high mortality and rapid disease progression, ovarian cancer remains one of the most common malignancies threatening the health of women. The present study was conducted to explore the anticancer effects and the underlying mechanisms of poricoic acid A (PAA), the main components of Poria cocos, on ovarian cancer. We investigated the anticancer effects of different concentrations of PAA in the SKOV3 cell line. Cell viability and proliferation were examined by CCK-8 assay. Cellular migration and invasion were assessed by the scratch and Transwell migration assays, respectively. The effect of PPA on cell apoptosis was measured by flow cytometry and caspase-3/8/9 colorimetric assay. Western blot was performed to detect protein level changes related to apoptosis and mTOR signaling pathways. The in vivo anticancer effect of PAA was evaluated using xenograft tumorigenesis model in nude mice. Our results showed that PAA suppressed SKOV3 cellular viability, migration, and invasion in a dosage-dependent manner. Flow cytometry results demonstrated PAA treatment could induce SKOV3 cell apoptosis. In addition, increased ratio of LC3-II/LC3-I (a marker for autophagosome formation) was observed after PAA treatment, as well as inhibition of m-TOR and p70s6k phosphorylation. In nude mice, PAA treatment reduced the xenograft tumor weight by 70% (P<0.05). In conclusion, our data suggested that PAA induced apoptosis and autophagy in ovarian cancer via modulating the mTOR/p70s6k signaling axis.
Collapse
Affiliation(s)
- Rui Ma
- Department of Pharmacy, The 305 Hospital of PLA, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Zhenhua Zhang
- Department of Gastroenterology, The 305 Hospital of PLA, Beijing, China
| | - Jin Xu
- Department of Neurology, Chang Zheng Hospital, Navy Medical University, Shangai, China
| | - Xueqi Liang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Zhao
- Department of Pharmacy, The 305 Hospital of PLA, Beijing, China
| |
Collapse
|
16
|
Xia Y, Chen R, Lu G, Li C, Lian S, Kang TW, Jung YD. Natural Phytochemicals in Bladder Cancer Prevention and Therapy. Front Oncol 2021; 11:652033. [PMID: 33996570 PMCID: PMC8120318 DOI: 10.3389/fonc.2021.652033] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022] Open
Abstract
Phytochemicals are natural small-molecule compounds derived from plants that have attracted attention for their anticancer activities. Some phytochemicals have been developed as first-line anticancer drugs, such as paclitaxel and vincristine. In addition, several phytochemicals show good tumor suppression functions in various cancer types. Bladder cancer is a malignant tumor of the urinary system. To date, few specific phytochemicals have been used for bladder cancer therapy, although many have been studied in bladder cancer cells and mouse models. Therefore, it is important to collate and summarize the available information on the role of phytochemicals in the prevention and treatment of bladder cancer. In this review, we summarize the effects of several phytochemicals including flavonoids, steroids, nitrogen compounds, and aromatic substances with anticancer properties and classify the mechanism of action of phytochemicals in bladder cancer. This review will contribute to facilitating the development of new anticancer drugs and strategies for the treatment of bladder cancer using phytochemicals.
Collapse
Affiliation(s)
- Yong Xia
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Ruijiao Chen
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Guangzhen Lu
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Changlin Li
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Sen Lian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Taek-Won Kang
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, South Korea
| | - Young Do Jung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
17
|
Jiang Y, Fan L. The effect of Poria cocos ethanol extract on the intestinal barrier function and intestinal microbiota in mice with breast cancer. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113456. [PMID: 33039631 DOI: 10.1016/j.jep.2020.113456] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/14/2020] [Accepted: 10/03/2020] [Indexed: 05/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Poria cocos Wolf has been used in traditional East-Asian medicine for centuries to effectively treat various gastrointestinal disorders such as diarrhea for its tonic, anti-fungal and anti-bacterial activities. Previous studies have revealed that the tumor development would induce intestinal microbiota dysbiosis and intestinal barrier dysfunction to the patients with breast cancer. AIM OF STUDY To investigate the effect and the mechanism of ethanol extract of Poria cocos (PC) on intestinal barrier function and intestinal microbiota in the mice with breast cancer. MATERIALS AND METHODS Thirty-six female BALB/c mice were randomly divided into four groups (the normal control, model, PC and positive control group). Intestinal histopathological was evaluated by H&E staining. The difference of the intestinal microbiota in each group was studied by 16S rDNA high-throughput sequencing. The level of plasma endotoxin, D -lactic acid (D-LA) and diamine oxidase (DAO) were measured by ELISA. The putrescine content in serum and urine were detected by HPLC. Expression of the tight junction (TJ) proteins, phosphorylated p38 MAPK and ERK1/2 were determined by western blotting. RESULTS Our results showed that tumor development prominently induced the intestinal damage and microbiome dysbiosis in mice. PC prominently remit such histologic damage through enhancing the expression of TJ proteins and decreasing the levels of DAO, D-LA and endotoxin via upregulating the expression of phosphorylated ERK1/2 and p38 MAPK. Furthermore, PC increased the diversity of the intestinal microbiota and strikingly changed the structure and composition of the gut microbiota in the mice by increasing the beneficial bacteria Lactobacillus, Bifidobacterium, and decreasing the sulfate-reducing bacteria Desulfovibrio and inflammatory associated bacteria Mucispirillum, S24-7 and Staphylococcus. Moreover, PICRUSt analysis and the putrescine detection might indicate that PC might be involved in the putrescine metabolism in the mice. Correlation analysis indicated that Prevotella, Rikenellaceae and Bacteroidetes were significantly correlated with Claudin-8 and p38-MAPK expression (p < 0.05). CONCLUSION PC could improve the dysbacteriosis and repair the intestinal barrier function in the mice with breast cancer. This study provide more data to support the application of PC in breast cancer treatment.
Collapse
Affiliation(s)
- Yu Jiang
- State Key Laboratory of Food Science & Technology, Jiangnan University, Wuxi, 214122, China
| | - Liuping Fan
- State Key Laboratory of Food Science & Technology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
18
|
Jung EJ, Lee WS, Paramanantham A, Kim HJ, Shin SC, Kim GS, Jung JM, Ryu CH, Hong SC, Chung KH, Kim CW. p53 Enhances Artemisia annua L. Polyphenols-Induced Cell Death Through Upregulation of p53-Dependent Targets and Cleavage of PARP1 and Lamin A/C in HCT116 Colorectal Cancer Cells. Int J Mol Sci 2020; 21:ijms21239315. [PMID: 33297377 PMCID: PMC7730414 DOI: 10.3390/ijms21239315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 12/11/2022] Open
Abstract
Plant-derived natural polyphenols exhibit anticancer activity without showing any noticeable toxicities to normal cells. The aim of this study was to investigate the role of p53 on the anticancer effect of polyphenols isolated from Korean Artemisia annua L. (pKAL) in HCT116 human colorectal cancer cells. We confirmed that pKAL induced reactive oxygen species (ROS) production, propidium iodide (PI) uptake, nuclear structure change, and acidic vesicles in a p53-independent manner in p53-null HCT116 cells through fluorescence microscopy analysis of DCF/PI-, DAPI-, and AO-stained cells. The pKAL-induced anticancer effects were found to be significantly higher in p53-wild HCT116 cells than in p53-null by hematoxylin staining, CCK-8 assay, Western blot, and flow cytometric analysis of annexin V/PI-stained cells. In addition, expression of ectopic p53 in p53-null cells was upregulated by pKAL in both the nucleus and cytoplasm, increasing pKAL-induced cell death. Moreover, Western bot analysis revealed that pKAL-induced cell death was associated with upregulation of p53-dependent targets such as p21, Bax and DR5 and cleavage of PARP1 and lamin A/C in p53-wild HCT116 cells, but not in p53-null. Taken together, these results indicate that p53 plays an important role in enhancing the anticancer effects of pKAL by upregulating p53 downstream targets and inducing intracellular cell death processes.
Collapse
Affiliation(s)
- Eun Joo Jung
- Departments of Biochemistry, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea; (E.J.J.); (C.W.K.)
| | - Won Sup Lee
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
- Correspondence: ; Tel.: +82-55-750-8733; Fax: +82-55-758-9122
| | - Anjugam Paramanantham
- Departments of Internal Medicine, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Hye Jung Kim
- Departments of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Jin-Myung Jung
- Departments of Neurosurgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Chung Ho Ryu
- Department of Food Technology, Research Institute of Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Soon Chan Hong
- Departments of Surgery, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Ky Hyun Chung
- Departments of Urology, Institute of Health Sciences, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju 52727, Korea;
| | - Choong Won Kim
- Departments of Biochemistry, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 52727, Korea; (E.J.J.); (C.W.K.)
| |
Collapse
|
19
|
Jiang Y, Fan L. Evaluation of anticancer activities of Poria cocos ethanol extract in breast cancer: In vivo and in vitro, identification and mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2020; 257:112851. [PMID: 32283190 DOI: 10.1016/j.jep.2020.112851] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/30/2020] [Accepted: 04/04/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Poria cocos Wolf (P. cocos), a well-known traditional East-Asian medicinal and edible fungus, is one of the most important components in Chinese medicine formulas like "Guizhi fuling wan" to treat hyperplasia of mammary glands and breast cancer. AIMING OF STUDY In this study, we attempted to verify the anticancer efficacy of the ethanol extract of P. cocos (PC) on the breast cancer as well as to investigate its most active compound and its underlying molecular mechanism in vivo and in vitro. MATERIALS AND METHODS The key anti-cancer components were separated and purified through chromatography and identified by spectral analyses. The in vivo anti-breast cancer efficacy and side effects of PC were evaluated in BALB/c nude mice that have been subcutaneously injected with breast cancer cells MDA-MB-231. Cytotoxicity, apoptosis and cell cycle arrest of PC were evaluated in vitro by cell viability assays and flow cytometry. The protein levels were examined via western blotting. RESULTS Pachymic acid (PA), separated and identified as the most active compound, induced the significant cytotoxicity on breast cancer cells MDA-MB-231(IC50 value, 2.13 ± 0.24 μg/mL) and was not active against the normal breast epithelium cells MCF-10A. The in vivo experiment revealed that PC could significantly inhibit the tumor development and the final mean tumor weight of the mice in the PC group (0.51 ± 0.12g) was significantly lower than that in the model group (1.22 ± 0.45g). Notably, compared to the first-line anticancer drug cisplatin, PC showed less side effects on the function of the vital organs and the muscle strength of the mice. Among in vitro study, PC significantly inhibited the cell growth of MDA-MB-231 by inducing cell apoptosis and cell cycle arrested at G0/G1 phase in a dose-dependent manner. The expression of cell cycle-associated cyclin D1, cyclin E, CDK2, and CDK4 were downregulated, while p53 and p21 expression were upregulated following the PA treatment. In addition, PA downregulated the apoptotic regulator Bcl-2, increased the expression of pro-apoptotic protein Bax, and promoted the release of cytochrome c and the activation of cleaved caspase-3, -9 and caspase -8 via mitochondria-mediated and death receptor-mediated signaling pathways. CONCLUSION This study verified the anticancer efficacy of PC on breast cancer in vivo and in vitro through induction of cell apoptosis and G0/G1 cell cycle arrest. The data also suggested that PA could be developed as an efficacious agent for breast cancer treatment with less side effects.
Collapse
Affiliation(s)
- Yu Jiang
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Liuping Fan
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
20
|
Ko YS, Jung EJ, Go SI, Jeong BK, Kim GS, Jung JM, Hong SC, Kim CW, Kim HJ, Lee WS. Polyphenols Extracted from Artemisia annua L. Exhibit Anti-Cancer Effects on Radio-Resistant MDA-MB-231 Human Breast Cancer Cells by Suppressing Stem Cell Phenotype, β-Catenin, and MMP-9. Molecules 2020; 25:molecules25081916. [PMID: 32326231 PMCID: PMC7221914 DOI: 10.3390/molecules25081916] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Artemisia annua L. has been reported to show anti-cancer activities. Here, we determined whether polyphenols extracted from Artemisia annua L. (pKAL) exhibit anti-cancer effects on radio-resistant MDA-MB-231 human breast cancer cells (RT-R-MDA-MB-231 cells), and further explored their molecular mechanisms. Cell viability assay and colony-forming assay revealed that pKAL inhibited cell proliferation on both parental and RT-R-MDA-MB-231 cells in a dose-dependent manner. The anti-proliferative effects of pKAL on RT-R-MDA-MB-231 cells were superior or similar to those on parental ones. Western blot analysis revealed that expressions of cluster of differentiation 44 (CD44) and Oct 3/4, matrix metalloproteinase-9 (MMP-9) and signal transducer and activator of transcription-3 (STAT-3) phosphorylation were significantly increased in RT-R-MDA-MB-231 cells compared to parental ones, suggesting that these proteins could be associated with RT resistance. pKAL inhibited the expression of CD44 and Oct 3/4 (CSC markers), and β-catenin and MMP-9 as well as STAT-3 phosphorylation of RT-R-MDA-MB-231. Regarding upstream signaling, the JNK or JAK2 inhibitor could inhibit STAT-3 activation in RT-R-MDA-MB-231 cells, but not augmented pKAL-induced anti-cancer effects. These findings suggest that c-Jun N-terminal kinase (JNK) or Janus kinase 2 (JAK2)/STAT3 signaling are not closely related to the anti-cancer effects of pKAL. In conclusion, this study suggests that pKAL exhibit anti-cancer effects on RT-R-MDA-MB-231 cells by suppressing CD44 and Oct 3/4, β-catenin and MMP-9, which appeared to be linked to RT resistance of RT-R-MDA-MB-231 cells.
Collapse
Affiliation(s)
- Young Shin Ko
- Department of Pharmacology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, 816 Beongil 15 Jinjudaero, Jinju 52727, Korea;
| | - Eun Joo Jung
- Departments of Biochemistry, Institute of Health Sciences, Gyeongsang National University College of Medicine, 90 Chilam-dong, Jinju 660-702, Korea; (E.J.J.); (C.W.K.)
| | - Se-il Go
- Departments of Internal Medicine, Institute of Health Sciences and Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, 90 Chilam-dong, Jinju 660-702, Korea;
| | - Bae Kwon Jeong
- Departments of Radiation Oncology, Institute of Health Sciences and Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, 90 Chilam-dong, Jinju 660-702, Korea;
| | - Gon Sup Kim
- School of Veterinary and Institute of Life Science, Gyeongsang National University, 900 Gajwadong, Jinju 660-701, Korea;
| | - Jin-Myung Jung
- Departments of Neurosurgery, Institute of Health Sciences and Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, 90 Chilam-dong, Jinju 660-702, Korea;
| | - Soon Chan Hong
- Departments of Surgery, Institute of Health Sciences and Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, 90 Chilam-dong, Jinju 660-702, Korea;
| | - Choong Won Kim
- Departments of Biochemistry, Institute of Health Sciences, Gyeongsang National University College of Medicine, 90 Chilam-dong, Jinju 660-702, Korea; (E.J.J.); (C.W.K.)
| | - Hye Jung Kim
- Department of Pharmacology, College of Medicine, Institute of Health Sciences, Gyeongsang National University, 816 Beongil 15 Jinjudaero, Jinju 52727, Korea;
- Correspondence: (H.J.K.); (W.S.L.); Tel.: +82-55-750-8733 (W.S.L.)
| | - Won Sup Lee
- Departments of Internal Medicine, Institute of Health Sciences and Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, 90 Chilam-dong, Jinju 660-702, Korea;
- Correspondence: (H.J.K.); (W.S.L.); Tel.: +82-55-750-8733 (W.S.L.)
| |
Collapse
|
21
|
Pang Y, Zhu S, Pei H. Pachymic acid protects against cerebral ischemia/reperfusion injury by the PI3K/Akt signaling pathway. Metab Brain Dis 2020; 35:673-680. [PMID: 32140824 DOI: 10.1007/s11011-020-00540-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/23/2020] [Indexed: 12/26/2022]
Abstract
Pachymic acid (PA) from medicinal fungus Poria cocos has a variety of pharmacological potentials. However, there are no reports of the effects of PA on cerebral ischemia/reperfusion (I/R) injury. The purpose of this study was to investigate the mechanisms of PA on cerebral I/R injury in rats. The effects of PA on cerebral infarction size, brain water content, neurological symptoms and cerebral blood flow were evaluated. Nissl staining was used to observe the damage of ischemic brain neurons after I/R in rats. Apoptosis of ischemic brain neurons after I/R was observed by TUNEL staining. The effect of PA on the expression of some components of PI3K/Akt was detected by Western blotting. PA significantly increased cerebral blood flow after I/R in rats, reduced infarct volume and brain water content, and downgrade neurological function scores, significantly reduced neuronal damage after I/R in rats, and significantly decreased neuronal apoptosis. The effect of PA on rat I/R can be eliminated by LY294002. In addition, PA significantly up-regulated the protein expression of p-PTEN (Ser380), p-PDK1 (Ser241), p-Akt (Ser473), pc-Raf (Ser259) and p-BAD (Ser136), and down-regulated Cleaved caspase protein expression. LY294002 can reverse the effect of PA on the expression of PI3K / Akt signaling pathway related protein in rats after I/R. PA had obviously neuroprotective effects on brain I/R injury and neuronal apoptosis, and its mechanism may be related to activation of PI3K / Akt signaling pathway.
Collapse
Affiliation(s)
- Yingqiao Pang
- Medical School of Qingdao University, No. 38 Dengzhou Road, Qingdao City, Shandong Province, 266021, People's Republic of China
| | - Shaozhi Zhu
- Medical School of Qingdao University, No. 38 Dengzhou Road, Qingdao City, Shandong Province, 266021, People's Republic of China
| | - Haitao Pei
- Department of Neurology, The Affiliated Hospital of the Qingdao University, No.19 Jiangsu Road, Qingdao City, Shandong Province, 266011, People's Republic of China.
| |
Collapse
|
22
|
Zhang P, Zhao S, Lu X, Shi Z, Liu H, Zhu B. Metformin enhances the sensitivity of colorectal cancer cells to cisplatin through ROS-mediated PI3K/Akt signaling pathway. Gene 2020; 745:144623. [PMID: 32222530 DOI: 10.1016/j.gene.2020.144623] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/08/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023]
Abstract
Metformin and cisplatin have been widely studied as antitumor agents. However, the effect of metformin combined with cisplatin has not been investigated in colorectal cancer (CRC) cells. This study was aimed to explore the effect of metformin or/and cisplatin on cell viability, apoptosis, and the related signaling pathways in CRC SW480 and SW620 cells. We found that metformin or cisplatin inhibited cell viability of SW480 and SW620 cells in a concentration- and time-dependent manner. Furthermore, metformin combined with cisplatin obviously inhibited cell viability, decreased colony formation, induced apoptosis, mediated cleavage of caspase-9, caspase-3 and PARP, activated mitochondrial membrane potential, downregulated Mcl-1 and Bcl-2 expression, upregulated Bak and Bax expression, and increased reactive oxygen species (ROS) production, compared to the individual agent in SW480 and SW620 cells, which were attenuated by N-acetyl-L-cysteine (NAC), a ROS scavenger. Moreover, NAC could recover the downregulation of p-PI3K and p-Akt treated with combination of metformin and cisplatin, which subsequently activated the PI3K/Akt signaling pathway. Taken together, our results demonstrated that metformin enhanced the sensitivity of CRC cells to cisplatin through ROS-mediated PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Pei Zhang
- School of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, China
| | - Surong Zhao
- School of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, China
| | - Xingyue Lu
- School of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, China
| | - Zongfen Shi
- School of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical College, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, China.
| | - Bing Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, Anhui, China.
| |
Collapse
|
23
|
Xu H, Wang Y, Zhao J, Jurutka PW, Huang D, Liu L, Zhang L, Wang S, Chen Y, Cheng S. Triterpenes from
Poria cocos
are revealed as potential retinoid X receptor selective agonists based on cell and in silico evidence. Chem Biol Drug Des 2020; 95:493-502. [DOI: 10.1111/cbdd.13610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/11/2019] [Accepted: 08/11/2019] [Indexed: 01/17/2023]
Affiliation(s)
- Hui Xu
- Department of Food Quality and Safety School of Engineering China Pharmaceutical University Nanjing China
| | - Yuchen Wang
- Laboratory of Molecular Design and Drug Discovery School of Science China Pharmaceutical University Nanjing China
| | - Junnan Zhao
- Laboratory of Molecular Design and Drug Discovery School of Science China Pharmaceutical University Nanjing China
| | - Peter W. Jurutka
- School of Mathematical and Natural Sciences Arizona State University Phoenix AZ USA
| | - Dechun Huang
- Department of Pharmaceutical Engineering School of Engineering China Pharmaceutical University Nanjing China
| | - Liangyun Liu
- Department of Food Quality and Safety School of Engineering China Pharmaceutical University Nanjing China
| | - Lange Zhang
- Department of Food Quality and Safety School of Engineering China Pharmaceutical University Nanjing China
| | - Suilou Wang
- Department of Food Quality and Safety School of Engineering China Pharmaceutical University Nanjing China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery School of Science China Pharmaceutical University Nanjing China
| | - Shujie Cheng
- Department of Food Quality and Safety School of Engineering China Pharmaceutical University Nanjing China
| |
Collapse
|
24
|
Ding B, Ji X, Sun X, Zhang T, Mu S. In vitro effect of pachymic acid on the activity of Cytochrome P450 enzymes. Xenobiotica 2020; 50:913-918. [PMID: 32026737 DOI: 10.1080/00498254.2020.1727062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pachymic acid is a wildly used traditional Chinese medicine with various pharmacological features. It also exists in many drugs which are wildly used in pediatric.The effect of pachymic acid on the activity of eight major CYP isoforms was investigated in human liver microsomes.The effects of pachymic acid on eight human liver CYP isoforms (i.e. 1A2, 3A4, 2A6, 2E1, 2D6, 2C9, 2C19 and 2C8) were investigated in vitro using human liver microsomes (HLMs), and the enzyme kinetic parameters were calculated.The activity of CP3A4, 2E1, and 2C9 was inhibited by pachymic acid in a concentration-dependent manner with IC50 values of 15.04, 27.95, and 24.22 μM, respectively. Pachymic acid is a non-competitive inhibitor of CYP3A4, with the Ki value of 6.47 μM. While the inhibition of CYP2E1 and 2C9 was performed in a competitive manner, with the Ki value of 11.96 and 10.94 μM, respectively. Moreover, the inhibition of CYP3A4 was in a time-dependent manner with the KI/Kinact value of 7.77/0.048 min-1 μM-1.The in vitro inhibitory effect of pachymic acid on the activity of CYP3A4, 2E1, and 2C9 indicated the potential drug-drug interaction with the drugs that metabolized by CYP3A4, 2E1, and 2C9. Further clinical and in vivo studies are needed to evaluate the significance of this interaction.
Collapse
Affiliation(s)
- Baodong Ding
- Department of Pediatrics, Weifang Yidu Central Hospital, Weifang, China
| | - Xiaofei Ji
- Department of Pediatrics, Weifang Yidu Central Hospital, Weifang, China
| | - Xueming Sun
- Department of Pediatrics, Weifang Yidu Central Hospital, Weifang, China
| | - Tongtong Zhang
- Weihai Songshan Community Health Service Center, Weihai, China
| | - Suping Mu
- Department of Pediatrics, Woman and Children's Hospital Of Weifang People's Hospital, Weifang, China
| |
Collapse
|
25
|
Cheng S, Castillo V, Sliva D. CDC20 associated with cancer metastasis and novel mushroom‑derived CDC20 inhibitors with antimetastatic activity. Int J Oncol 2019; 54:2250-2256. [PMID: 31081056 DOI: 10.3892/ijo.2019.4791] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/26/2019] [Indexed: 11/06/2022] Open
Abstract
Aberrant expression of cell division cycle 20 (CDC20) is associated with malignant progression and poor prognosis in various types of cancer. The development of specific CDC20 inhibitors may be a novel strategy for the treatment of cancer with elevated expression of CDC20. The aim of the current study was to elucidate the role of CDC20 in cancer cell invasiveness and to identify novel natural inhibitors of CDC20. The authors found that CDC20 knockdown inhibited the migration of chemoresistant PANC‑1 pancreatic cancer cells and the metastatic MDA‑MB‑231 breast cancer cell line. By contrast, the overexpression of CDC20 by plasmid transfection promoted the metastasizing capacities of the PANC‑1 cells and MCF‑7 breast cancer cells. It was also identified that a triterpene mixture extracted from the mushroom Poria cocos (PTE), purified triterpenes dehydropachymic acid, and polyporenic acid C (PPAC) downregulated the expression of CDC20 in PANC‑1 cells dose‑dependently. Migration was also suppressed by PTE and PPAC in a dose‑dependent manner, which was consistent with expectations. Taken together, the present study is the first, to the best of our knowledge, to demonstrate that CDC20 serves an important role in cancer metastasis and that triterpenes from P. cocos inhibit the migration of pancreatic cancer cells associated with CDC20. Further investigations are in progress to investigate the specific mechanism associated with CDC20 and these triterpenes, which may have future potential use as natural agents in the treatment of metastatic cancer.
Collapse
Affiliation(s)
- Shujie Cheng
- Department of Food Quality and Safety, School of Engineering, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Victor Castillo
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN 46202, USA
| | - Daniel Sliva
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN 46202, USA
| |
Collapse
|
26
|
Miao G, Han J, Zhang J, Wu Y, Tong G. Targeting Pyruvate Kinase M2 and Hexokinase II, Pachymic Acid Impairs Glucose Metabolism and Induces Mitochondrial Apoptosis. Biol Pharm Bull 2019; 42:123-129. [DOI: 10.1248/bpb.b18-00730] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Guopeng Miao
- Department of Bioengineering, Huainan Normal University
| | - Juan Han
- Department of Bioengineering, Huainan Normal University
| | - Jifeng Zhang
- Department of Bioengineering, Huainan Normal University
| | - Yihai Wu
- Department of Bioengineering, Huainan Normal University
| | - Guanhe Tong
- Department of Bioengineering, Huainan Normal University
| |
Collapse
|
27
|
Bonam SR, Wu YS, Tunki L, Chellian R, Halmuthur MSK, Muller S, Pandy V. What Has Come out from Phytomedicines and Herbal Edibles for the Treatment of Cancer? ChemMedChem 2018; 13:1854-1872. [PMID: 29927521 DOI: 10.1002/cmdc.201800343] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/19/2018] [Indexed: 12/20/2022]
Abstract
Several modern treatment strategies have been adopted to combat cancer with the aim of minimizing toxicity. Medicinal plant-based compounds with the potential to treat cancer have been widely studied in preclinical research and have elicited many innovations in cutting-edge clinical research. In parallel, researchers have eagerly tried to decrease the toxicity of current chemotherapeutic agents either by combining them with herbals or in using herbals alone. The aim of this article is to present an update of medicinal plants and their bioactive compounds, or mere changes in the bioactive compounds, along with herbal edibles, which display efficacy against diverse cancer cells and in anticancer therapy. It describes the basic mechanism(s) of action of phytochemicals used either alone or in combination therapy with other phytochemicals or herbal edibles. This review also highlights the remarkable synergistic effects that arise between certain herbals and chemotherapeutic agents used in oncology. The anticancer phytochemicals used in clinical research are also described; furthermore, we discuss our own experience related to semisynthetic derivatives, which are developed based on phytochemicals. Overall, this compilation is intended to facilitate research and development projects on phytopharmaceuticals for successful anticancer drug discovery.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- UMR 7242 CNRS, Biotechnology and Cell Signaling, University of Strasbourg, Laboratory of Excellence Medalis, Illkirch, 67400, France.,Vaccine Immunology Laboratory, Natural Product Chemistry Division, CSIR - Indian Institute of Chemical Technology (IICT), Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, CSIR - Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Yuan Seng Wu
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lakshmi Tunki
- Vaccine Immunology Laboratory, Natural Product Chemistry Division, CSIR - Indian Institute of Chemical Technology (IICT), Hyderabad, 500007, India
| | - Ranjithkumar Chellian
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mahabalarao Sampath Kumar Halmuthur
- Vaccine Immunology Laboratory, Natural Product Chemistry Division, CSIR - Indian Institute of Chemical Technology (IICT), Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, CSIR - Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Sylviane Muller
- UMR 7242 CNRS, Biotechnology and Cell Signaling, University of Strasbourg, Laboratory of Excellence Medalis, Illkirch, 67400, France.,University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, 67000, France
| | - Vijayapandi Pandy
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.,Department of Pharmacology, Chalapathi Institute of Pharmaceutical Sciences, Lam, Guntur, Andhra Pradesh, 522034, India
| |
Collapse
|
28
|
Qiao X, Wang X, Shang Y, Li Y, Chen SZ. Azithromycin enhances anticancer activity of TRAIL by inhibiting autophagy and up-regulating the protein levels of DR4/5 in colon cancer cells in vitro and in vivo. Cancer Commun (Lond) 2018; 38:43. [PMID: 29970185 PMCID: PMC6029027 DOI: 10.1186/s40880-018-0309-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 06/06/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Azithromycin is a member of macrolide antibiotics, and has been reported to inhibit the proliferation of cancer cells. However, the underlying mechanisms are not been fully elucidated. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively targets tumor cells without damaging healthy cells. In the present study, we examined whether azithromycin is synergistic with TRAIL, and if so, the underlying mechanisms in colon cancers. METHODS HCT-116, SW480, SW620 and DiFi cells were treated with azithromycin, purified TRAIL, or their combination. A sulforhoddamine B assay was used to examine cell survival. Apoptosis was examined using annexin V-FITC/PI staining, and autophagy was observed by acridine orange staining. Western blot analysis was used to detect protein expression levels. In mechanistic experiments, siRNAs were used to knockdown death receptors (DR4, DR5) and LC-3B. The anticancer effect of azithromycin and TRAIL was also examined in BALB/c nude mice carrying HCT-116 xenografts. RESULTS Azithromycin decreased the proliferation of HCT-116 and SW480 cells in a dose-dependent manner. Combination of azithromycin and TRAIL inhibited tumor growth in a manner that could not be explained by additive effects. Azithromycin increased the expressions of DR4, DR5, p62 and LC-3B proteins and potentiated induction of apoptosis by TRAIL. Knockdown of DR4 and DR5 with siRNAs increased cell survival rate and decreased the expression of cleaved-PARP induced by the combination of azithromycin and TRAIL. LC-3B siRNA and CQ potentiated the anti-proliferation activity of TRAIL alone, and increased the expressions of DR4 and DR5. CONCLUSION The synergistic antitumor effect of azithromycin and TRAIL mainly relies on the up-regulations of DR4 and DR5, which in turn result from LC-3B-involved autophagy inhibition.
Collapse
Affiliation(s)
- Xinran Qiao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P.R. China
| | - Xiaofei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P.R. China
| | - Yue Shang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P.R. China
| | - Yi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P.R. China
| | - Shu-Zhen Chen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P.R. China.
| |
Collapse
|
29
|
Wang Z, Qi F, Cui Y, Zhao L, Sun X, Tang W, Cai P. An update on Chinese herbal medicines as adjuvant treatment of anticancer therapeutics. Biosci Trends 2018; 12:220-239. [DOI: 10.5582/bst.2018.01144] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Zhixue Wang
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University
| | - Fanghua Qi
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University
| | - Yangang Cui
- Department of Chemotherapy, Shandong Provincial Hospital affiliated to Shandong University
| | - Lin Zhao
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University
| | - Xiaogang Sun
- Department of Tumor Minimally Invasive Surgery, Shandong Provincial Hospital affiliated to Shandong University
| | - Wei Tang
- National Center for Global Health and Medicine
| | - Pingping Cai
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affiliated to Shandong University
| |
Collapse
|
30
|
Xu Y, Tong Y, Ying J, Lei Z, Wan L, Zhu X, Ye F, Mao P, Wu X, Pan R, Peng B, Liu Y, Zhu J. Chrysin induces cell growth arrest, apoptosis, and ER stress and inhibits the activation of STAT3 through the generation of ROS in bladder cancer cells. Oncol Lett 2018; 15:9117-9125. [PMID: 29805643 PMCID: PMC5958737 DOI: 10.3892/ol.2018.8522] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022] Open
Abstract
Chrysin is a natural flavone that has various biological activities, including antitumor effects. However, the effect of chrysin on bladder cancer cells remains elusive. The present study investigated the effects of chrysin on bladder cancer cells and its underlying mechanisms. The results demonstrated that chrysin induced apoptosis via the intrinsic pathway, as evidenced by activation of caspase-9 and caspase-3, however not caspase-8. In addition, chrysin reduced the expression of anti-apoptotic B cell lymphoma (Bcl) proteins including Bcl-2, Mcl-1, Bcl-xl, and promoted the protein expression of pro-apoptotic Bcl-2 associated X, apoptosis regulator. Chrysin also induced endoplasmic reticulum stress via activation of the unfolded protein response of PRKR-like endoplasmic reticulum kinase, eIF2α and activating transcription factor 4 in bladder cancer cells. Additionally, chrysin inhibited the signal transducer and activator of transcription 3 pathway. Furthermore, the generation of reactive oxygen species (ROS) was detected following treatment with chrysin. The ROS scavenger N-acetylcysteine inhibited the antitumor effect of chrysin. Collectively, these results indicate chrysin may act as a promising therapeutic candidate for targeting bladder cancer.
Collapse
Affiliation(s)
- Yi Xu
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Yanyue Tong
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Junjie Ying
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Zhangming Lei
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Lijun Wan
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Xiuwen Zhu
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Feng Ye
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Penglei Mao
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Xinkuan Wu
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Renbing Pan
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Bo Peng
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Yukun Liu
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| | - Jianyong Zhu
- Department of Urology, Quzhou People's Hospital, Quzhou, Zhejiang 324000, P.R. China
| |
Collapse
|
31
|
Lu C, Cai D, Ma J. Pachymic Acid Sensitizes Gastric Cancer Cells to Radiation Therapy by Upregulating Bax through Hypoxia. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:875-890. [PMID: 29737213 DOI: 10.1142/s0192415x18500465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We have previously shown that pachymic acid (PA) inhibited tumorigenesis of gastric cancer (GC) cells. However, the exact mechanism underlying the radiation response of GC was still elusive. To evaluate the effects of PA treatment on radiation response of GC cell lines both in vitro and in vivo, a colony formation assay and xenograft mouse model were employed. Changes in Bax and HIF1[Formula: see text] expressions were assessed in GC cells following PA treatment. Luciferase reporter and chromatin immune-precipitation assays were carried out to investigate the regulation of Bax through HIF1[Formula: see text]. Stable HIF1[Formula: see text] knockdown was introduced into GC cells to further study the mechanism underlying PA-enhanced response to radiation both in vitro and in vivo. PA greatly enhanced the sensitivity of GC cells to radiation in vitro and in vivo, upregulated Bax expression and inhibited hypoxia. Bax expression was under hypoxia inhibition, and PA increased Bax expression through repressing HIF1[Formula: see text]. Stable HIF1[Formula: see text] overexpression in GC cells abolished the sensitizing effect of PA on GC cells to radiation both in vitro and in vivo. PA functions as a radiation sensitizing compound in GC. PA treatment induces the expression of pro-apoptotic factor Bax by inhibiting hypoxia/HIF1[Formula: see text], supporting the therapeutic potential of PA in radiation therapy against GC.
Collapse
Affiliation(s)
- Chunwei Lu
- * Zhongshan Hospital, Fudan University, Xuhui District, Shanghai 200032, P. R. China
| | - Dingfang Cai
- * Zhongshan Hospital, Fudan University, Xuhui District, Shanghai 200032, P. R. China
| | - Jun Ma
- * Zhongshan Hospital, Fudan University, Xuhui District, Shanghai 200032, P. R. China
| |
Collapse
|
32
|
Aras A, Khalid S, Jabeen S, Farooqi AA, Xu B. Regulation of cancer cell signaling pathways by mushrooms and their bioactive molecules: Overview of the journey from benchtop to clinical trials. Food Chem Toxicol 2018; 119:206-214. [PMID: 29680270 DOI: 10.1016/j.fct.2018.04.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 12/14/2022]
Abstract
Mushrooms represent a tremendous source of biologically useful and pharmacologically active molecules. Recent breakthroughs in cancer genetics, genomics, proteomics and translational research have helped us to develop a better understanding of the underlying mechanisms which are contributory in cancer development and progression. Different signaling pathways particularly, Wnt, SHH, TGF/SMAD and JAK/STAT have been shown to modulate cancer progression and development. Increasingly it is being realized that genetic/epigenetic mutations and loss of apoptosis also mandate a 'multi-molecular' perspective for the development of therapies to treat cancer. In this review we attempted to provide an overview of the regulation of different signaling pathways by mushrooms and their bioactive compounds. Regulation of Wnt and JAK-STAT pathways by mushrooms is deeply studied but we do not have comprehensive information about regulation of TGF/SMAD, Notch and TRAIL induced signaling pathways because of superficially available data. There are outstanding questions related to modulation of oncogenic and tumor suppressor microRNAs by mushrooms in different cancers. Therefore, detailed mechanistic insights related to targeting of multiple pathways by extracts or bioactive compounds from mushrooms will be helpful in bridging our current knowledge gaps and translation of medicinally precious bioactive molecules to clinically effective therapeutics.
Collapse
Affiliation(s)
- Aliye Aras
- Department of Botany, Faculty of Science, Istanbul University, Istanbul 34460, Turkey
| | - Sumbul Khalid
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Saima Jabeen
- Department of Zoology, University of Gujrat, Sub-Campus, Rawalpindi, Pakistan
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan.
| | - Baojun Xu
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
33
|
Ji L, Zhong B, Jiang X, Mao F, Liu G, Song B, Wang CY, Jiao Y, Wang JP, Xu ZB, Li X, Zhan B. Actein induces autophagy and apoptosis in human bladder cancer by potentiating ROS/JNK and inhibiting AKT pathways. Oncotarget 2017; 8:112498-112515. [PMID: 29348843 PMCID: PMC5762528 DOI: 10.18632/oncotarget.22274] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023] Open
Abstract
Human bladder cancer is a common genitourinary malignant cancer worldwide. However, new therapeutic strategies are required to overcome its stagnated survival rate. Triterpene glycoside Actein (ACT), extracted from the herb black cohosh, suppresses the growth of human breast cancer cells. Our study attempted to explore the role of ACT in human bladder cancer cell growth and to reveal the underlying molecular mechanisms. We found that ACT significantly impeded the bladder cancer cell proliferation via induction of G2/M cycle arrest. Additionally, ACT administration triggered autophagy and apoptosis in bladder cancer cells, proved by the autophagosome formation, LC3B-II accumulation, improved cleavage of Caspases/poly (ADP-ribose) polymerase (PARP). Furthermore, reduction of reactive oxygen species (ROS) and p-c-Jun N-terminal kinase (JNK) could markedly reverse ACT-induced autophagy and apoptosis. In contrast, AKT and mammalian target of rapamycin (mTOR) were greatly de-phosphorylated by ACT, while suppressing AKT and mTOR activity could enhance the effects of ACT on apoptosis and autophagy induction. In vivo, ACT reduced the tumor growth with little toxicity. Taken together, our findings indicated that ACT suppressed cell proliferation, induced autophagy and apoptosis through promoting ROS/JNK activation, and blunting AKT pathway in human bladder cancer, which indicated that ACT might be an effective candidate against human bladder cancer in future.
Collapse
Affiliation(s)
- Lu Ji
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bing Zhong
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Xi Jiang
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Fei Mao
- Department of Urology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Gang Liu
- Department of Orthopaedics, Huai’an First People's Hospital, Nanjing Medical University, Huai’an 223300, China
| | - Bin Song
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Cheng-Yuan Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Yong Jiao
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Jiang-Ping Wang
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Zhi-Bin Xu
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Xing Li
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| | - Bo Zhan
- Branch of Raw Material and Natural Products, Far East Biological Products Co. LTD., Nanjing 210009, China
| |
Collapse
|
34
|
Park C, Lee WS, Han MH, Song KS, Hong SH, Nagappan A, Kim GY, Kim GS, Jung JM, Ryu CH, Shin SC, Hong SC, Choi YH. Lonicera japonica Thunb. Induces caspase-dependent apoptosis through death receptors and suppression of AKT in U937 human leukemic cells. Phytother Res 2017; 32:504-513. [PMID: 29193390 DOI: 10.1002/ptr.5996] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 11/07/2022]
Abstract
Decoctions obtained from the dried flowers of Lonicera japonica Thunb. (Indongcho) have been utilized in folk remedies against inflammatory diseases. Recently, many agents that have used for inflammatory diseases are showing anticancer effects. Here, we have isolated polyphenols extracted from lyophilized Lonicera japonica Thunb (PELJ) and investigated the anticancer effects of PELJ on U937 cells. Here, we demonstrated that PELJ induced apoptosis by upregulation of DR4 and Fas, and further it is augmented by suppression of XIAP. In addition, The PELJ-induced apoptosis is at least in part by blocking PI3K/Akt pathway. These findings suggest that PELJ may provide evidence of anticancer activities on U937 cells. Further study for detailed mechanism and the effects on animal models is warranted to determine whether PELJ provide more conclusive evidence that PELJ which may provide a beneficial effect for treating cancer.
Collapse
Affiliation(s)
- Cheol Park
- Department of Molecular Biology, College of Natural Sciences and Human Ecology, Dongeui University, Busan, 614-714, South Korea
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 660-702, South Korea
| | - Min-Ho Han
- Natural products Research Team, National Marine Biodiversity Institute of Korea, -gun, Seocheon, 325-902, South Korea
| | - Kyoung Seob Song
- Department of Physiology, Kosin University College of Medicine, Busan, 602-703, South Korea
| | - Su-Hyun Hong
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan, 614-052, South Korea
| | - Arulkumar Nagappan
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 660-702, South Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju, 690-756, South Korea
| | - Gon Sup Kim
- School of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, 660-701, South Korea
| | - Jin-Myung Jung
- Department Neurosurgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 660-702, South Korea
| | - Chung Ho Ryu
- Division of Applied Life Science(BK 21 Program), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 660-701, South Korea
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju, 660-701, South Korea
| | - Soon Chan Hong
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 660-702, South Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine and anti-Aging Research Center, Dongeui University, Busan, 614-052, South Korea
| |
Collapse
|
35
|
Lu JN, Panchanathan R, Lee WS, Kim HJ, Kim DH, Choi YH, Kim GS, Shin SC, Hong SC. Anthocyanins from the Fruit of Vitis Coignetiae Pulliat Inhibit TNF-Augmented Cancer Proliferation, Migration, and Invasion in A549 Cells. Asian Pac J Cancer Prev 2017; 18:2919-2923. [PMID: 29172259 PMCID: PMC5773771 DOI: 10.22034/apjcp.2017.18.11.2919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Objective: Anthocyanins belong to a class of flavonoids, exhibiting antioxidant and anti-inflammatory actions have been reported to have anti-cancer effects. Here, we investigated whether anthocyanins can inhibit cancer cell proliferation, invasion, and angiogenesis in human lung cancer A549 cells, which are critically involved in cancer metastasis. Methods: We used anthocyanins from fruits of Vitis coignetiae Pulliat (AIMs) which has been used in Korean folk medicine for the treatment of inflammatory diseases and cancers. We have performed cell proliferation assays, cell invasion assay, gelatin zymography, wound healing assay and western blotting to examine whether anthocyanins can inhibit cancer cell proliferation, invasion, and angiogenesis in A549 cells. Result: AIMs did not inhibit cancer cell proliferation on A549 cells. Also, AIMs suppressed cancer migration, and invasion by supressing MMP-2 and MMP-9 expression. The Immuno-blotting results also revealed that AIMs suppressed the proteins involved in cancer proliferation (COX-2, C-myc, cyclin D1), migration and invasion (MMP-2, MMP-9), anti-apoptosis (XIAP, and c-IAP2), adhesion and angiogenesis (ICAM-1, VEGF). Conclusion: This study demonstrates that the anthocyanins isolated from fruits of Vitis coignetiae Pulliat inhibit cancer proliferation, cancer migration, and invasion that is involve in cancer-metastasis. This study provides evidence that AIMs might have anti-cancer effects on human lung cancer.
Collapse
Affiliation(s)
- Jing Nan Lu
- Department of Internal Medicine, Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, 660-702 Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Pachymic acid inhibits the tumorigenicity of gastric cancer cells by the mitochondrial pathway. Anticancer Drugs 2017; 28:170-179. [PMID: 27792037 DOI: 10.1097/cad.0000000000000449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Pachymic acid (PA), a lanostane-type triterpenoid derived from traditional Chinese herbals, has been reported to have antitumor activity in versatile cancer cells. However, the antitumor effect of PA in gastric cancer (GC) cells remains unclear. In this study, we aimed to explore the efficacy and mechanisms of PA in GC. The antiproliferative effect of PA was assessed by a growth assay and a colony formation assay. Flow cytometry was used to detect changes in cell cycle distribution. Apoptosis was assessed by an annexin V/propidium iodide double-staining assay. The expressions of the apoptosis-related proteins were measured by western blot. The mitochondrial capacity was observed by immunostaining of Mito Tracker Red and mitochondrial function protein MT. Xenograft models of GC were constructed by a subcutaneous injection of SGC-7901 and MKN-49P cells pretreated with PA. PA could potently inhibit GC cell growth and colony formation. PA significantly induced G1, G2/M, and inhibited G0 phase arrest in GC cell lines SGC-7901 and MKN-49P. PA induced cell apoptosis by regulating the expressions of apoptosis-related proteins (caspase-3, PARP, Bcl-2, and Bax) and suppressing the mitochondrial capacity of GC cell lines in vitro in a dose-dependent manner. In addition, PA suppressed the tumor growth of xenograft models of GC and prolonged the survival of animals markedly. In brief, the present study shows that PA induces apoptosis through inhibition of mitochondrial capacity in human GC cells. Our findings suggest that PA may have therapeutic potential in GC.
Collapse
|
37
|
The anti-tumor effect of pachymic acid on osteosarcoma cells by inducing PTEN and Caspase 3/7-dependent apoptosis. J Nat Med 2017; 72:57-63. [DOI: 10.1007/s11418-017-1117-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/21/2017] [Indexed: 10/19/2022]
|
38
|
Can Z, Lele S, Zhirui Z, Qiong P, Yuzhong C, Lingling L, Surong Z, Yiming S, Pei Z, Chenchen J, Liu H. 3-Bromopyruvate enhances TRAIL-induced apoptosis in human nasopharyngeal carcinoma cells through CHOP-dependent upregulation of TRAIL-R2. Anticancer Drugs 2017; 28:739-749. [PMID: 28471808 DOI: 10.1097/cad.0000000000000502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Past reports have shown that the sensitivity of cancer cells to TRAIL-induced apoptosis is related to their expression of TRAIL-death receptors on the cell surface. However, the level of TRAIL-death receptors expression on cancer cells is always low. Our previous research showed that nasopharyngeal carcinoma (NPC) cells have a poor sensitivity to low doses of TRAIL. Here, we evaluated combined treatment with the energy inhibitor 3-bromopyruvate (3BP) and TRAIL as a method to produce an increased apoptotic response in NPC cells. The results showed that 3BP and TRAIL together produced higher cytotoxicity and increased TRAIL-R2 expression in NPC cells compared with the effects of either 3BP or TRAIL alone. These findings led us to hypothesize that 3BP may sensitize NPC cells to TRAIL. 3BP is a metabolic blocker that inhibits hexokinase II activity, suppresses ATP production, and induces endoplasmic reticulum (ER) stress. Our results showed that 3BP also activated AMP-activated protein kinase, which we found to play an important role in the induction of ER stress by 3BP. Furthermore, the induction of TRAIL-R2 expression and the sensitization of the NPC cells to TRAIL by 3BP were reduced when we inhibited the expression of CHOP. Taken together, our results showed that a low dose of 3BP sensitized NPC cells to TRAIL-induced apoptosis by the upregulation of CHOP, which was mediated by the activation of AMP-activated protein kinase and ER stress. The results showed that 3BP is a promising candidate agent for enhancing the therapeutic response to TRAIL in NPC.
Collapse
Affiliation(s)
- Zhou Can
- aFaculty of Pharmacy, Bengbu Medical College bDepartment of Pharmacy cDepartment of Surgical Oncology, The First Affiliated Hospital of Bengbu Medical College dDepartment of Pharmacy, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China eSchool of Medicine and Public Health, Faculty of Health, Newcastle University, Newcastle, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhang YH, Zhang Y, Li XY, Feng XD, Jian W, Li RQ. Antitumor activity of the pachymic acid in nasopharyngeal carcinoma cells. Ultrastruct Pathol 2017; 41:245-251. [PMID: 28414554 DOI: 10.1080/01913123.2017.1296522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yan-Hua Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Yong Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Xiu-Ying Li
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Xu-Dong Feng
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Wei Jian
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| | - Rong-Qing Li
- Department of Radiation Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, P.R. China
| |
Collapse
|
40
|
Lee WS, Jung JH, Panchanathan R, Yun JW, Kim DH, Kim HJ, Kim GS, Ryu CH, Shin SC, Hong SC, Choi YH, Jung JM. Ursodeoxycholic Acid Induces Death Receptor-mediated Apoptosis in Prostate Cancer Cells. J Cancer Prev 2017; 22:16-21. [PMID: 28382282 PMCID: PMC5380185 DOI: 10.15430/jcp.2017.22.1.16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/17/2017] [Accepted: 02/17/2017] [Indexed: 12/03/2022] Open
Abstract
Background Bile acids have anti-cancer properties in a certain types of cancers. We determined anticancer activity and its underlying molecular mechanism of ursodeoxycholic acid (UDCA) in human DU145 prostate cancer cells. Methods Cell viability was measured with an MTT assay. UDCA-induced apoptosis was determined with flow cytometric analysis. The expression levels of apoptosis-related signaling proteins were examined with Western blotting. Results UDCA treatment significantly inhibited cell growth of DU145 in a dose-dependent manner. It induced cellular shrinkage and cytoplasmic blebs and accumulated the cells with sub-G1 DNA contents. Moreover, UDCA activated caspase 8, suggesting that UDCA-induced apoptosis is associated with extrinsic pathway. Consistent to this finding, UDCA increased the expressions of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor, death receptor 4 (DR4) and death receptor 5 (DR5), and TRAIL augmented the UDCA-induced cell death in DU145 cells. In addition, UDCA also increased the expressions of Bax and cytochrome c and decreased the expression of Bcl-xL in DU145 cells. This finding suggests that UDCA-induced apoptosis may be involved in intrinsic pathway. Conclusions UDCA induces apoptosis via extrinsic pathway as well as intrinsic pathway in DU145 prostate cancer cells. UDCA may be a promising anti-cancer agent against prostate cancer.
Collapse
Affiliation(s)
- Won Sup Lee
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea; Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| | - Ji Hyun Jung
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea; Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| | - Radha Panchanathan
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea; Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| | - Jeong Won Yun
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea; Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| | - Dong Hoon Kim
- Department of Emergency Medicine, Gyeongsang National University School of Medicine, Jinju, Korea; Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| | - Hye Jung Kim
- Department of Pharmacology, Gyeongsang National University School of Medicine, Jinju, Korea; Institute of Health Sciences, Gyeongsang National University, Jinju, Korea
| | - Gon Sup Kim
- School of Veterinary Medicine, Gyeongsang National University, Jinju, Korea; Research Institute of Life Science, Gyeongsang National University, Jinju, Korea
| | - Chung Ho Ryu
- Division of Applied Life Science (BK 21 Program), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, Korea
| | - Sung Chul Shin
- Research Institute of Life Science, Gyeongsang National University, Jinju, Korea; Department of Chemistry, College of Natural Sciences, Gyeongsang National University, Jinju, Korea
| | - Soon Chan Hong
- Institute of Health Sciences, Gyeongsang National University, Jinju, Korea; Department of Surgery, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan, Korea; Anti-Aging Research Center & Blue-Bio Industry RIC, Dongeui University, Busan, Korea
| | - Jin-Myung Jung
- Institute of Health Sciences, Gyeongsang National University, Jinju, Korea; Department of Neurosurgery, Gyeongsang National University School of Medicine, Jinju, Korea
| |
Collapse
|
41
|
Nagappan A, Lee WS, Yun JW, Lu JN, Chang SH, Jeong JH, Kim GS, Jung JM, Hong SC. Tetraarsenic hexoxide induces G2/M arrest, apoptosis, and autophagy via PI3K/Akt suppression and p38 MAPK activation in SW620 human colon cancer cells. PLoS One 2017; 12:e0174591. [PMID: 28355296 PMCID: PMC5371332 DOI: 10.1371/journal.pone.0174591] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/11/2017] [Indexed: 01/11/2023] Open
Abstract
Tetraarsenic hexoxide (As4O6) has been used in Korean folk medicines for the treatment of cancer, however its anti-cancer mechanisms remain obscured. Here, this study investigated the anti-cancer effect of As4O6 on SW620 human colon cancer cells. As4O6 has showed a dose-dependent inhibition of SW620 cells proliferation. As4O6 significantly increased the sub-G1 and G2/M phase population, and Annexin V-positive cells in a dose-dependent manner. G2/M arrest was concomitant with augment of p21 and reduction in cyclin B1, cell division cycle 2 (cdc 2) expressions. Nuclear condensation, cleaved nuclei and poly (adenosine diphosphate‑ribose) polymerase (PARP) activation were also observed in As4O6-treated SW620 cells. As4O6 induced depolarization of mitochondrial membrane potential (MMP, ΔΨm) but not reactive oxygen species (ROS) generation. Further, As4O6 increased death receptor 5 (DR5), not DR4 and suppressed the B‑cell lymphoma‑2 (Bcl-2) and X-linked inhibitor of apoptosis protein (XIAP) family proteins. As4O6 increased the formation of AVOs (lysosomes and autophagolysosomes) and promoted the conversion of microtubule-associated protein 1A/1B-light chain 3 (LC3)-I to LC3-II in a dose- and time- dependent manner. Interestingly, a specific phosphoinositide 3-kinase (PI3K)/Akt inhibitor (LY294002) augmented the As4O6 induced cell death; whereas p38 mitogen-activated protein kinases (p38 MAPK) inhibitor (SB203580) abrogated the cell death. Thus, the present study provides the first evidence that As4O6 induced G2/M arrest, apoptosis and autophagic cell death through PI3K/Akt and p38 MAPK pathways alteration in SW620 cells.
Collapse
Affiliation(s)
- Arulkumar Nagappan
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, 90 Chilam-dong Jinju, Korea
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, 90 Chilam-dong Jinju, Korea
- * E-mail: ,
| | - Jeong Won Yun
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, 90 Chilam-dong Jinju, Korea
| | - Jing Nan Lu
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, 90 Chilam-dong Jinju, Korea
| | - Seong-Hwan Chang
- Department of Surgery, Konkuk University School of Medicine, Seoul, Korea
| | - Jae-Hoon Jeong
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, 900 Gajwadong, Jinju, Korea
| | - Jin-Myung Jung
- Department of Neurosurgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, 90 Chilam-dong Jinju, Korea
| | - Soon Chan Hong
- Department of Surgery, Institute of Health Sciences, Gyeongsang National University School of Medicine, 90 Chilam-dong Jinju, Korea
| |
Collapse
|
42
|
Secondary Metabolites from Higher Fungi. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 106 2017; 106:1-201. [DOI: 10.1007/978-3-319-59542-9_1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
43
|
Farooqi AA, Gadaleta CD, Ranieri G, Fayyaz S, Marech I. New Frontiers in Promoting TRAIL-Mediated Cell Death: Focus on Natural Sensitizers, miRNAs, and Nanotechnological Advancements. Cell Biochem Biophys 2016; 74:3-10. [PMID: 26972296 DOI: 10.1007/s12013-015-0712-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer is a multifaceted and genomically complex disease, and rapidly emerging scientific evidence is emphasizing on intra-tumor heterogeneity within subpopulations of tumor cells and rapidly developing resistance against different molecular therapeutics. There is an overwhelmingly increasing list of agents currently being tested for efficacy against cancer. In accordance with the concept that therapeutic agents must have fewer off target effects and considerable efficacy, TRAIL has emerged as one among the most deeply investigated proteins reportedly involved in differential killing of tumor cells. Considerable killing activity of TRAIL against different cancers advocated its entry into clinical trials. However, data obtained through preclinical and cell culture studies are deepening our understanding of wide-ranging mechanisms which induce resistance against TRAIL-based therapeutics. These include downregulation of death receptors, overexpression of oncogenes, inactivation of tumor suppressor genes, imbalance of pro- and anti-apoptotic proteins, and inactivation of intrinsic and extrinsic pathways. Substantial fraction of information has been added into existing pool of knowledge related to TRAIL biology and recently accumulating evidence is adding new layers to regulation of TRAIL-induced apoptosis. Certain hints have emerged underscoring miR135a-3p- and miR-143-mediated regulation of TRAIL-induced apoptosis, and natural agents have shown remarkable efficacy in improving TRAIL-based therapeutics by increasing expression of tumor suppressor miRNAs. In this review, we summarize most recent breakthroughs related to naturopathy and strategies to nanotechnologically deliver TRAIL to the target site in xenografted mice. We also set spotlight on positive and negative regulators of TRAIL-mediated signaling. Comprehensive knowledge of genetics and proteomics of TRAIL-based signaling network obtained from cancer patients of different populations will be helpful in getting a step closer to personalized medicine.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad, Pakistan.
| | - Cosmo Damiano Gadaleta
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Girolamo Ranieri
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Sundas Fayyaz
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | - Ilaria Marech
- Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
44
|
Nagappan A, Venkatarame Gowda Saralamma V, Hong GE, Lee HJ, Shin SC, Kim EH, Lee WS, Kim GS. Proteomic analysis of selective cytotoxic anticancer properties of flavonoids isolated from Citrus platymamma on A549 human lung cancer cells. Mol Med Rep 2016; 14:3814-22. [PMID: 27573346 DOI: 10.3892/mmr.2016.5666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 06/29/2016] [Indexed: 11/05/2022] Open
Abstract
Citrus platymamma Hort. ex Tanaka (Byungkyul in Korean) has been used in Korean folk medicine for the treatment of inflammatory disorders and cancer. However, the molecular mechanism underlying the anticancer properties of flavonoids isolated from C. platymamma (FCP) remains to be elucidated. Therefore, the present study attempted to identify the key proteins, which may be important in the anticancer effects of FCP on A549 cells using a proteomic approach. FCP showed a potent cytotoxic effect on the A549 human lung cancer cells, however, it had no effect on WI‑38 human fetal lung fibroblasts at the same concentrations. Furthermore, 15 differentially expressed protein spots (spot intensities ≥2‑fold change; P<0.05) were obtained from comparative proteome analysis of two‑dimensional gel electrophoresis maps of the control (untreated) and FCP‑treated A549 cells. Finally, eight differentially expressed proteins, one of which was upregulated and seven of which were downregulated, were successfully identified using matrix‑assisted laser desorption/ionization time‑of‑flight/time‑of‑flight tandem mass spectrometry and peptide mass fingerprinting analysis. Specifically, proteins involved in signal transduction were significantly downregulated, including annexin A1 (ANXA1) and ANXA4, whereas 14‑3‑3ε was upregulated. Cytoskeletal proteins, including cofilin‑1 (CFL1), cytokeratin 8 (KRT8) and KRT79, and molecular chaperones/heat shock proteins, including endoplasmin, were downregulated. Proteins involved in protein metabolism, namely elongation factor Ts were also downregulated. Consistent with results of the proteome analysis, the immunoblotting results showed that 14‑3‑3ε was upregulated, whereas CFL1, ANXA4 and KRT8 were downregulated in the FCP‑treated A549 cells. The majority of the proteins were involved in tumor growth, cell cycle, apoptosis, migration and signal transduction. These findings provide novel insights into the molecular mechanisms underlying FCP-induced anticancer effects on A549 cells.
Collapse
Affiliation(s)
- Arulkumar Nagappan
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Gyeongnam 660‑702, Republic of Korea
| | - Venu Venkatarame Gowda Saralamma
- Research Institute of Life Science and College of Veterinary Medicine (BK21 Plus Project), Gyeongsang National University, Jinju, Gyeongnam 660‑701, Republic of Korea
| | - Gyeong Eun Hong
- Research Institute of Life Science and College of Veterinary Medicine (BK21 Plus Project), Gyeongsang National University, Jinju, Gyeongnam 660‑701, Republic of Korea
| | - Ho Jeong Lee
- Research Institute of Life Science and College of Veterinary Medicine (BK21 Plus Project), Gyeongsang National University, Jinju, Gyeongnam 660‑701, Republic of Korea
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju, Gyeongnam 660‑701, Republic of Korea
| | - Eun Hee Kim
- Department of Nursing Science, International University of Korea, Jinju, Gyeongnam 660‑759, Republic of Korea
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, Gyeongsang National University School of Medicine, Jinju, Gyeongnam 660‑702, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine (BK21 Plus Project), Gyeongsang National University, Jinju, Gyeongnam 660‑701, Republic of Korea
| |
Collapse
|
45
|
Nagappan A, Lee HJ, Saralamma VVG, Park HS, Hong GE, Yumnam S, Raha S, Charles SN, Shin SC, Kim EH, Lee WS, Kim GS. Flavonoids isolated from Citrus platymamma induced G2/M cell cycle arrest and apoptosis in A549 human lung cancer cells. Oncol Lett 2016; 12:1394-1402. [PMID: 27446443 PMCID: PMC4950876 DOI: 10.3892/ol.2016.4793] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 04/29/2016] [Indexed: 12/28/2022] Open
Abstract
Citrus platymamma hort. ex Tanaka belongs to the Rutaceae family and is widely used in folk medicines in Korea due to its anti-proliferative, anti-cancer, anti-oxidant, anti-inflammatory and anti-diabetic activities. However, the molecular mechanism of its anti-cancer effect is not well understood. The present study was conducted to elucidate the anti-cancer effect and molecular mechanism of flavonoids from Citrus platymamma (FCP) on A549 cells. FCP displayed concentration-dependent inhibition on A549 cells proliferation. Further, flow cytometry revealed that FCP significantly increased the sub-G1 (apoptotic cell population) and G2/M phase population, and the total number of apoptotic cells, in a dose-dependent manner. Nuclear condensation and fragmentation were also observed upon staining with Hoechst 33342 in FCP-treated A549 cells. Immunoblotting demonstrated a dose-dependent downregulation of cyclin B1, cyclin-dependent kinase 1, cell division cycle 25c, pro-caspases −3, −6, −8 and −9, and poly (adenosine diphosphate-ribose) polymerase (PARP) in FCP-treated A549 cells. In addition, FCP induced caspase-3 activation and subsequent PARP cleavage, and increased the B-cell lymphoma (Bcl)-2-associated X protein/Bcl-extra large ratio in A549 cells. These findings suggest that FCP induced G2/M arrest and apoptosis of A549 cells. The present study provides evidence that FCP may be useful in the treatment of human lung cancer.
Collapse
Affiliation(s)
- Arulkumar Nagappan
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, School of Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-702, Republic of Korea
| | - Ho Jeong Lee
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Venu Venkatarame Gowda Saralamma
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Hyeon Soo Park
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Gyeong Eun Hong
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Silvia Yumnam
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Suchismita Raha
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Shobana Nancy Charles
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, School of Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-702, Republic of Korea
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| | - Eun Hee Kim
- Department of Nursing Science, International University of Korea, Jinju, Gyeongnam 660-759, Republic of Korea
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences and Gyeongnam Regional Cancer Center, School of Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-702, Republic of Korea
| | - Gon Sup Kim
- Brain Korea 21 Program for Leading Universities and Students, Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongnam 660-701, Republic of Korea
| |
Collapse
|