1
|
Liao Y, Wei F, He Z, He J, Ai Y, Guo C, Zhou L, Luo D, Li C, Wen Y, Zeng J, Ma X. Animal-derived natural products for hepatocellular carcinoma therapy: current evidence and future perspectives. Front Pharmacol 2024; 15:1399882. [PMID: 38803433 PMCID: PMC11129636 DOI: 10.3389/fphar.2024.1399882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) has a high morbidity and mortality rate, and the survival rate of HCC patients remains low. Animal medicines have been used as potential therapeutic tools throughout the long history due to their different structures of biologically active substances with high affinity to the human body. Here, we focus on the effects and the mechanism of action of animal-derived natural products against HCC, which were searched in databases encompassing Web of Science, PubMed, Embase, Science Direct, Springer Link, and EBSCO. A total of 24 natural products from 12 animals were summarized. Our study found that these natural products have potent anti-hepatocellular carcinoma effects. The mechanism of action involving apoptosis induction, autophagy induction, anti-proliferation, anti-migration, and anti-drug resistance via phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR), Ras/extracellular signal regulated kinases (ERK)/mitogen-activated protein kinase (MAPK), Wnt/β-catenin, and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways. Huachansu injection and sodium cantharidate have been used in clinical applications with good efficacy. We review the potential of animal-derived natural products and their derivatives in the treatment of HCC to date and summarize their application prospect and toxic side effects, hoping to provide a reference for drug development for HCC.
Collapse
Affiliation(s)
- Yichao Liao
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Wei
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhelin He
- Endoscopy Center, Guang’an Hospital of Traditional Chinese Medicine, Guang’an, China
| | - Jingxue He
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanlin Ai
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cui Guo
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Luo
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chengen Li
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
2
|
Pan MS, Cao J, Fan YZ. Insight into norcantharidin, a small-molecule synthetic compound with potential multi-target anticancer activities. Chin Med 2020; 15:55. [PMID: 32514288 PMCID: PMC7260769 DOI: 10.1186/s13020-020-00338-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
Norcantharidin (NCTD) is a demethylated derivative of cantharidin, which is an anticancer active ingredient of traditional Chinese medicine, and is currently used clinically as a routine anti-cancer drug in China. Clarifying the anticancer effect and molecular mechanism of NCTD is critical for its clinical application. Here, we summarized the physiological, chemical, pharmacokinetic characteristics and clinical applications of NCTD. Besides, we mainly focus on its potential multi-target anticancer activities and underlying mechanisms, and discuss the problems existing in clinical application and scientific research of NCTD, so as to provide a potential anticancer therapeutic agent for human malignant tumors.
Collapse
Affiliation(s)
- Mu-Su Pan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Jin Cao
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Yue-Zu Fan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| |
Collapse
|
3
|
Chi J, Jiang Z, Qiao J, Zhang W, Peng Y, Liu W, Han B. Antitumor evaluation of carboxymethyl chitosan based norcantharidin conjugates against gastric cancer as novel polymer therapeutics. Int J Biol Macromol 2019; 136:1-12. [PMID: 31158420 DOI: 10.1016/j.ijbiomac.2019.05.216] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
Novel polymer-drug conjugates (CNC) were prepared from carboxymethyl chitosan (CMCS) and norcantharidin (NCTD) via amidation reaction and characterized by FTIR and 1H NMR spectroscopy. The aim of this study was to elucidate the antitumor efficacy of CNC on gastric cancer and the possible underlying mechanisms. The CNC conjugates possessed significant inhibitory effects on the proliferation of SGC-7901 cells and suppressed the migration as well as tube formation of HUVECs. Besides, Hoechst 33258 staining and Annexin V-FITC/PI detection suggested that the conjugates were more effective in triggering apoptosis of SGC-7901 cells compared with free NCTD. Moreover, CNC remarkably reduced systemic toxicity and enhanced the antitumor efficacy in vivo with a tumor suppression rate of 59.57% against SGC-7901 gastric tumor in BALB/c nude mice. Further investigation about the underlying mechanisms indicated that CNC could upregulate expressions of TNF-α and Bax, and downregulate expressions of VEGF, Bcl-2, MMP-2 and MMP-9, thereby inhibiting tumor metastasis and inducing apoptosis in vivo. Overall, our results demonstrated that CNC might be a promising and feasible polymer therapeutics for gastrointestinal tumor therapy.
Collapse
Affiliation(s)
- Jinhua Chi
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zhiwen Jiang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China
| | - Jing Qiao
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Wei Zhang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Yanfei Peng
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China
| | - Wanshun Liu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Baoqin Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China.
| |
Collapse
|
4
|
Chen F, Wang S, Wei Y, Wu J, Huang G, Chen J, Shi J, Xia J. Norcantharidin modulates the miR-30a/Metadherin/AKT signaling axis to suppress proliferation and metastasis of stromal tumor cells in giant cell tumor of bone. Biomed Pharmacother 2018; 103:1092-1100. [DOI: 10.1016/j.biopha.2018.04.100] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/24/2022] Open
|
5
|
Mo L, Zhang X, Shi X, Wei L, Zheng D, Li H, Gao J, Li J, Hu Z. Norcantharidin enhances antitumor immunity of GM-CSF prostate cancer cells vaccine by inducing apoptosis of regulatory T cells. Cancer Sci 2018; 109:2109-2118. [PMID: 29770533 PMCID: PMC6029826 DOI: 10.1111/cas.13639] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 12/20/2022] Open
Abstract
Norcantharidin (NCTD) is a promising antitumor drug with low toxicity. It was reported to be able to regulate immunity, but the mechanism is not yet clear. Here we explored whether NCTD could enhance the antitumor immunity induced by prostate cancer cell vaccine. The results of the in vitro study showed that NCTD induced apoptosis and inhibited proliferation of regulatory T cells (Tregs). Mechanistic research showed that NCTD inhibited Akt activation and activated FOXO1 transcription, resulting in a pro‐apoptotic effect. The results of the in vivo study showed that more tumor‐infiltrating Tregs existed within peripheral blood and tumor tissue after treatment with the vaccine. Adding NCTD to vaccine treatment could decrease the number of tumor‐infiltrating Tregs and increase the number of CD4+ and CD8+ T cells. Combination therapy with NCTD and vaccine was more effective in inhibiting tumor growth than the vaccine alone. In general, this is the first report that NCTD could induce apoptosis of Tregs and enhance the vaccine‐induced immunity.
Collapse
Affiliation(s)
- Lijun Mo
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xinji Zhang
- Department of Urology, Shunde Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojun Shi
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Wei
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Dianpeng Zheng
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Hongwei Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jinlong Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhiming Hu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Guo J, Wu Y, Du J, Yang L, Chen W, Gong K, Dai J, Miao S, Jin D, Xi S. Deregulation of UBE2C-mediated autophagy repression aggravates NSCLC progression. Oncogenesis 2018; 7:49. [PMID: 29904125 PMCID: PMC6002383 DOI: 10.1038/s41389-018-0054-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/12/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023] Open
Abstract
The roles of aberrantly regulated autophagy in human malignancy and the mechanisms that initiate and sustain the repression of autophagy in carcinogenesis are less well defined. Activation of the oncogene UBE2C and repression of autophagy are concurrently underlying the initiation, progression, and metastasis of lung cancer and exploration of essential association of UBE2C with autophagy will confer more options in searching novel molecular therapeutic targets in lung cancer. Here we report that aberrant activation of UBE2C in lung tumors from patients associates with adverse prognosis and enhances cell proliferation, clonogenicity, and invasive growth of NSCLC. UBE2C selectively represses autophagy in NSCLC and disruption of UBE2C-mediated autophagy repression attenuates cell proliferation, clonogenicity, and invasive growth of NSCLC. Autophagy repression is essentially involved in UBE2C-induced cell proliferation, clonogenicity, and invasive growth of NSCLC. Interference of UBE2C-autophagy repression axis by Norcantharidin arrests NSCLC progression. UBE2C is repressed post-transcriptionally via tumor suppressor miR-381 and epitranscriptionally stabilized with maintenance of lower m6A level within its mature RNAs due to the upregulation of m6A demethylase ALKBH5 in NSCLC. Collectively, our results indicated that deregulated UBE2C-autophagy repression axis drives NSCLC progression which renders varieties of potential molecular targets in cancer therapy of NSCLC.
Collapse
Affiliation(s)
- Jiwei Guo
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Yan Wu
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Jing Du
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Lijuan Yang
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Weiwei Chen
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Kaikai Gong
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Juanjuan Dai
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Shuang Miao
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Dan Jin
- Department of Pain Ward, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Sichuan Xi
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China.
| |
Collapse
|
7
|
YAP and TAZ in Lung Cancer: Oncogenic Role and Clinical Targeting. Cancers (Basel) 2018; 10:cancers10050137. [PMID: 29734788 PMCID: PMC5977110 DOI: 10.3390/cancers10050137] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/17/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in the world and there is no current treatment able to efficiently treat the disease as the tumor is often diagnosed at an advanced stage. Moreover, cancer cells are often resistant or acquire resistance to the treatment. Further knowledge of the mechanisms driving lung tumorigenesis, aggressiveness, metastasization, and resistance to treatments could provide new tools for detecting the disease at an earlier stage and for a better response to therapy. In this scenario, Yes Associated Protein (YAP) and Trascriptional Coactivator with PDZ-binding motif (TAZ), the final effectors of the Hippo signaling transduction pathway, are emerging as promising therapeutic targets. Here, we will discuss the most recent advances made in YAP and TAZ biology in lung cancer and, more importantly, on the newly discovered mechanisms of YAP and TAZ inhibition in lung cancer as well as their clinical implications.
Collapse
|
8
|
Guo J, Wu Y, Yang L, Du J, Gong K, Chen W, Dai J, Li X, Xi S. Repression of YAP by NCTD disrupts NSCLC progression. Oncotarget 2018; 8:2307-2319. [PMID: 27903989 PMCID: PMC5356801 DOI: 10.18632/oncotarget.13668] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/21/2016] [Indexed: 11/25/2022] Open
Abstract
The efficacy of available lung cancer therapeutic interference is significantly limited by various resistance mechanisms to those drugs. Activation of the oncogene YAP underlying the initiation, progression, and metastasis of lung cancer associates with poor prognosis and confers drug resistance against targeted therapy. In this study, we evaluated the specificity of norcantharidin (NCTD) in repressing YAP to inhibit non-small cell lung carcinoma (NSCLC) progression. Our study revealed that YAP signal pathways were aberrantly activated in lung cancer tissues and cells which rendered more proliferative and invasive phenotypes to human lung cancer cells. We confirmed that NCTD specifically repressed YAP signaling pathway to interfere the YAP-mediated non-small cell lung carcinoma progression and metastasis via arresting cell cycle, enhancing apoptosis and inducing senescence. We also found NCTD-mediated repression of YAP decreased epithelial-to-mesenchymal transition (EMT) and reduced the motile and invasive cellular phenotype in vitro via enhancing E-cadherin and decreasing fibronectin/vimentin. Mechanistic investigations revealed that NCTD transcriptionally downregulated YAP and post-translationally modulated the subcellular redistribution of YAP between nucleus and cytoplasm. Collectively, our results indicated that NCTD is a novel therapeutic drug candidate for NSCLC which specifically and sensitively target YAP signal pathway.
Collapse
Affiliation(s)
- Jiwei Guo
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - Yan Wu
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - Lijuan Yang
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - Jing Du
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - Kaikai Gong
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - Weiwei Chen
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - Juanjuan Dai
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - XueLin Li
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| | - Sichuan Xi
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou 256603, P.R. China
| |
Collapse
|
9
|
Jayasooriya RGPT, Molagoda IMN, Park C, Jeong JW, Choi YH, Moon DO, Kim MO, Kim GY. Molecular chemotherapeutic potential of butein: A concise review. Food Chem Toxicol 2017; 112:1-10. [PMID: 29258953 DOI: 10.1016/j.fct.2017.12.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/13/2022]
Abstract
Butein is a biologically active flavonoid isolated from the bark of Rhus verniciflua Stokes, which is known to have therapeutic potential against various cancers. Notably, butein inhibits cancer cell growth by inducing G2/M phase arrest and apoptosis. Butein-induced G2/M phase arrest is associated with increased phosphorylation of ataxia telangiectasia mutated (ATM) and Chk1/2, and consequently, with reduced cdc25C levels. In addition, butein-induced apoptosis is mediated through the activation of caspase-3, which is associated with changes in the expression of Bcl-2 and Bax proteins. Intriguingly, butein sensitizes cells to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis via ERK-mediated Sp1 activation, which promotes the transcription of specific death receptor 5. Butein also inhibits the migration and invasion of human cancer cells by suppressing nuclear factor-κB- and extracellular signal-regulated kinases 1/2-mediated expression of matrix metalloproteinase-9 and vascular endothelial growth factor. Additionally, butein downregulates the expression of human telomerase reverse transcriptase and causes a concomitant decrease in telomerase activity. These findings provide the basis for the pharmaceutical development of butein. The aim of this review is to provide an update on the mechanisms underlying the anticancer activity of butein, with a special focus on its effects on different cellular signaling cascades.
Collapse
Affiliation(s)
- Rajapaksha Gedara Prasad Tharanga Jayasooriya
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea; Department of Biological Sciences, Faculty of Applied Science, University of Rajarata, Mihintale 50300, Sri Lanka
| | | | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences and Human Ecology, Dongeui University, Busan 67340, Republic of Korea
| | - Jin-Woo Jeong
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Dong-Oh Moon
- Department of Biology Education, Daegu University, Jillyang, Gyeongsan, Gyeonsangbuk-do 38453, Republic of Korea
| | - Mun-Ock Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Chungcheongbuk-do 28116, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
10
|
Lin CL, Chen CM, Lin CL, Cheng CW, Lee CH, Hsieh YH. Norcantharidin induces mitochondrial-dependent apoptosis through Mcl-1 inhibition in human prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1867-1876. [PMID: 28760656 DOI: 10.1016/j.bbamcr.2017.07.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/20/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022]
Abstract
Norcantharidin (NCTD) is the demethylated form of cantharidin that exhibits anticancer potential in many cancer cell types. Recent reports suggest that NCTD targeting ROS/AMPK and DNA replication signaling pathway could be an effective strategy for the treatment of PCa cells. However, supportive evidence is limited to the effect of NCTD that induction of apoptosis through suppression of the Mcl-1. Here, we show that NCTD induced PCa cell apoptosis and triggered caspase activation, which was associated with mitochondria dysfunction. Mechanistic investigations suggested that NCTD modulated the Akt signaling via increased nuclear translocation and interaction with the myeloid cell leukemia-1 (Mcl-1) promoter by FOXO4, resulting in an apoptotic effect. Moreover, miR-320d, which targets Mcl-1, was significantly upregulated after NCTD treatment. Overexpression of miR-320d by NCTD induced mitochondria dysfunction and apoptosis, which was notably attenuated with a miR-320d inhibitor. In vivo xenograft analysis revealed that NCTD significantly reduced tumor growth in mice with PC3 tumor xenografts. Taken together, our results provide new insights into the critical role of NCTD in suppressing Mcl-1 via epigenetic upregulation of miR-320d, resulting in PCa cell apoptosis.
Collapse
Affiliation(s)
- Chu-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Min Chen
- Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan; School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Hsing Lee
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Division of Pediatric Surgery, Department of Surgery, Children's Hospital of China Medical University, Taichung. Taiwan.
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan; Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Clinical laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
11
|
Tao R, Wang ZF, Qiu W, He YF, Yan WQ, Sun WY, Li HJ. Role of S100A3 in human hepatocellular carcinoma and the anticancer effect of sodium cantharidinate. Exp Ther Med 2017; 13:2812-2818. [PMID: 28588665 PMCID: PMC5450779 DOI: 10.3892/etm.2017.4294] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 11/15/2016] [Indexed: 12/11/2022] Open
Abstract
The fifth most common cancer worldwide is hepatocellular carcinoma (HCC), which has an annual mortality rate of ~800,000. Although surgical procedures for HCC, such as hepatic resection and liver transplantation, have progressed and the outcomes of patients have improved, HCC is still characterized by frequent recurrence, even after liver transplantation. In the present study the expression of the protein coding gene, S100 calcium binding protein A3 (S100A3), was observed in 62 HCC tissues and tumor-surrounding tissues. The present study indicated that S100A3 activation was involved in tumorigenesis and tumor aggressiveness. The protein and mRNA expression levels of S100A3 in the human HCC cell line (HepG2) were investigated using western blotting and reverse transcription-quantitative polymerase chain reaction analysis, respectively. The function of sodium cantharidinate in inducing HCC cell apoptosis was also investigated. Sodium cantharidinate inhibited the protein and gene expression of S100A3 in HepG2 cells in vitro. These data suggested that S100A3 has an important role in human HCC. The present study indicates that the functional properties of sodium cantharidinate are promising for the development of a novel drug that may control the expression of S100A3 and improve the treatment of human HCC in the near future.
Collapse
Affiliation(s)
- Ran Tao
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China.,Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhong-Feng Wang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Qiu
- Department of Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yu-Fang He
- Institute of Phytochemistry, Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130012, P.R. China
| | - Wei-Qun Yan
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wen-Yi Sun
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hai-Jun Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
12
|
Han Z, Li B, Wang J, Zhang X, Li Z, Dai L, Cao M, Jiang J. Norcantharidin Inhibits SK-N-SH Neuroblastoma Cell Growth by Induction of Autophagy and Apoptosis. Technol Cancer Res Treat 2017; 16:33-44. [PMID: 26755751 PMCID: PMC5616112 DOI: 10.1177/1533034615624583] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/01/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022] Open
Abstract
Norcantharidin, a low-toxic analog of the active anticancer compound cantharidin in Mylabris, can inhibit proliferation and induce apoptosis of multiple types of cancer cells. However, the anticancer activities of norcantharidin with respect to neuroblastoma, and its underlying mechanisms, have not been investigated. Therefore, our study was designed to determine the efficacy of norcantharidin on SK-N-SH neuroblastoma cell death and to elucidate detailed mechanisms of activity. In the present study, norcantharidin suppressed the proliferation and cloning ability of SK-N-SH cells in a dose-dependent manner, apparently by reducing the mitochondrial membrane potential and arresting SK-N-SH cells at the G2/M stage, accompanied by elevated expressions of p21 and decreased expressions of cyclin B1 and cell division control 2. Treatment by norcantharidin induced significant mitophagy and autophagy, as demonstrated by a decrease in Translocase Of Outer Mitochondrial Membrane 20 (TOM20), increased beclin1 and LC3-II protein expression, reduced protein SQSTM1/p62 expression, and accumulation of punctate LC3 in the cytoplasm of SK-N-SH cells. In addition, norcantharidin induced apoptosis through regulating the expression of B-cell lymphoma 2-associated X protein/B-cell lymphoma 2 and B-cell lymphoma 2-associated X protein/myeloid cell leukemia 1 and activating caspase-3 and caspase-9-dependent endogenous mitochondrial pathways. We also observed an increase in phosphor-AMP-activated protein kinase accompanied with a decrease in phosphor-protein kinase B and mammalian target of rapamycin expression after treatment with norcantharidin. Subsequent studies indicated that norcantharidin participates in cellular autophagy and apoptosis via activation of the c-Jun NH2-terminal kinases/c-Jun pathway. In conclusion, our results demonstrate that norcantharidin can reduce the mitochondrial membrane potential, induce mitophagy, and subsequently arouse cellular autophagy and apoptosis; the AMP-activated protein kinase, protein kinase B/mammalian target of rapamycin, and c-Jun NH2-terminal kinases/c-Jun signaling pathways are widely involved in these processes. Thus, the traditional Chinese medicine norcantharidin could be a novel therapeutic strategy for treating neuroblastoma.
Collapse
Affiliation(s)
- Zeping Han
- Department of Laboratory, Central Hospital of Panyu, Guangzhou, China
| | - Baoxia Li
- State Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Juanjuan Wang
- Department of Biochemistry, Medical College, Jinan University, Guangzhou, China
| | - Xiangqiang Zhang
- Department of Biochemistry, Medical College, Jinan University, Guangzhou, China
| | - Zhenhua Li
- Department of Biochemistry, Medical College, Jinan University, Guangzhou, China
| | - Liting Dai
- Department of Biochemistry, Medical College, Jinan University, Guangzhou, China
| | - Mingrong Cao
- Department of General Surgery, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jianwei Jiang
- Department of Biochemistry, Medical College, Jinan University, Guangzhou, China
| |
Collapse
|
13
|
Yang PY, Hu DN, Kao YH, Lin IC, Chou CY, Wu YC. Norcantharidin induces apoptosis in human prostate cancer cells through both intrinsic and extrinsic pathways. Pharmacol Rep 2016; 68:874-80. [PMID: 27351942 DOI: 10.1016/j.pharep.2016.04.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/12/2016] [Accepted: 04/15/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Norcantharidin, a modified pure compound from blister beetles, was previously demonstrated to induce apoptosis of cancer cells. This study investigated its anti-cancer activity in prostate cancer cells and the mechanisms involved. METHODS Two human prostate cancer cell lines, 22Rv1 and Du145, were treated with norcantharidin at concentrations ranging from 3 to 30μg/ml. Cytotoxic effect of norcantharidin was determined by use of the 3-(4,5-dimethylthiazol-yl)-diphenyl tetrazoliumbromide (MTT) assay. The effects of apoptosis were evaluated by cell death assay, Caspase-3, -8, -9 activity and cytochrome c release. The apoptotic related protein expressions (Bcl-2 family and inhibitor of apoptosis proteins) were determined using western blotting. RESULTS An MTT assay revealed that norcantharidin induced cytotoxicity against both prostate cancer cells in dose- and time-dependent manners. Treatment with norcantharidin at 3μg/ml or higher significantly increased oligonucleosomal formation with concomitant appearance of PARP cleavage, implicating the induction of apoptosis. Norcantharidin intrinsically elevated cytosolic cytochrome c levels and activated caspase-3, -8, and -9. Extrinsically, it upregulated the expression of not only the death receptors Fas and DR5 in 22Rv1 cells, but also of RIP and TRADD adaptor proteins in Du145 cells. Mechanistically, norcantharidin increased ratios of pro-/anti-apoptotic proteins and decreased expression of IAP family member proteins, including cIAP1 and survivin, regardless of the distinct status of androgen receptor expression in both cells. CONCLUSIONS Norcantharidin exhibited cytotoxicity against 22Rv1 and Du145 prostate cancer cells by inducing both intrinsic and extrinsic apoptotic pathways and could thus potentially be a remedy for prostate cancer.
Collapse
Affiliation(s)
- Pei-Yu Yang
- Department of Medical Research, Show Chwan Memorial Hospital, Changhua, Taiwan, ROC.
| | - Dan-Ning Hu
- Tissue Culture Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY, USA.
| | - Ying-Hsien Kao
- Department of Medical Research, E-DA Hospital, Kaohsiung, Taiwan, ROC.
| | - I-Ching Lin
- Department of Family Medicine, Changhua Christian Hospital, Changhua, Taiwan, ROC; School of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC; School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC.
| | - Chih-Yuan Chou
- Division of Urology, Department of Surgery, Show-Chwan Memorial Hospital, Changhua, Taiwan, ROC.
| | - Yang-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
14
|
Hsieh MJ, Yeh CB, Chiou HL, Hsieh MC, Yang SF. Dioscorea nipponica Attenuates Migration and Invasion by Inhibition of Urokinase-Type Plasminogen Activator through Involving PI3K/Akt and Transcriptional Inhibition of NF-κB and SP-1 in Hepatocellular Carcinoma. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:177-95. [DOI: 10.1142/s0192415x16500129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
High mortality and morbidity rates for hepatocellular carcinoma (HCC) in Taiwan primarily result from uncontrolled tumor metastasis. In our previous studies, we have reported that Dioscorea nipponica Makino extract (DNE) has anti-metastasis effects on human oral cancer cells. However, the effect of DNE on hepatoma metastasis have not been thoroughly investigated and remains poorly understood. To determine the effects of DNE on the migration and invasion in HCC cells we used a wound healing model, Boyden chamber assays, gelatin/casein zymography and Western blotting. Transcriptional levels of matrix metalloproteinase-9 (MMP-9) and urokinase-type plasminogen activator (u-PA) were detected by real-time PCR and promoter assays. In this study, DNE treatment significantly inhibited the migration/invasion capacities of Huh7 cell lines. The results of gelatin/casein zymography and Western blotting revealed that the activities and protein levels of the MMP-9 and u-PA were inhibited by DNE. Tests of the mRNA levels, real-time PCR, and promoter assays evaluated the inhibitory effects of DNE on u-PA expression in human hepatoma cells. A chromatin immunoprecipitation (ChIP) assay showed not only that DNE inhibits u-PA expression, but also the inhibitory effects were associated with the down-regulation of the transcription factors of NF-[Formula: see text]B and SP-1 signaling pathways. Western blot analysis also showed that DNE inhibits PI3K and phosphorylation of Akt. In conclusion, these results show that u-PA expression may be a potent therapeutic target in the DNE-mediated suppression of HCC invasion/migration. DNE may have potential use as a chemo-preventive agent against liver cancer metastasis.
Collapse
Affiliation(s)
- Ming-Ju Hsieh
- Cancer Research Center, Changhua Christian Hospital, Changhua 50006, Taiwan
- School of Optometry, Chung Shan Medical University, Taichung 40201, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chao-Bin Yeh
- Department of Emergency Medicine, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Ming-Chang Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
15
|
Xiong X, Wu M, Zhang H, Li J, Lu B, Guo Y, Zhou T, Guo H, Peng R, Li X, Tian Q, Wang Y. Atg5 siRNA inhibits autophagy and enhances norcantharidin-induced apoptosis in hepatocellular carcinoma. Int J Oncol 2015; 47:1321-8. [PMID: 26240015 DOI: 10.3892/ijo.2015.3103] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 06/22/2015] [Indexed: 11/06/2022] Open
Abstract
Cantharidin is a terpenoid isolated from Chinese blister beetles, and norcantharidin (NCTD) is a demethylated analog of cantharidin. It has been reported that cantharidin and norcantharidin have anticancer activities. Growing evidence suggests that inhibiting autophagy can induce apoptosis in the human hepatoma cell line HepG2. The objective of the present study was to determine whether inhibition of autophagy enhances NCTD-induced apoptosis in HepG2 cells. HepG2 cells were cultured in DMEM containing NCTD. Autophagy was upregulated in the presence of HBSS media supplemented with Ca2+ and Mg2+ and 10 mM HEPES and downregulated in the presence of 3-methyladenine (3-MA) and Atg5 siRNA. Autophagy, cell viability, and the expression of apoptotic proteins were assessed in HepG2 cells. Our data showed that cell apoptosis generally increased after norcantharidin treatment in HepG2 cells. Expression of LC3-II, an autophagosome marker, increased when cells were treated with HBSS media. It also increased cell viability. However, in the presence of 3-MA and Atg5 siRNA, autophagy was inhibited, LC3-II expression decreased and cell apoptosis increased. There was increased expression of Bax, cytochrome c, cleaved caspase-3, caspase-9 and PARP and the mitochondrial membrane potential was disrupted. Additionally, increased apoptosis was accompanied by increased reactive oxygen species (ROS) production. NCTD has anticancer activity, and Atg5 siRNA-mediated downregulation of autophagy enhanced its anticancer actions due to ROS generation and activation of the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Xuanxuan Xiong
- Department Of Gastroenterology 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Mingbo Wu
- Department Of Gastroenterology 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Haiyan Zhang
- Department Of Gastroenterology 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Jin Li
- Department of Oncological Surgery 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), The Tumor Research Institute of the Southeast University (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Bo Lu
- Department Of Gastroenterology 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Yonggao Guo
- Department Of Gastroenterology 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Tian Zhou
- Department Of Gastroenterology 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Hao Guo
- Department of Oncological Surgery 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), The Tumor Research Institute of the Southeast University (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Rui Peng
- Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiangcheng Li
- Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qingzhong Tian
- Department of Oncological Surgery 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), The Tumor Research Institute of the Southeast University (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| | - Yun Wang
- Department of Oncological Surgery 2, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xuzhou), The Tumor Research Institute of the Southeast University (Xuzhou), Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
16
|
Wan Z, Pan H, Liu S, Zhu J, Qi W, Fu K, Zhao T, Liang J. Downregulation of SNAIL sensitizes hepatocellular carcinoma cells to TRAIL-induced apoptosis by regulating the NF-κB pathway. Oncol Rep 2015; 33:1560-6. [PMID: 25607597 DOI: 10.3892/or.2015.3743] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/09/2015] [Indexed: 11/05/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer and the second most lethal cancer worldwide. Evidence has shown HCC cell resistance to TRAIL‑mediated apoptosis. In a previous study, we verified that silencing SNAIL downregulated the growth of HCC cells. In addition, the mechanism of resistance to TRAIL in HCC cells was connected with the activation of nuclear factor-κB (NF-κB). Thus, it was hypothesized that the downregultaion of SNAIL sensitizes HCC cells to TRAIL-induced apoptosis by regulating the NF-κB pathway. In the present study, the most effective lentiviral vectors carrying shRNA against SNAIL were selected and adenoviral vectors harboring TRAIL were constructed. The expression of SNAIL and TRAIL was detected by quantitative PCR and western blotting. HCC cell viability and apoptosis were assessed using an MTT assay and the Hoechst test. To determine how to sensitize HCC cells to TRAIL-induced apoptosis after silencing SNAIL, p53 was assessed by western blot analysis. We also investigated the expression of Bcl-xL, cIAP2, survivin and Raf-1 protein using western blot analysis and the apoptotic degree of HuH-7 cells was detected using the Hoechst test following the suppression of each gene, which was a possible molecular mechanism to sensitive TRAIL-induced apoptosis through the downregulation of SNAIL in HCC cells. Silencing SNAIL resulted in increased apoptosis by enhancing sensitization to TRAIL in all the HCC cells. Additionally, p53 protein was upregulated in HuH-7 cells. Expression of Bcl-xL, cIAP2, survivin and Raf-1 was downregulated following silencing of SNAIL, while down-regulation of any of the proteins contributed to SNAIL suppression enhancing HCC cell sensitivity to TRAIL‑induced apoptosis, with the exception of cIAP2. The results demonstrated that silencing SNAIL can sensitize TRAIL-induced apoptosis in HCC cells by upregulating p53 protein and by regulating related genes of the NF-κB pathway such as Bcl-xL, survivin and Raf-1.
Collapse
Affiliation(s)
- Zhaojun Wan
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Huazheng Pan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Shihai Liu
- Department of Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jingjuan Zhu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Weiwei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Kai Fu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Teng Zhao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jun Liang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
17
|
Firempong CK, Cao X, Tong S, Yu J, Xu X. Prospects for multitarget lipid-raft-coated silica beads: a remarkable online biomaterial for discovering multitarget antitumor lead compounds. RSC Adv 2015. [DOI: 10.1039/c5ra08322b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Application of lipid raft biomaterial with multiple cancer-related receptors for screening novel multitarget antitumour lead compounds.
Collapse
Affiliation(s)
- Caleb Kesse Firempong
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Xia Cao
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Shanshan Tong
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Jiangnan Yu
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| | - Ximing Xu
- Department of Pharmaceutics
- School of Pharmacy
- Centre for Nano Drug/Gene Delivery and Tissue Engineering
- Jiangsu University
- Zhenjiang
| |
Collapse
|
18
|
Fayyaz S, Yaylim I, Turan S, Kanwal S, Farooqi AA. Hepatocellular carcinoma: targeting of oncogenic signaling networks in TRAIL resistant cancer cells. Mol Biol Rep 2014; 41:6909-17. [PMID: 25037270 DOI: 10.1007/s11033-014-3577-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/01/2014] [Indexed: 01/18/2023]
Abstract
Apoptotic response in hepatocellular carcinoma (HCC) cells is impaired because of interconnectivity of proteins into complexes and signaling networks that are highly divergent in time and space. TNF-related apoptosis-inducing ligand (TRAIL) has emerged as an attractive anticancer agent reported to selectively induce apoptosis in cancer cells. Although diametrically opposed roles of TRAIL are reported both as an inducer of apoptosis and regulator of metastasis, overwhelmingly accumulating experimental evidence highlighting apoptosis inducing activity of TRAIL is directing TRAIL into clinical trials. Insights from TRAIL mediated signaling in HCC research are catalyzing new lines of study that should not only explain molecular mechanisms of disease but also highlight emerging paradigms in restoration of TRAIL mediated apoptosis in resistant cancer cells. It is becoming progressively more understandable that phytochemicals derived from edible plants have shown potential in modelling their interactions with their target proteins. Rapidly accumulating in vitro and in-vivo evidence indicates that phytonutrients have anticancer activity in rodent models of hepatocellular carcinoma. In this review we bring to limelight how phytonutrients restore apoptosis in hepatocellular carcinoma cells by rebalancing pro-apoptotic and anti-apoptotic proteins. Evidence has started to emerge, that reveals how phytonutrients target pharmacologically intractable proteins to suppress cancer. Target-based small-molecule discovery has entered into the mainstream research in the pharmaceutical industry and a better comprehension of the genetics of patients will be essential for identification of responders and non-responders.
Collapse
Affiliation(s)
- Sundas Fayyaz
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | | | | | | | | |
Collapse
|
19
|
Antognelli C, Palumbo I, Aristei C, Talesa VN. Glyoxalase I inhibition induces apoptosis in irradiated MCF-7 cells via a novel mechanism involving Hsp27, p53 and NF-κB. Br J Cancer 2014; 111:395-406. [PMID: 24918814 PMCID: PMC4102940 DOI: 10.1038/bjc.2014.280] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/24/2014] [Accepted: 04/30/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Glyoxalase I (GI) is a cellular defence enzyme involved in the detoxification of methylglyoxal (MG), a cytotoxic byproduct of glycolysis, and MG-derived advanced glycation end products (AGEs). Argpyrimidine (AP), one of the major AGEs coming from MG modifications of proteins arginines, is a pro-apoptotic agent. Radiotherapy is an important modality widely used in cancer treatment. Exposure of cells to ionising radiation (IR) results in a number of complex biological responses, including apoptosis. The present study was aimed at investigating whether, and through which mechanism, GI was involved in IR-induced apoptosis. METHODS Apoptosis, by TUNEL assay, transcript and protein levels or enzymatic activity, by RT-PCR, western blot and spectrophotometric methods, respectively, were evaluated in irradiated MCF-7 breast cancer cells, also in experiments with appropriate inhibitors or using small interfering RNA. RESULTS Ionising radiation induced a dramatic reactive oxygen species (ROS)-mediated inhibition of GI, leading to AP-modified Hsp27 protein accumulation that, in a mechanism involving p53 and NF-κB, triggered an apoptotic mitochondrial pathway. Inhibition of GI occurred at both functional and transcriptional levels, the latter occurring via ERK1/2 MAPK and ERα modulation. CONCLUSIONS Glyoxalase I is involved in the IR-induced MCF-7 cell mitochondrial apoptotic pathway via a novel mechanism involving Hsp27, p53 and NF-κB.
Collapse
Affiliation(s)
- C Antognelli
- Department of Experimental Medicine, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - I Palumbo
- Radiation Oncology Section, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - C Aristei
- Radiation Oncology Section, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| | - V N Talesa
- Department of Experimental Medicine, University of Perugia, Sant'Andrea delle Fratte, 06132 Perugia, Italy
| |
Collapse
|
20
|
Effects of quercetin on hedgehog signaling in chronic myeloid leukemia KBM7 cells. Chin J Integr Med 2014; 20:776-81. [PMID: 24928376 DOI: 10.1007/s11655-014-1817-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate the effects of quercetin on Hedgehog (Hh) signaling in chronic myeloid leukemia KBM7 cells. METHODS The KBM7 cells were treated with 50, 100 and 200 μmol/L quercetin for 48 h respectively. And then the trypan blue assay was used to examine the proliferative inhibition of quercetin. Apoptotic cells and cell cycle were measured by flow cytometry. The mRNA and protein expression were detected by quantitative real-time polymerase chain reaction (PCR) and Western blot, respectively. RESULTS Quercetin significantly inhibited KBM7 cell proliferation, induced cell apoptosis, and blocked cell cycle at G1 phase, which were in dose-dependent manners. The mRNA and protein expression of Smoothened and Glioma1 (Gli1), the members of Hh pathway decreased after treatment with quercetin. The Bcl-2 and Cyclin D1, targets of Hh signaling, also decreased after treatment with quercetin, respectively. Quercetin also could increase p53 and Caspase-3 expression. Bcr-abl mRNA copies decreased, but no changes of phosphorylated Bcr-abl and Bcr-abl proteins were observed, after treatment with quercetin. CONCLUSION Quercetin could inhibit Hh signaling and its downstream targets in the KBM7 cells. And it might be one of mechanisms of inducing apoptosis and inhibiting cell cycle by quercetin.
Collapse
|
21
|
Lu S, Gao Y, Huang X, Wang X. Cantharidin exerts anti-hepatocellular carcinoma by miR-214 modulating macrophage polarization. Int J Biol Sci 2014; 10:415-25. [PMID: 24719559 PMCID: PMC3979994 DOI: 10.7150/ijbs.8002] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/22/2014] [Indexed: 01/04/2023] Open
Abstract
Norcantharidin (NCTD), a demethylated form of cantharidin, has been used as a routine anticancer drug in China. In this study, the effect and mechanism of NCTD on anti-hepatocellular carcinoma (HCC) was examined. In vivo antitumor activity was investigated in hepatoma-bearing mice by intraperitoneal injection of different concentration of NCTD. The levels of MicroRNAs (miRNAs) and mRNA were detected by real-time PCR. The concentrations of IL-10 and IL-12 in BMDMs, Raw 264.7 cells or tumor-associated macrophages (TAMs) were measured with ELISA kit. The effects of TAMs on H22 cell survival and invasion were assayed via the CCK-8 and tumor invasion assay, respectively. Anti-miR-214 or pre-miR-214 was used to down-regulate or up-regulated miR-214 expression. The results showed that NCTD drastically impaired tumor growth in hepatoma-bearing mice, correlating with increased anti-tumor activity of TAMs. Moreover, NCTD stimulation led to an alteration of HCC microenvironment, reflected by a decrease in a shift from M2 to M1 polarization and the populations of CD4+/CD25+Foxp3 T cells. The activation of STAT3 was inhibited in TAMs from hepatoma-bearing mice injected with NCTD. Addition of NCTD to treat RAW264.7 or TAMs enhanced M1 polarization through increase of miR-214 expression. NCTD significantly inhibited β-catenin expression, which could be reversed by miR-214 inhibitor. Conditioned media from TAMs in hepatoma-bearing mice treated with NCTD or TAMs transfected with pre-miR-214 inhibited survival and invasion of H22 cells. This finding reveals a novel role for NCTD on inhibition of HCC through miR-214 modulating macrophage polarization.
Collapse
Affiliation(s)
- Sen Lu
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, Ministry of Health, Nanjing 210029, China
| | - Yun Gao
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, Ministry of Health, Nanjing 210029, China
| | - Xinli Huang
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, Ministry of Health, Nanjing 210029, China
| | - Xuehao Wang
- Center of Liver Transplantation, The First Affiliated Hospital of Nanjing Medical University, The Key Laboratory of Living Donor Liver Transplantation, Ministry of Health, Nanjing 210029, China
| |
Collapse
|
22
|
Prasad SB, Verma AK. Cantharidin-mediated ultrastructural and biochemical changes in mitochondria lead to apoptosis and necrosis in murine Dalton's lymphoma. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2013; 19:1377-1394. [PMID: 24029497 DOI: 10.1017/s143192761301324x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Cantharidin, a type of terpenoid, is the blistering agent of blister beetles frequently used in traditional medicine. The isolation and anticancer activity of cantharidin from blister beetles, Mylabris cichorii has been recently reported by us. This study deals with changes in mitochondrial structure and function and understanding their significance in the underlying mechanism(s) in cantharidin-mediated antitumor effects in Dalton's lymphoma (DL) bearing mice. Cantharidin treatment caused the appearance of abnormal mitochondrial features which included roundish mitochondria with thickened membranes, irregularity in cristae, and appearance of small and large size vacuoles in mitochondria of DL cells. Cantharidin treatment resulted in a decrease in mitochondrial reduced glutathione, succinate dehydrogenase activity, mitochondrial membrane potential, and induced apoptosis and necrosis in DL cells. The decrease/release of mitochondrial cytochrome c were also observed after cantharidin treatment. Flow cytometry-based cell cycle analysis showed a time-dependent accumulation of the sub-G0 population of DL cells, thus, confirming the involvement of apoptosis in tumor cells in cantharidin-mediated antitumor activity. These finding signify that the apoptosis induced by cantharidin in DL cells should involve mitochondrial-dependent pathways. It is suggested that these cantharidin-mediated changes in mitochondria may play a crucial role in its antitumor activity.
Collapse
Affiliation(s)
- Surya B Prasad
- Department of Zoology, Cell and Tumor Biology Laboratory, North-Eastern Hill University, Shillong-793 022, India
| | | |
Collapse
|
23
|
Chen S, Qu X, Wan P, Li QW, Wang Z, Guo F, Bai L, Hu Z, Tan W, Li J. Norcantharidin inhibits pre-replicative complexes assembly of HepG2 cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2013; 41:665-82. [PMID: 23711148 DOI: 10.1142/s0192415x13500468] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Norcantharidin (NCTD) is currently used for anticancer therapy but the exact mechanism of action remains unknown. Pre-replicative complexes (pre-RCs) are essential for cell DNA replication and highly related to malignant proliferation. Here, we examined the inhibitory effect of NCTD on pre-RC components in HepG2 cells. We showed that NCTD induced degradation of Cdc6 and Mcm2 in a dose-dependent manner. Under 100 μM NCTD concentration, about 70% of Cdc6 and 50% of Mcm2 were degraded. In addition, the nuclear translocation of Mcm6 was inhibited by NCTD. Further studies aiming at G1 synchronous cells showed that, NCTD reduced the chromatin-bound Cdc6, Mcm2 and Mcm6. Moreover, the cells were blocked from entering the S phase and accumulated at the G1 phase when released synchronously into the cell cycle. Consistently, the DNA replication was inhibited by NCTD. Finally, the combination NCTD with Cdc6 depletion lead to more severe cytotoxicity (88%) than NCTD (52%) and Cdc6 depletion (39%) alone. A synergic cytotoxicity was observed between Cdc6 depletion and NCTD. In conclusion, our results demonstrate that NCTD inhibits pre-RC assembly; subsequently blocks the G1 to S transition; and inhibits DNA replication in HepG2 cells. Pre-RCs are an intriguing target for cancer therapy, which merits further investigations for anticancer development.
Collapse
Affiliation(s)
- Sansan Chen
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Liu B, Sun WY, Zhi CY, Lu TC, Gao HM, Zhou JH, Yan WQ, Gao HC. Role of S100A3 in human colorectal cancer and the anticancer effect of cantharidinate. Exp Ther Med 2013; 6:1499-1503. [PMID: 24255681 PMCID: PMC3829748 DOI: 10.3892/etm.2013.1344] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/19/2013] [Indexed: 11/06/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality. The early diagnosis and treatment of CRC is the key to improving the survival of patients who may benefit from adjuvant chemotherapy. In the present study, the protein expression of S100A3 was observed in a cohort of 20 patients with cancer, which indicated that S100A3 activation was involved in tumorigenesis. In addition, the anticancer activity of cantharidinate was investigated using immunohistochemistry and quantitative polymerase chain reaction (qPCR) analysis. The protein expression of S100A3 was observed to increase by 2.4-fold in human CRC cells compared with the expression level in normal control cells (P<0.01). Cantharidinate inhibited the protein and gene expression of S100A3 in UCT-116 human CRC cells in vitro. These results suggested that S100A3 is important in human CRC. Cantharidinate has the potential to be considered as a novel adjuvant drug for controlling the expression of S100A3 in human CRC as it exhibits preventive effects.
Collapse
Affiliation(s)
- Bin Liu
- Department of General Surgery, Jilin University Second Hospital, Changchun, Jilin 130041, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Yang LL, Liang CY, Lu TC, Zhi CY, Liu B, Zhou JH, Liu XM, Gao HC, Huang W. Role of tissue transglutaminase and effect of cantharidinate in human colorectal cancer. Mol Med Rep 2013; 8:1812-6. [PMID: 24085483 DOI: 10.3892/mmr.2013.1706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 09/17/2013] [Indexed: 11/06/2022] Open
Abstract
The role of tissue transglutaminase (tTG) in cancer development remains an important field of study. The aim of the current study was to understand the involvement of tTG in cancer and the inhibitory effect of cantharidinate on the expression of tTG in human colorectal cancer (CRC) using immunohistochemical and PCR analysis. The results showed that the expression of tTG increased in human CRC and cantharidinate inhibited the expression of tTG. These results suggested that tTG is significant in human CRC and that tTG may be an important target for tumor chemoprevention and treatment. Cantharidinate may be considered as a novel co‑therapy for controlling tTG expression in human CRC.
Collapse
Affiliation(s)
- Lan-Lan Yang
- Department of Gastroenterology and Hepatology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Verma AK, Prasad SB. Antitumor effect of blister beetles: an ethno-medicinal practice in Karbi community and its experimental evaluation against a murine malignant tumor model. JOURNAL OF ETHNOPHARMACOLOGY 2013; 148:869-879. [PMID: 23707211 DOI: 10.1016/j.jep.2013.05.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 04/09/2013] [Accepted: 05/06/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL IMPORTANCE The blister beetles Epicauta hirticornis and Mylabris cichorii are used as a folk medicine by the Karbi tribe in Karbi Anglong district of Assam, India for the treatment of different human ailments, including cancer cases. AIM OF THE STUDY It includes field survey related to zoo-therapeutic aspects of two blister beetles in Karbi community, isolation of bio-active compound and evaluation of its antitumor potential with possible mode of action against murine Ehrlich ascites carcinoma (EAC). MATERIALS AND METHODS The main bio-active compound of blister beetles was isolated from ethyl acetate extract and the structure was confirmed as cantharidin using NMR, IR, Mass and X-ray diffractometer. The effect of cantharidin on apoptosis, necrosis, autophagy and the apoptosis related signaling pathways were determined using different bioassays, including cell cycle analysis, mitochondrial membrane potential, western blot analysis of cytochrome c, caspases 9, 3/7 assays, and lactate dehydrogenase (LDH) assay. RESULTS Cantharidin induced apoptosis, necrosis and autophagy cell death in EAC cells. The decrease in mitochondrial membrane potential was observed, which may help to release cytochrome c from mitochondria to cytosol. Cantharidin treatment caused up-regulation of caspases 9 and -3/7 and a decrease in LDH activity in EAC cells. CONCLUSION The major bioactive compound of these blister beetles is cantharidin which induces severe apoptosis in EAC cells involving mitochondrial intrinsic pathway. Cantharidin-mediated inhibition of LDH activity may lead to short supply of NAD(+) and cut off energy and anabolic supply to cancer cells.
Collapse
Affiliation(s)
- Akalesh Kumar Verma
- Cell and Tumor Biology Laboratory, Department of Zoology, School of Life Sciences, North-Eastern Hill University, Shillong 793 022, India
| | | |
Collapse
|