1
|
Guo H, Ren W, Guo M, Wu X, Guo Q. A Comprehensive Review on Ethnopharmacology, Phytochemistry of Mylabris, and Pharmacology of Cantharidin. Chem Biodivers 2025; 22:e202500266. [PMID: 40095765 DOI: 10.1002/cbdv.202500266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/19/2025]
Abstract
Mylabris, the dried body of the Chinese blister beetle, has been utilized in traditional medicine across Asia, Europe, South Africa, and North America for the treatment of tumors, carbuncles, and scrofula. Phytochemical studies revealed cantharidin and its derivatives as the main constituents. Mylabris extracts and its phytochemicals have demonstrated promising pharmacological efficacy, including antitumor, cardiovascular protective, anti-osteoporotic, antidiabetic, and antileishmanial properties. Despite its extensive history of medicinal use and promising therapeutic potential, comprehensive reviews addressing the chemical constituents and pharmacological activities of Mylabris are still limited. This review aims to provide a detailed and systematic overview of ethnopharmacology and phytochemistry of Mylabris, as well as the pharmacology of cantharidin, highlighting the potential of Mylabris as a source of novel therapeutic agents. By summarizing the research findings, this review seeks to enhance the scientific understanding of Mylabris, support its rational clinical application, and guide future research directions, ultimately contributing to the development of new and effective treatments for various diseases.
Collapse
Affiliation(s)
- Huan Guo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, P. R. China
| | - Wenshuo Ren
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, P. R. China
| | - Meizhu Guo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, P. R. China
| | - Xia Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, P. R. China
| | - Qiang Guo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, P. R. China
| |
Collapse
|
2
|
Nguyen TTK, Woo SM, Seo SU, Banstola A, Kim H, Duwa R, Vu ATT, Hong IS, Kwon TK, Yook S. Enhanced anticancer efficacy of TRAIL-conjugated and odanacatib-loaded PLGA nanoparticles in TRAIL resistant cancer. Biomaterials 2025; 312:122733. [PMID: 39106819 DOI: 10.1016/j.biomaterials.2024.122733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/27/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) demonstrates unique characteristics in anticancer therapies as it selectively induces apoptosis in cancer cells. However, most cancer cells are TRAIL-resistant. Odanacatib (ODN), a cathepsin K inhibitor, is considered a novel sensitizer for cancer treatment. Combination therapy between TRAIL and sensitizers is considered a potent platform that improves TRAIL-based anticancer therapies beyond TRAIL monotherapy. Herein, we developed ODN loaded poly(lactic-co-glycolic) nanoparticles conjugated to GST-TRAIL (TRAIL-ODN-PLGA-NPs) to target and treat TRAIL-resistant cancer. TRAIL-ODN-PLGA-NPs demonstrated a significant increase in cellular uptake via death receptors (DR5 and DR4) on surface of cancer cells. TRAIL-ODN-PLGA-NPs exposure destroyed more TRAIL-resistant cells compared to a single treatment with free drugs. The released ODN decreased the Raptor protein, thereby increasing damage to mitochondria by elevating reactive oxygen species (ROS) generation. Additionally, Bim protein stabilization improved TRAIL-resistant cell sensitization to TRAIL-induced apoptosis. The in vivo biodistribution study revealed that TRAIL-ODN-PLGA-NPs demonstrated high location and retention in tumor sites via the intravenous route. Furthermore, TRAIL-ODN-PLGA-NPs significantly inhibited xenograft tumor models of TRAIL-resistant Caki-1 and TRAIL-sensitive MDA-MB-231 cells.The inhibition was associated with apoptosis activation, Raptor protein stabilizing Bim protein downregulation, Bax accumulation, and mitochondrial ROS generation elevation. Additionally, TRAIL-ODN-PLGA-NPs affected the tumor microenvironment by increasing tumor necrosis factor-α and reducing interleukin-6. In conclusion, we evealed that our formulation demonstrated synergistic effects against TRAIL compared with the combination of free drug in vitro and in vivo models. Therefore, TRAIL-ODN-PLGA-NPs may be a novel candidate for TRAIL-induced apoptosis in cancer treatment.
Collapse
Affiliation(s)
- Thoa Thi Kim Nguyen
- College of Pharmacy, Keimyung University, Daegu, 42602, Republic of Korea; Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Seon Min Woo
- Department of Immunology, School of medicine, Keimyung University, Daegu, Republic of Korea
| | - Seung Un Seo
- Department of Immunology, School of medicine, Keimyung University, Daegu, Republic of Korea
| | - Asmita Banstola
- Department of Dermatology, Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
| | - Haesoo Kim
- College of Pharmacy, Keimyung University, Daegu, 42602, Republic of Korea
| | - Ramesh Duwa
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Radiology, Molecular Imaging Program at Stanford (MIPS), School of medicine, Stanford University, Stanford, CA, 94305, USA
| | - An Thi Thanh Vu
- College of Pharmacy, Keimyung University, Daegu, 42602, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 406-840, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of medicine, Keimyung University, Daegu, Republic of Korea; Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
3
|
Xu WL, Tang WJ, Yang WY, Sun LC, Zhang ZQ, Li W, Zang XX. Multiorgan dysfunction syndrome due to high-dose cantharidin poisoning: A case report. World J Clin Cases 2024; 12:2074-2078. [PMID: 38680272 PMCID: PMC11045515 DOI: 10.12998/wjcc.v12.i12.2074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/07/2024] [Accepted: 03/28/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND This report delves into the diagnostic and therapeutic journey undertaken by a patient with high-dose cantharidin poisoning and multiorgan dysfunction syndrome (MODS). Particular emphasis is placed on the comprehensive elucidation of the clinical manifestations of high-dose cantharidin poisoning, the intricate path to diagnosis, and the exploration of potential underlying mechanisms. CASE SUMMARY A patient taking 10 g of cantharidin powder orally subsequently developed MODS. The patient was treated with supportive care, fluid hydration and antibiotics, and hemoperfusion and hemofiltration therapy for 24 h and successfully recovered 8 d after hospital admission. Cantharidin poisoning can cause life-threatening MODS and is rare clinically. This case underscores the challenge in diagnosis and highlights the need for early clinical differentiation to facilitate accurate assessment and prompt intervention. CONCLUSION This article has reported and analyzed the clinical data, diagnosis, treatment, and prognosis of a case of high-dose cantharidin poisoning resulting in MODS and reviewed the relevant literature to improve the clinical understanding of this rare condition.
Collapse
Affiliation(s)
- Wan-Ling Xu
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Wen-Jing Tang
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Wei-Ying Yang
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Li-Chao Sun
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Ze-Qun Zhang
- Department of Chinese Traditional Medicine, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Wei Li
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Xiu-Xian Zang
- Department of Emergency Medicine, First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
4
|
Li S, Duan X, Zhang Y, Zhao C, Yu M, Li X, Li X, Zhang J. Lipidomics reveals serum lipid metabolism disorders in CTD-induced liver injury. BMC Pharmacol Toxicol 2024; 25:10. [PMID: 38225635 PMCID: PMC10790540 DOI: 10.1186/s40360-024-00732-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/03/2024] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND Cantharidin (CTD), the main toxic component of Mylabris, has been extensively used for tumor treatment in recent years. CTD-induced liver toxicity has attracted significant interest in clinic. METHODS In this study, biochemical parameters and liver pathological changes were analyzed after CTD was administered to mice by gavage. Subsequently, a lipidomic approach was used to investigate serum lipid metabolism disorders, and the mechanism underlying CTD-induced liver injury in mice was explored. RESULTS The results showed that the levels of TC and LDL-C were significantly increased after CTD intervention. Besides, pathological results showed inflammatory cell infiltration and hepatocyte necrosis in the liver. Furthermore, lipidomics found that a total of 18 lipid metabolites were increased and 40 were decreased, including LPC(20:4), LPC(20:3), PC(22:6e/2:0), PE(14:0e/21:2), PC(18:2e/22:6), glycerophospholipids, CE(16:0), CE(18:0) Cholesterol esters and TAG(12:0/12:0/22:3), TAG(16:1/16:2/20:4), TAG(18:1/18:1/20:0), TAG(16:2/18:2/18:2), TAG(18:0/18:0/20:0), TAG(13:1/19:0/19:0) glycerolipids. Metabolic pathway analysis found that glycerophospholipid, glycerol ester and glycosylphosphatidylinositol (GPI)-anchored biosynthetic metabolic pathways were dysregulated and the increase in PE caused by glycophoric metabololism and GPI may be the source of lipid metabolism disorders caused by CTD. Overall, the present study provided new insights into the mechanism of CTD-induced liver injury and increased drug safety during clinical application.
Collapse
Affiliation(s)
- Shan Li
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xiaotong Duan
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Yixin Zhang
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Cancan Zhao
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Ming Yu
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xiaofei Li
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| | - Xiaomei Li
- Cancer Research Laboratory, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China.
| | - Jianyong Zhang
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
5
|
Yan J, Gao YM, Deng XL, Wang HS, Shi GT. Integrative analysis of the molecular signature of target genes involved in the antitumor effects of cantharidin on hepatocellular carcinoma. BMC Cancer 2023; 23:1161. [PMID: 38017425 PMCID: PMC10685469 DOI: 10.1186/s12885-023-11594-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Cantharidin (CTD) is the active ingredient of Chinese medicine, which has been traditionally used in multiple cancers treatment, especially in hepatocellular carcinoma (HCC). However, a comprehensive analysis of the CTD-related molecular mechanism is still necessary to understand its functions in HCC treatment. This study aimed to reveal the novel molecular targets and regulatory networks of CTD in HCC. METHODS A model of H22 tumour-bearing mice was constructed, and the function of CTD in tumour growth was evaluated. An integrated approach of CTD associated transcriptional profiling and biological systems analysis was used to identify key regulators involved in antitumour pathways. The identified differential expression patterns were supported by the results of Gene Ontology (GO) term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyse, and by protein-protein interaction (PPI) network construction. The relationships between gene expression and tumour immunity were evaluated using Tumour Immune Estimation Resource (TIMER). Prognostic value was analyzed with Kaplan-Meier plotter. RESULTS In the present study, the therapeutic effect of CTD on HCC was evaluated in vivo. We obtained the CTD-related transcriptional profiles, systematically and intuitively illustrated its possible pharmacological mechanisms in HCC through multiple targets and signalling pathways. These results revealed that the CTD-related differentially expressed genes were involved in autophagy, transcription factors (TFs) related transcriptional regulation, fatty acid metabolism and immune response in HCC. We found that MAPT, TOP2A, CENPF and MEFV were hub genes of CTD targets involved in autophagy regulation. Totally, 14 TFs have been confirmed to be critical for transcriptional regulation, and 33 TF targets were identified as the hub genes in transcriptional mis-regulation pathway in cancer. These TFs were associated with the immune response and immune cell infiltration. In addition, the downregulated genes were significantly enriched in metabolic regulation pathways, especially fatty acid metabolism after CTD treatment. Furthermore, the network of CTD associated miRNAs with these fatty acid metabolism-related targets was constructed in HCC. CONCLUSIONS Taken together, our results comprehensively elucidated that CTD could act on multiple targets in HCC therapy, affecting autophagy, transcriptional regulation, the immune response and fatty acid metabolism. Our results provide a foundation for the study of the molecular mechanistic of CTD and its clinical application in the treatment of HCC.
Collapse
Affiliation(s)
- Jia Yan
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yu Min Gao
- School of Public health, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiu Ling Deng
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| | - Hai Sheng Wang
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| | - Gui Tao Shi
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
6
|
Chen J, Lin Y, Zheng S, Chen Q, Tang S, Zhong X. CBX3 promotes clear cell renal carcinoma through PI3K/AKT activation and aberrant immunity. J Transl Med 2023; 21:600. [PMID: 37674204 PMCID: PMC10483741 DOI: 10.1186/s12967-023-04478-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND A chromobox homologue 3 (CBX3) is elevated in various cancers and significantly contributes to the promotion of malignant behavior; despite this, its exact involvement in clear cell renal cell carcinoma (ccRCC) is yet unknown. METHODS The Cancer Genome Atlas database served to evaluate CBX3 production and its connection to survival in patients with ccRCC. Our team evaluated the effects of knockdown of CBX3 levels in ccRCC cell populations using in vitro together with in vivo models. CBX3, proteins related to death, and epithelial-to-mesenchymal transition (EMT)-related proteins were measured in ccRCC cells using western blotting and immunohistochemical assays. Through the analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) and GeneOntology (GO) and Gene Set Enrichment Analysis (GSEA), the biological processes and signal pathways related to CBX3 expression were identified. Immune-related activity reduced by CBX3 was assessed using various online tools. RESULTS Both genomic and protein expression showed that CBX3 was upregulated in ccRCC. Further functional analyses revealed that CBX3 played a crucial role in enhancing cell growth, migration, and EMT in vitro along with in vivo. Moreover, the study results provided distinct mechanistic evidence that CBX3 exerts its pathological functions in ccRCC by activating the PI3K/AKT pathway. Finally, immunoassays revealed that CBX3, a possible biomarker of ccRCC, was significantly associated with immunity. CONCLUSIONS Our results suggest that the overexpression of CBX3 promotes ccRCC advancement through PI3K/AKT activation and even immunological dysregulation, making it a potentially viable and beneficial therapeutic target.
Collapse
Affiliation(s)
- Jiasheng Chen
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yuxin Lin
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Shukai Zheng
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Qingshan Chen
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Xiaoping Zhong
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China.
| |
Collapse
|
7
|
Jin D, Huang NN, Wei JX. Hepatotoxic mechanism of cantharidin: insights and strategies for therapeutic intervention. Front Pharmacol 2023; 14:1201404. [PMID: 37383714 PMCID: PMC10293652 DOI: 10.3389/fphar.2023.1201404] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/01/2023] [Indexed: 06/30/2023] Open
Abstract
Cantharidin (CTD), a natural compound derived from Mylabris, is widely used in traditional Oriental medicine for its potent anticancer properties. However, its clinical application is restricted due to its high toxicity, particularly towards the liver. This review provides a concise understanding of the hepatotoxic mechanisms of CTD and highlights novel therapeutic strategies to mitigate its toxicity while enhancing its anticancer efficacy. We systematically explore the molecular mechanisms underlying CTD-induced hepatotoxicity, focusing on the involvement of apoptotic and autophagic processes in hepatocyte injury. We further discuss the endogenous and exogenous pathways implicated in CTD-induced liver damage and potential therapeutic targets. This review also summarizes the structural modifications of CTD derivatives and their impact on anticancer activity. Additionally, we delve into the advancements in nanoparticle-based drug delivery systems that hold promise in overcoming the limitations of CTD derivatives. By offering valuable insights into the hepatotoxic mechanisms of CTD and outlining potential avenues for future research, this review contributes to the ongoing efforts to develop safer and more effective CTD-based therapies.
Collapse
Affiliation(s)
- Dian Jin
- Department of Pharmacy, Sixth People’s Hospital of Chengdu, Chengdu, China
| | - Na-Na Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing-Xia Wei
- Department of Pharmacy, Sixth People’s Hospital of Chengdu, Chengdu, China
| |
Collapse
|
8
|
Norcantharidin induces G2/M arrest and apoptosis via activation of ERK and JNK, but not p38 signaling in human renal cell carcinoma ACHN cells. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2021; 71:267-278. [PMID: 33151174 DOI: 10.2478/acph-2021-0012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/15/2020] [Indexed: 01/19/2023]
Abstract
Renal cell carcinoma (RCC) is generally acknowledged as the most resistant primary malignancy unresponsive to conventional radiotherapy and chemotherapy treatments. Norcantharidin (NCTD), a therapeutic compound derived from medicinal plants, has been shown to trigger apoptosis, as well as antimetastatic and antioxidant activities in several tumor cells. However, NCTD's mechanism of antitumor activity in the RCC cell line remains unclear. In this study, we report that NCTD led to a time- and dose-dependent inhibition of cell proliferation. It had also markedly induced apoptosis and G2/M phase cell cycle arrest in a dose-dependent manner by decreasing the expressions of pro-caspase-3, pro-caspase-9, cyclin B1, and pCDC25C while increasing active caspase-3, cleaved-PARP, P21, and pCDC2 levels. Interestingly, NCTD treatment provoked the phosphorylation of extracellular-regulated protein kinase (ERK) and c-Jun-N-terminal kinase (JNK), but not of p38 MAPK. Moreover, SCH772984 and SP600125, ERK and JNK inhibitors, respectively, could partially abolish NCTD-induced apoptosis and G2/M phase cell cycle arrest. Collectively, these findings suggest that NCTD might activate JNK and ERK signaling pathways, consequently inducing apoptosis and G2/M arrest through the modulation of related proteins. This study provided evidence that NCTD is a promising therapeutic drug for the treatment of RCC.
Collapse
|
9
|
Naz F, Wu Y, Zhang N, Yang Z, Yu C. Anticancer Attributes of Cantharidin: Involved Molecular Mechanisms and Pathways. Molecules 2020; 25:E3279. [PMID: 32707651 PMCID: PMC7397086 DOI: 10.3390/molecules25143279] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is a preeminent threat to the human race, causing millions of deaths each year on the Earth. Traditionally, natural compounds are deemed promising agents for cancer treatment. Cantharidin (CTD)-a terpenoid isolated from blister beetles-has been used extensively in traditional Chinese medicines for healing various maladies and cancer. CTD has been proven to be protein phosphatase 2A (PP2A) and heat shock transcription factor 1 (HSF-1) inhibitor, which can be potential targets for its anticancer activity. Albeit, it harbors some toxicities, its immense anticancer potential cannot be overlooked, as the cancer-specific delivery of CTD could help to rescue its lethal effects. Furthermore, several derivatives have been designed to weaken its toxicity. In light of extensive research, the antitumor activity of CTD is evident in both in vitro as well as in vivo cancer models. CTD has also proven efficacious in combination with chemotherapy and radiotherapy and it can also target some drug-resistant cancer cells. This mini-review endeavors to interpret and summarize recent information about CTD anticancer potential and underlying molecular mechanisms. The pertinent anticancer strength of CTD could be employed to develop an effective anticarcinogenic drug.
Collapse
Affiliation(s)
| | | | | | - Zhao Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (F.N.); (Y.W.); (N.Z.)
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (F.N.); (Y.W.); (N.Z.)
| |
Collapse
|
10
|
Liu F, Duan C, Zhang J, Li X. Cantharidin‐induced LO2 cell autophagy and apoptosis via endoplasmic reticulum stress pathway in vitro. J Appl Toxicol 2020; 40:1622-1635. [DOI: 10.1002/jat.4022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/06/2020] [Accepted: 05/19/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Fang Liu
- Basic Medical School Zunyi Medical University Zunyi China
| | - Cancan Duan
- Key Lab Basic Pharmacology of Ministry of Education and Joint International Research laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi China
| | - Jianyong Zhang
- Key Lab Basic Pharmacology of Ministry of Education and Joint International Research laboratory of Ethnomedicine of Ministry of Education Zunyi Medical University Zunyi China
- School of pharmacy Zunyi Medical University Zunyi China
| | - Xiaofei Li
- Basic Medical School Zunyi Medical University Zunyi China
| |
Collapse
|
11
|
Li YD, Mao Y, Dong XD, Lei ZN, Yang Y, Lin L, Ashby CR, Yang DH, Fan YF, Chen ZS. Methyl-Cantharidimide (MCA) Has Anticancer Efficacy in ABCB1- and ABCG2-Overexpressing and Cisplatin Resistant Cancer Cells. Front Oncol 2020; 10:932. [PMID: 32676451 PMCID: PMC7333678 DOI: 10.3389/fonc.2020.00932] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/12/2020] [Indexed: 01/16/2023] Open
Abstract
In this study, we investigated the efficacy of methyl-cantharidimide (MCA), a cantharidin (CTD) analog, as an anticancer drug, in cancer cells overexpressing either ABCB1 or ABCG2 transporters and in cisplatin-resistant cancer cells. The results indicated that: (i) MCA was efficacious in the ABCB1-overexpressing cell line, KB-C2, and the ABCB1-gene-transfected cell line, HEK293/ABCB1 (IC50 from 6.37 to 8.44 mM); (ii) MCA was also efficacious in the ABCG2-overexpressing cell line, NCI-H460/MX20, and the ABCG2-gene-transfected cell lines, HEK293/ABCG2-482-R2, HEK293/ABCG2-482-G2, and the HEK293/ABCG2-482-T7 cell lines (IC50 from 6.37 to 9.70 mM); (iii) MCA was efficacious in the cisplatin resistant cancer cell lines, KCP-4 and BEL-7404/CP20 (IC50 values from 7.05 to 8.16 mM); (iv) MCA (up to 16 mM) induced apoptosis in both BEL-7404 and BEL-7404/CP20 cancer cells; (v) MCA arrested both BEL-7404 and BEL-7404/CP20 cancer cells in the G0/G1 phase of the cell cycle; (vi) MCA (8 mM) upregulated the expression level of the protein, unc-5 netrin receptor B (UNC5B) in HepG2 and BEL-7404 cancer cells. Overall, our results indicated that MCA's efficacy in ABCB1- and ABCG2-overexpressing and cisplatin resistant cancer cells is due to the induction of apoptosis and cell cycle arrest in the G0/G1 phase.
Collapse
Affiliation(s)
- Yi-Dong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xing-Duo Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Lizhu Lin
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Dong-Hua Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Ying-Fang Fan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States.,Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| |
Collapse
|
12
|
Nazim UM, Yin H, Park SY. Downregulation of c‑FLIP and upregulation of DR‑5 by cantharidin sensitizes TRAIL‑mediated apoptosis in prostate cancer cells via autophagy flux. Int J Mol Med 2020; 46:280-288. [PMID: 32319535 PMCID: PMC7255450 DOI: 10.3892/ijmm.2020.4566] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 03/06/2020] [Indexed: 11/17/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apop-tosis-inducing ligand (TRAIL), a type II transmembrane protein, is a part of the TNF superfamily of cytokines. Cantharidin, a type of terpenoid, is extracted from the blister beetles (Mylabris genus) used in Traditional Chinese Medicine. Cantharidin elicits antibiotic, antiviral and antitumor effects, and can affect the immune response. The present study demonstrated that a cantharidin and TRAIL combination treatment regimen elicited a synergistic outcome in TRAIL-resistant DU145 cells. Notably, it was also identified that cantharidin treatment initiated the downregulation of cellular FLICE-like inhibitory protein (c-FLIP) and upregulation of death receptor 5 (DR-5), and sensitized cells to TRAIL-mediated apoptosis by initiating autophagy flux. In addition, cantharidin treatment increased lipid-modified microtubule-associated proteins 1A/1B light chain 3B expression and significantly attenuated sequestosome 1 expression. Attenuation of autophagy flux by a specific inhibitor such as chloroquine and genetic modification using ATG5 small interfering RNA abrogated the cantharidin-mediated TRAIL-induced apoptosis. Overall, the results of the present study revealed that cantharidin effectively sensitized cells to TRAIL-mediated apoptosis and its effects are likely to be mediated by autophagy, the downregulation of c-FLIP and the upregulation of DR-5. They also suggested that the combination of cantharidin and TRAIL may be a successful therapeutic strategy for TRAIL-resistant prostate cancer.
Collapse
Affiliation(s)
- Uddin Md Nazim
- Department of Veterinary Medicine, Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| | - Honghua Yin
- Department of Veterinary Medicine, Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| | - Sang-Youel Park
- Department of Veterinary Medicine, Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| |
Collapse
|
13
|
Song M, Wang X, Luo Y, Liu Z, Tan W, Ye P, Fu Z, Lu F, Xiang W, Tang L, Yao L, Nie Y, Xiao J. Cantharidin suppresses gastric cancer cell migration/invasion by inhibiting the PI3K/Akt signaling pathway via CCAT1. Chem Biol Interact 2020; 317:108939. [PMID: 31945315 DOI: 10.1016/j.cbi.2020.108939] [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: 10/12/2019] [Revised: 12/17/2019] [Accepted: 01/08/2020] [Indexed: 12/28/2022]
Abstract
Cantharidin (CTD) is a traditional Chinese medicine that shows an anticancer effects in multiple types of cancer cells. However, the mechanism of CTD anti-cancer function in gastric cancer (GC) is still unclear. The aim of the present study was to investigate the underlying mechanism that CTD inhibits proliferation and migration through suppression of the PI3K/Akt signaling. CTD induced GC cell apoptosis and inhibited metastasis measured by CCK8 assays as well as wound healing assays and transwell assays. Mechanistic investigations suggested that CTD modulated the PI3K/Akt signaling via western-blot and quantitative q-PCR. In addition, we identified and confirmed CCAT1 as a novel direct target of CTD inhibited PI3K/AKt signaling expression. In conclusion, our results provide new point into the critical role of CTD in suppressing PI3K/Akt signaling via down-regulation of CCAT1, resulting in suppression GC cell growth and migration/invasion.
Collapse
Affiliation(s)
- Mengyun Song
- The Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China; Nankai University, School of Medicine, Tianjin, People's Republic of China
| | - Xianfei Wang
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Yajun Luo
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Zilin Liu
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Wang Tan
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Pengcheng Ye
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Zhiming Fu
- The Department of Gastrointestinal Surgery 1, Hainan general hospital, Haikou, Hainan, People's Republic of China
| | - Fei Lu
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Wanping Xiang
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Linghan Tang
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Lin Yao
- The Department of Gastroenterology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Yuqiang Nie
- The Department of Gastroenterology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, People's Republic of China
| | - Jiangwei Xiao
- The Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
14
|
Huang X, Xie W, Yu X, Fan C, Wang J, Cao Y, Li J. Methyl-Cantharidimide Inhibits Growth of Human Hepatocellular Carcinoma Cells by Inducing Cell Cycle Arrest and Promoting Apoptosis. Front Oncol 2019; 9:1234. [PMID: 31803617 PMCID: PMC6873211 DOI: 10.3389/fonc.2019.01234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022] Open
Abstract
Methyl-Cantharidimide (MCA) is a derivative of cantharidin which has potential anticancer activity. This study investigates the effect of MCA on the growth and metastasis of human hepatocellular carcinoma (HCC) cells. Human HCC HepG2 and Hep3B2.1-7 cells, and normal hepatocytes (L02) were treated with a series of concentrations of MCA. The inhibition ability of these cells was examined by CCK-8 assay. Cell cycle and cell apoptosis were determined using Flow Cytometry. The effect of MCA on cell migration and invasion was evaluated through scratch wound healing and transwell migration assays. Furthermore, Western blot was used to evaluate biomarkers associated with cell cycle and apoptosis. It was found that: (i) MCA inhibited cell proliferation in HCC cells in a dose- and time-dependent manner, especially in HepG2 cells; (ii) MCA arrested HCC cells in G-1 phase cell cycle; (iii) MCA induced HCC cells apoptosis; (iv) MCA inhibited the migration ability of HCC cells; and (v) MCA treatment significantly increased cleaved-caspase3 and decreased NF-κB protein in HCC cells. These results suggest that MCA has cytotoxic effect on HCC cells by inducing cell cycle arrest and promoting apoptosis. MCA could be developed as an previous anticancer drug for the treatment of human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiangzhong Huang
- Department of Interventional Therapy, Affiliated Jiangyin Hospital, Medical College of Southeast University, Jiangyin, China
| | - Wen Xie
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xiaofan Yu
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Caiyun Fan
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jin Wang
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yi Cao
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jianxiang Li
- School of Public Health, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Feng IC, Hsieh MJ, Chen PN, Hsieh YH, Ho HY, Yang SF, Yeh CB. Cantharidic acid induces apoptosis through the p38 MAPK signaling pathway in human hepatocellular carcinoma. ENVIRONMENTAL TOXICOLOGY 2018; 33:261-268. [PMID: 29159945 DOI: 10.1002/tox.22513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/03/2017] [Accepted: 11/07/2017] [Indexed: 06/07/2023]
Abstract
Cantharidin analogs exhibit anticancer activities, including apoptosis. However, the molecular mechanisms underlying the effects of cantharidic acid (CA), a cantharidin analog, on apoptosis in hepatocellular carcinoma (HCC) cells are unclear. Thus, in this study, we evaluated the anticancer activities of CA by investigating its ability to trigger apoptosis in SK-Hep-1 cells. Our data demonstrated that CA effectively inhibited the proliferation of SK-Hep-1 cells in a dose-dependent manner. Furthermore, CA effectively triggered cell cycle arrest and induced apoptosis, as determined by flow cytometric analysis. Western blotting revealed that CA significantly activated proapoptotic signaling including caspase-3, -8, and -9 in SK-Hep-1 cells. Moreover, treatment of SK-Hep-1 cells with CA induced the activation of ERK, p38, and c-Jun N-terminal kinase. Moreover, the inhibition of p38 by specific inhibitors abolished CA-induced cell apoptosis. In conclusion, our results indicated that CA induces apoptosis in SK-Hep-1 cells through a p38-mediated apoptotic pathway and could be a new HCC therapeutic agent.
Collapse
Affiliation(s)
- I-Che Feng
- Division of Gastroenterology and Hepatology, Chi Mei Medical Center, Yongkang District, Tainan, Taiwan
- Department of Internal Medicine, Chi Mei Medical Center, Yongkang District, Tainan, Taiwan
| | - Ming-Ju Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsin-Yu Ho
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chao-Bin Yeh
- Department of Emergency Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
16
|
Liu YP, Li L, Xu L, Dai EN, Chen WD. Cantharidin suppresses cell growth and migration, and activates autophagy in human non-small cell lung cancer cells. Oncol Lett 2018; 15:6527-6532. [PMID: 29731854 DOI: 10.3892/ol.2018.8141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022] Open
Abstract
Cantharidin (CTD), a component of Mylabris (blister beetle), is a traditional Chinese medicine that exerts an anticancer effect in multiple types of cancer cells. The aim of the present study was to investigate whether CTD exhibited anti-metastatic and inhibitory cell proliferation effects against human non-small cell lung cancer (NSCLC) A549 cells, and the possible underlying mechanism by which this occurs. The results of the present study demonstrated that CTD arrested proliferation, suppressed invasion and migration and induced apoptosis in A549 cells in vitro. Alterations of apoptosis-associated protein levels, including B-cell lymphoma-2 (Bcl-2), Bcl-associated X (Bax) and active caspase-3, were detected. Furthermore, the present study demonstrated that CTD activated autophagy through downregulation of p62 expression and upregulation of microtubule-associated proteins 1A/1B light chain 3B and Beclin-1 expression. Additionally, western blot analysis identified that CTD inhibited the phosphatidylinositol 3-kinase (PI3K)/RAC serine/threonine protein kinase (Akt)/mechanistic target of rapamycin (mTOR) signaling pathway in NSCLC, demonstrating that the levels of phosphorylated (p-)Akt, p-mTOR, phosphorylated ribosomal p70S6 protein kinase (p-p70-S6K) and cyclin D1 were significantly decreased following treatment with CTD. In conclusion, the results of the present study indicated that CTD impeded cell growth and migration by inhibiting PI3K/Akt/mTOR signaling in NSCLC, and promoted autophagy and apoptosis. CTD exhibited anticancer activity against NSCLC in vitro, revealing it as a potential candidate for the treatment of NSCLC.
Collapse
Affiliation(s)
- Yan-Peng Liu
- Department of Internal Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Ling Li
- Department of Surgery, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| | - Liang Xu
- Department of Surgery, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| | - E-Nuo Dai
- Department of Surgery, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| | - Wei-Da Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| |
Collapse
|