1
|
Li C, Ma Z, Wei X, Wang Y, Wu J, Li X, Sun X, Ding Z, Yang C, Zou Y. Bufalin Ameliorates Myocardial Ischemia/Reperfusion Injury by Suppressing Macrophage Pyroptosis via P62 Pathway. J Cardiovasc Transl Res 2025; 18:221-236. [PMID: 39733202 PMCID: PMC12043737 DOI: 10.1007/s12265-024-10577-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/29/2024] [Indexed: 12/30/2024]
Abstract
Bufalin, which is isolated from toad venom, exerts positive effects on hearts under pathological circumstance. We aimed to investigate the effects and mechanisms of bufalin on myocardial I/R injury. In vivo, bufalin ameliorated myocardial I/R injury, which characteristics with better ejection function, decreased infarct size and less apoptosis. The levels of pyroptotic proteins were increased in I/R-treated macrophages and inflammatory cytokines expressed more in I/R-induced mouse, which could be attenuated by bufalin. Bufalin also reduced H/R-treated macrophage pyroptosis in vitro. Autophagic flux blockage and ROS accumulation were reduced by bufalin in impaired macrophages. Overexpression of p62 abrogated the anti-proptosis and anti-oxidative effects of bufalin. The levels of apoptosis related proteins were changed and TUNEL-positive ratio was raised in cardiomyocytes that received conditioned medium treatment with H/R-treated macrophages, while bufalin pretreatment could reduce apoptosis. These findings indicate that bufalin may attenuate myocardial I/R injury by suppressing macrophage pyroptosis via P62 pathway.
Collapse
Affiliation(s)
- Chang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhen Ma
- Institutes of Biomedical Sciences, Fudan University, 131 Dong'an Road, Shanghai, 200032, China
| | - Xiang Wei
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai, 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xuan Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiaolei Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhiwen Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, 131 Dong'an Road, Shanghai, 200032, China.
| | - Cheng Yang
- Department of Cardiac Surgery, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, 131 Dong'an Road, Shanghai, 200032, China.
- State Key Laboratory of Genetic Engineering, Fudan University, 138 Yixueyuan Road, Shanghai, 200438, China.
| |
Collapse
|
2
|
Zhang Y, Pan R, Li K, Cheang LH, Zhao J, Zhong Z, Li S, Wang J, Zhang X, Cheng Y, Zheng X, He R, Wang H. HSPD1 Supports Osteosarcoma Progression through Stabilizing ATP5A1 and thus Activation of AKT/mTOR Signaling. Int J Biol Sci 2024; 20:5162-5190. [PMID: 39430254 PMCID: PMC11489178 DOI: 10.7150/ijbs.100015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Malignant transformation is concomitant with excessive activation of stress response pathways. Heat shock proteins (HSPs) are stress-inducible proteins that play a role in folding and processing proteins, contributing to the non-oncogene addiction of stressed tumor cells. However, the detailed role of the HSP family in osteosarcoma has not been investigated. Bulk and single-cell transcriptomic data from the GEO and TARGET databases were used to identify HSPs associated with prognosis in osteosarcoma patients. The expression level of HSPD1 was markedly increased in osteosarcoma, correlating with a negative prognosis. Through in vitro and in vivo experiments, we systematically identified HSPD1 as an important contributor to the regulation of proliferation, metastasis, and apoptosis in osteosarcoma by promoting the epithelial-mesenchymal transition (EMT) and activating AKT/mTOR signaling. Subsequently, ATP5A1 was determined as a potential target of HSPD1 using immunoprecipitation followed by mass spectrometry. Mechanistically, HSPD1 may interact with ATP5A1 to reduce the K48-linked ubiquitination and degradation of ATP5A1, which ultimately activates the AKT/mTOR pathway to ensure osteosarcoma progression and EMT process. These findings expand the potential mechanisms by which HSPD1 exerts biological effects and provide strong evidence for its inclusion as a potential therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Ruilin Pan
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Kun Li
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of the Chinese Ministry of Education, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Lek Hang Cheang
- Department of Orthopedic Surgery, Centro Hospitalar Conde de Sao Januario, Macau, China
| | - Jing Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, China
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, China
| | - Shaoping Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, China
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, Guangzhou, China
- Department of Orthopedics, NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaofang Zhang
- Department of Pharmacy, the First Affiliated Hospital, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, Guangzhou, China
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Yanmei Cheng
- Department of Cardiothoracic Surgery ICU, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Rongrong He
- State Key Laboratory of Bioactive Molecules and Drug Ability Assessment, Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of the Chinese Ministry of Education, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Chemistry and the Potential Antiviral, Anticancer, and Anti-Inflammatory Activities of Cardiotonic Steroids Derived from Toads. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196586. [PMID: 36235123 PMCID: PMC9571018 DOI: 10.3390/molecules27196586] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Cardiotonic steroids (CTS) were first documented by ancient Egyptians more than 3000 years ago. Cardiotonic steroids are a group of steroid hormones that circulate in the blood of amphibians and toads and can also be extracted from natural products such as plants, herbs, and marines. It is well known that cardiotonic steroids reveal effects against congestive heart failure and atrial fibrillation; therefore, the term "cardiotonic" has been coined. Cardiotonic steroids are divided into two distinct groups: cardenolides (plant-derived) and bufadienolides (mainly of animal origin). Cardenolides have an unsaturated five-membered lactone ring attached to the steroid nucleus at position 17; bufadienolides have a doubly unsaturated six-membered lactone ring. Cancer is a leading cause of mortality in humans all over the world. In 2040, the global cancer load is expected to be 28.4 million cases, which would be a 47% increase from 2020. Moreover, viruses and inflammations also have a very nebative impact on human health and lead to mortality. In the current review, we focus on the chemistry, antiviral and anti-cancer activities of cardiotonic steroids from the naturally derived (toads) venom to combat these chronic devastating health problems. The databases of different research engines (Google Scholar, PubMed, Science Direct, and Sci-Finder) were screened using different combinations of the following terms: “cardiotonic steroids”, “anti-inflammatory”, “antiviral”, “anticancer”, “toad venom”, “bufadienolides”, and “poison chemical composition”. Various cardiotonic steroids were isolated from diverse toad species and exhibited superior anti-inflammatory, anticancer, and antiviral activities in in vivo and in vitro models such as marinobufagenin, gammabufotalin, resibufogenin, and bufalin. These steroids are especially difficult to identify. However, several compounds and their bioactivities were identified by using different molecular and biotechnological techniques. Biotechnology is a new tool to fully or partially generate upscaled quantities of natural products, which are otherwise only available at trace amounts in organisms.
Collapse
|
4
|
Soumoy L, Ghanem GE, Saussez S, Journe F. Bufalin for an innovative therapeutic approach against cancer. Pharmacol Res 2022; 184:106442. [PMID: 36096424 DOI: 10.1016/j.phrs.2022.106442] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022]
Abstract
Bufalin is an endogenous cardiotonic steroid, first discovered in toad venom but also found in the plasma of healthy humans, with anti-tumour activities in different cancer types. The current review is focused on its mechanisms of action and highlights its very large spectrum of effects both in vitro and in vivo. All leads to the conclusion that bufalin mediates its effects by affecting all the hallmarks of cancer and seems restricted to cancer cells avoiding side effects. Bufalin decreases cancer cell proliferation by acting on the cell cycle and inducing different mechanisms of cell death including apoptosis, necroptosis, autophagy and senescence. Bufalin also moderates metastasis formation by blocking migration and invasion as well as angiogenesis and by inducing a phenotype switch towards differentiation and decreasing cancer cell stemness. Regarding its various mechanisms of action in cancer cells, bufalin blocks overactivated signalling pathways and modifies cell metabolism. Moreover, bufalin gained lately a huge interest in the field of drug resistance by both reversing various drug resistance mechanisms and affecting the immune microenvironment. Together, these data support bufalin as a quite promising new anti-cancer drug candidate.
Collapse
Affiliation(s)
- Laura Soumoy
- Laboratory of Human Anatomy & Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000 Mons, Belgium.
| | - Ghanem E Ghanem
- Laboratory of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Sven Saussez
- Laboratory of Human Anatomy & Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000 Mons, Belgium
| | - Fabrice Journe
- Laboratory of Human Anatomy & Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000 Mons, Belgium; Laboratory of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| |
Collapse
|
5
|
Tu D, Ning J, Zou L, Wang P, Zhang Y, Tian X, Zhang F, Zheng J, Ge G. Unique Oxidative Metabolism of Bufalin Generates Two Reactive Metabolites That Strongly Inactivate Human Cytochrome P450 3A. J Med Chem 2022; 65:4018-4029. [DOI: 10.1021/acs.jmedchem.1c01875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Dongzhu Tu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Ning
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Liwei Zou
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yani Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangge Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550025, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
6
|
Chen R, Guan Z, Zhong X, Zhang W, Zhang Y. Network Pharmacology Prediction: The Possible Mechanisms of Cinobufotalin against Osteosarcoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:3197402. [PMID: 35069780 PMCID: PMC8776428 DOI: 10.1155/2022/3197402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To explore the active compounds and targets of cinobufotalin (huachansu) compared with the osteosarcoma genes to obtain the potential therapeutic targets and pharmacological mechanisms of action of cinobufotalin on osteosarcoma through network pharmacology. METHODS The composition of cinobufotalin was searched by literature retrieval, and the target was selected from the CTD and TCMSP databases. The osteosarcoma genes, found from the GeneCards, OMIM, and other databases, were compared with the cinobufotalin targets to obtain potential therapeutic targets. The protein-protein interaction (PPI) network of potential therapeutic targets, constructed through the STRING database, was inputted into Cytoscape software to calculate the hub genes, using the NetworkAnalyzer. The hub genes were inputted into the Kaplan-Meier Plotter online database for exploring the survival curve. Functional enrichment analysis was identified using the DAVID database. RESULTS 28 main active compounds of cinobufotalin were explored, including bufalin, adenosine, oleic acid, and cinobufagin. 128 potential therapeutic targets on osteosarcoma are confirmed among 184 therapeutic targets form cinobufotalin. The hub genes included TP53, ACTB, AKT1, MYC, CASP3, JUN, TNF, VEGFA, HSP90AA1, and STAT3. Among the hub genes, TP53, ACTB, MYC, TNF, VEGFA, and STAT3 affect the patient survival prognosis of sarcoma. Through function enrichment analysis, it is found that the main mechanisms of cinobufotalin on osteosarcoma include promoting sarcoma apoptosis, regulating the cell cycle, and inhibiting proliferation and differentiation. CONCLUSION The possible mechanisms of cinobufotalin against osteosarcoma are preliminarily predicted through network pharmacology, and further experiments are needed to prove these predictions.
Collapse
Affiliation(s)
- Riyu Chen
- Guangzhou University of Chinese Medicine, 510000 Guangzhou, China
| | - Zeyi Guan
- Southern Medical University, 510000 Guangzhou, China
| | - Xianxing Zhong
- Guangzhou University of Chinese Medicine, 510000 Guangzhou, China
| | - Wenzheng Zhang
- Department of Joint Sports Medicine, Taian City Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 271000 Taian, China
| | - Ya Zhang
- Department of Pathology, The Second Affiliated Hospital of Shandong First Medical University, 271000 Taian, China
| |
Collapse
|
7
|
Lallier M, Marchandet L, Moukengue B, Charrier C, Baud’huin M, Verrecchia F, Ory B, Lamoureux F. Molecular Chaperones in Osteosarcoma: Diagnosis and Therapeutic Issues. Cells 2021; 10:cells10040754. [PMID: 33808130 PMCID: PMC8067202 DOI: 10.3390/cells10040754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/12/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is the most common form of primary bone tumor affecting mainly children and young adults. Despite therapeutic progress, the 5-year survival rate is 70%, but it drops drastically to 30% for poor responders to therapies or for patients with metastases. Identifying new therapeutic targets is thus essential. Heat Shock Proteins (HSPs) are the main effectors of Heat Shock Response (HSR), the expression of which is induced by stressors. HSPs are a large family of proteins involved in the folding and maturation of other proteins in order to maintain proteostasis. HSP overexpression is observed in many cancers, including breast, prostate, colorectal, lung, and ovarian, as well as OS. In this article we reviewed the significant role played by HSPs in molecular mechanisms leading to OS development and progression. HSPs are directly involved in OS cell proliferation, apoptosis inhibition, migration, and drug resistance. We focused on HSP27, HSP60, HSP70 and HSP90 and summarized their potential clinical uses in OS as either biomarkers for diagnosis or therapeutic targets. Finally, based on different types of cancer, we consider the advantage of targeting heat shock factor 1 (HSF1), the major transcriptional regulator of HSPs in OS.
Collapse
Affiliation(s)
- Morgane Lallier
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
| | - Louise Marchandet
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
| | - Brice Moukengue
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
| | - Celine Charrier
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
| | - Marc Baud’huin
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
- CHU Nantes, 44035 Nantes, France
| | - Franck Verrecchia
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
| | - Benjamin Ory
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
| | - François Lamoureux
- UMR1238, Phy-OS, Sarcomes Osseux et Remodelage des Tissus Calcifiés, INSERM, Université de Nantes, 44035 Nantes, France; (M.L.); (L.M.); (B.M.); (C.C.); (M.B.); (F.V.); (B.O.)
- Correspondence:
| |
Collapse
|
8
|
Bufalin down-regulates Axl expression to inhibit cell proliferation and induce apoptosis in non-small-cell lung cancer cells. Biosci Rep 2021; 40:222485. [PMID: 32219334 PMCID: PMC7146032 DOI: 10.1042/bsr20193959] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 12/29/2022] Open
Abstract
Axl, a member of the TAM (Tyro3, AXL, Mer) receptor tyrosine kinase family, plays critical roles in cell growth, proliferation, apoptosis, and migration. In the present study, we demonstrated that the anti-cancer activity of bufalin, a major bioactive component of the Chinese traditional medicine Chan Su, is mediated by the down-regulation of Axl in non-small-cell lung cancer (NSCLC) cells. We observed the inhibitory effect of bufalin on the proliferation of A549 and H460 NSCLC cells and the clonogenicity of these cells was reduced by bufalin treatment in a dose-dependent manner. Next, we found that the protein level of Axl was decreased in proportion to the concentration of bufalin in both A549 and H460 cells. Moreover, the promoter activity of the Axl gene was decreased by bufalin in a dose- and time-dependent manner, indicating that bufalin down-regulates Axl gene expression at the transcriptional level. We further examined if the anti-proliferative property of bufalin is influenced by Axl at the protein level. Axl overexpression attenuated the effect of bufalin in inhibiting cell proliferation and colony formation and inducing apoptosis in H460 cells, while knockdown of Axl gene expression induced the opposite effect. Taken together, our data indicate that the anti-proliferative and pro-apoptotic effects of bufalin were associated with the protein level of Axl, suggesting that Axl is a potent therapeutic target of bufalin in suppressing proliferation and inducing apoptosis in NSCLC cells.
Collapse
|
9
|
Deng LJ, Li Y, Qi M, Liu JS, Wang S, Hu LJ, Lei YH, Jiang RW, Chen WM, Qi Q, Tian HY, Han WL, Wu BJ, Chen JX, Ye WC, Zhang DM. Molecular mechanisms of bufadienolides and their novel strategies for cancer treatment. Eur J Pharmacol 2020; 887:173379. [PMID: 32758567 DOI: 10.1016/j.ejphar.2020.173379] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022]
Abstract
Bufadienolides are cardioactive C24 steroids with an α-pyrone ring at position C17. In the last ten years, accumulating studies have revealed the anticancer activities of bufadienolides and their underlying mechanisms, such as induction of autophagy and apoptosis, cell cycle disruption, inhibition of angiogenesis, epithelial-mesenchymal transition (EMT) and stemness, and multidrug resistance reversal. As Na+/K+-ATPase inhibitors, bufadienolides have inevitable cardiotoxicity. Short half-lives, poor stability, low plasma concentration and oral bioavailability in vivo are obstacles for their applications as drugs. To improve the drug potency of bufadienolides and reduce their side effects, prodrug strategies and drug delivery systems such as liposomes and nanoparticles have been applied. Therefore, systematic and recapitulated information about the antitumor activity of bufadienolides, with special emphasis on the molecular or cellular mechanisms, prodrug strategies and drug delivery systems, is of high interest. Here, we systematically review the anticancer effects of bufadienolides and the molecular or cellular mechanisms of action. Research advancements regarding bufadienolide prodrugs and their tumor-targeting delivery strategies are critically summarized. This work highlights recent scientific advances regarding bufadienolides as effective anticancer agents from 2011 to 2019, which will help researchers to understand the molecular pathways involving bufadienolides, resulting in a selective and safe new lead compound or therapeutic strategy with improved therapeutic applications of bufadienolides for cancer therapy.
Collapse
Affiliation(s)
- Li-Juan Deng
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China; School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yong Li
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Ming Qi
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Jun-Shan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Sheng Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Li-Jun Hu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Yu-He Lei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, 518034, China
| | - Ren-Wang Jiang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wei-Min Chen
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Qi Qi
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Hai-Yan Tian
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wei-Li Han
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Bao-Jian Wu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Wen-Cai Ye
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China.
| | - Dong-Mei Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
10
|
Bordon KDCF, Cologna CT, Fornari-Baldo EC, Pinheiro-Júnior EL, Cerni FA, Amorim FG, Anjolette FAP, Cordeiro FA, Wiezel GA, Cardoso IA, Ferreira IG, de Oliveira IS, Boldrini-França J, Pucca MB, Baldo MA, Arantes EC. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front Pharmacol 2020; 11:1132. [PMID: 32848750 PMCID: PMC7396678 DOI: 10.3389/fphar.2020.01132] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022] Open
Abstract
Animal poisons and venoms are comprised of different classes of molecules displaying wide-ranging pharmacological activities. This review aims to provide an in-depth view of toxin-based compounds from terrestrial and marine organisms used as diagnostic tools, experimental molecules to validate postulated therapeutic targets, drug libraries, prototypes for the design of drugs, cosmeceuticals, and therapeutic agents. However, making these molecules applicable requires extensive preclinical trials, with some applications also demanding clinical trials, in order to validate their molecular target, mechanism of action, effective dose, potential adverse effects, as well as other fundamental parameters. Here we go through the pitfalls for a toxin-based potential therapeutic drug to become eligible for clinical trials and marketing. The manuscript also presents an overview of the current picture for several molecules from different animal venoms and poisons (such as those from amphibians, cone snails, hymenopterans, scorpions, sea anemones, snakes, spiders, tetraodontiformes, bats, and shrews) that have been used in clinical trials. Advances and perspectives on the therapeutic potential of molecules from other underexploited animals, such as caterpillars and ticks, are also reported. The challenges faced during the lengthy and costly preclinical and clinical studies and how to overcome these hindrances are also discussed for that drug candidates going to the bedside. It covers most of the drugs developed using toxins, the molecules that have failed and those that are currently in clinical trials. The article presents a detailed overview of toxins that have been used as therapeutic agents, including their discovery, formulation, dosage, indications, main adverse effects, and pregnancy and breastfeeding prescription warnings. Toxins in diagnosis, as well as cosmeceuticals and atypical therapies (bee venom and leech therapies) are also reported. The level of cumulative and detailed information provided in this review may help pharmacists, physicians, biotechnologists, pharmacologists, and scientists interested in toxinology, drug discovery, and development of toxin-based products.
Collapse
Affiliation(s)
- Karla de Castro Figueiredo Bordon
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Takeno Cologna
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ernesto Lopes Pinheiro-Júnior
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe Augusto Cerni
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernanda Gobbi Amorim
- Postgraduate Program in Pharmaceutical Sciences, Vila Velha University, Vila Velha, Brazil
| | | | - Francielle Almeida Cordeiro
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Gisele Adriano Wiezel
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Iara Aimê Cardoso
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isabela Gobbo Ferreira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Isadora Sousa de Oliveira
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | | | - Mateus Amaral Baldo
- Health and Science Institute, Paulista University, São José do Rio Pardo, Brazil
| | - Eliane Candiani Arantes
- Laboratory of Animal Toxins, Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
11
|
Wei WL, Hou JJ, Wang X, Yu Y, Li HJ, Li ZW, Feng ZJ, Qu H, Wu WY, Guo DA. Venenum bufonis: An overview of its traditional use, natural product chemistry, pharmacology, pharmacokinetics and toxicology. JOURNAL OF ETHNOPHARMACOLOGY 2019; 237:215-235. [PMID: 30905791 DOI: 10.1016/j.jep.2019.03.042] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/16/2019] [Accepted: 03/16/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The animal medicine of Venenum Bufonis (VB), a product of the secretions of Bufo gargarizans Cantor or B. melanostictus Schneider, has long been used as a traditional Chinese medicine (TCM) for the treatment of sunstroke and faint, acute filthy disease - abdominal pain or vomiting and diarrhea, etc. AIM OF THE REVIEW: This review is aimed at providing the comprehensive and up-to-date information of VB as regards its ethnopharmacological uses, constituents and their metabolism, pharmacokinetics, pharmacology and toxicology, all of which could be used as fundamental data for future research as well as development of new drugs. MATERIALS AND METHODS The information and data about the studies of VB were collected from scientific journals, material medica, historical documents, library, and electronic databases (PubMed, Google Scholar, Science Direct, Researchgate, Web of Science and CNKI). RESULTS To date, about 142 bufadienolides and 16 indole alkaloids have been isolated from VB in total. The extract and isolated compounds showed a wide range of in vitro and in vivo pharmacologic effects, such as cardiotonic, anti-tumor, antinociceptive, anti-inflammatory, anesthetic and antimicrobial activities. Especially, bufadienolides have been extensively studied due to its powerful anti-tumor activities against various cancer cells. Furthermore, their metabolites and metabolic pathways were concluded in detail, and the main metabolic pathways of bufadienolides were hydroxylation, 3-isomerization, 3-keto, 16-hydrolyzation, 3-O-sulfate and 3-O-glucuronide. CONCLUSIONS Although VB possesses significant anti-tumor effect against various cancer cell lines, the development of new drugs still remains to be a challenge due to its pharmacodynamic effects in vivo, druggability and toxicology. The main problem lies in its side effects in vivo, poor bioavailability, fast metabolism, cardiotoxicity and neurovirulence. Besides, studies on its metabolism and toxicology in vitro and in vivo, as well as clinical trials should be further conducted for the new drug development and the establishment of optimal dosage of consumption of its administration.
Collapse
Affiliation(s)
- Wen-Long Wei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Jin-Jun Hou
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Xia Wang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Yang Yu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Hao-Jv Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Zhen-Wei Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Zi-Jin Feng
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Hua Qu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Wan-Ying Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China.
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China.
| |
Collapse
|
12
|
Cheng CS, Wang J, Chen J, Kuo KT, Tang J, Gao H, Chen L, Chen Z, Meng Z. New therapeutic aspects of steroidal cardiac glycosides: the anticancer properties of Huachansu and its main active constituent Bufalin. Cancer Cell Int 2019; 19:92. [PMID: 31011289 PMCID: PMC6458819 DOI: 10.1186/s12935-019-0806-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
Aim of the review In the past decade, increasing research attention investigated the novel therapeutic potential of steroidal cardiac glycosides in cancer treatment. Huachansu and its main active constituent Bufalin have been studied in vitro, in vivo and clinical studies. This review aims to summarize the multi-target and multi-pathway pharmacological effects of Bufalin and Huachansu in the last decade, with the aim of providing a more comprehensive view and highlighting the recently discovered molecular mechanisms. Results Huachansu and its major derivative, Bufalin, had been found to possess anti-cancer effects in a variety of cancer cell lines both in vitro and in vivo. The underlying anti-cancer molecular mechanisms mainly involved anti-proliferation, apoptosis induction, anti-metastasis, anti-angiogenesis, epithelial-mesenchymal transition inhibition, anti-inflammation, Na+/K+-ATPase activity targeting, the steroid receptor coactivator family inhibitions, etc. Moreover, the potential side-effects and toxicities of the toad extract, Huachansu, and Bufalin, including hematological, gastrointestinal, mucocutaneous and cardiovascular adverse reactions, were reported in animal studies and clinic trails. Conclusions Further research is needed to elucidate the potential drug-drug interactions and multi-target interaction of Bufalin and Huachansu. Large-scale clinical trials are warranted to translate the knowledge of the anticancer actions of Bufalin and Huachansu into clinical applications as effective and safe treatment options for cancer patients in the future.
Collapse
Affiliation(s)
- Chien-Shan Cheng
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Jiaqiang Wang
- 2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433 China.,5Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| | - Jie Chen
- 3School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China.,6Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Kuei Ting Kuo
- 3School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Jian Tang
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Huifeng Gao
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Lianyu Chen
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Zhen Chen
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Zhiqiang Meng
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| |
Collapse
|
13
|
Lan YL, Lou JC, Jiang XW, Wang X, Xing JS, Li S, Zhang B. A research update on the anticancer effects of bufalin and its derivatives. Oncol Lett 2019; 17:3635-3640. [PMID: 30915168 DOI: 10.3892/ol.2019.10062] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022] Open
Abstract
Bufalin (BF) is a cardiotonic steroid that has recently been found to have substantial anticancer activity; however, more efforts should be directed toward clarifying the detailed molecular mechanisms underlying this activity. BF could exert its anticancer effect by inducing apoptosis in various human cancer cells and thus triggering autophagic cancer cell death. The anti-inflammatory activities of BF are potentially important for its anticancer functions. Notably, some promising synthetic BF derivatives, including poly (ethylene glycol)-based polymeric prodrug of BF and BF211, have shown potent anticancer activity. Additionally, clinical trials regarding the use of BF-related agents in patients have supported the positive effect of BF as an anticancer treatment. Currently, large-scale randomized, double-blind, placebo or positive drug parallel controlled studies are required to confirm the anticancer potential of BF in various cancer types in the clinical setting. The present review will evaluate the potential mechanisms mediated by BF in intracellular signaling events in cancer cells and various promising BF derivatives that may have greater anticancer activity, thereby clarifying BF-mediated anticancer effects. The experimental and clinical results reviewed strongly emphasize the importance of this topic in future investigations.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China.,Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jia-Cheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Xue-Wen Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Xun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Jin-Shan Xing
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
14
|
Jing J, Tupally KR, Kokil GR, Qu Z, Chen S, Parekh HS. Development of a hybrid peptide dendrimer micellar carrier system and its application in the reformulation of a hydrophobic therapeutic agent derived from traditional Chinese medicine. RSC Adv 2019; 9:2458-2463. [PMID: 35520530 PMCID: PMC9059851 DOI: 10.1039/c8ra09606f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/06/2019] [Indexed: 11/29/2022] Open
Abstract
The discovery that a cane toad poison-derived steroid, bufalin can significantly impact cancer cell proliferation supports its potential use in cancer therapy. However, its poor aqueous solubility and tissue deposition characteristics hamper its broader application as an anticancer therapeutic agent in its own right. To address this we developed an amphiphilic dendrimer-based delivery system, which self-assembles into discrete micelles in an aqueous environment. The bufalin-micelle inclusion complex was prepared by the co-precipitation method and their presence was confirmed by dynamic light scattering (DLS), zeta potential and differential scanning calorimetry (DSC) and transmission electron microscopy (TEM) measurements. The self-assembled bufalin-containing micelles were found to form at/above the dendrimer concentration of 105.38 μmol L-1, and showed a more than threefold increase in the aqueous solubility (142.9 μg mL-1) of bufalin, when compared with a saturated bufalin aqueous solution (42.4 μg mL-1), and two non-assembling peptides of similar composition (79.3 and 62.5 μg mL-1 respectively).
Collapse
Affiliation(s)
- Jing Jing
- School of Pharmacy, The University of Queensland 20 Cornwall Street, Woolloongabba QLD 4012 Australia
| | - Karnaker R Tupally
- School of Pharmacy, The University of Queensland 20 Cornwall Street, Woolloongabba QLD 4012 Australia
| | - Ganesh R Kokil
- School of Pharmacy, The University of Queensland 20 Cornwall Street, Woolloongabba QLD 4012 Australia
| | - Zhi Qu
- School of Pharmacy, The University of Queensland 20 Cornwall Street, Woolloongabba QLD 4012 Australia
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute Shenzhen 518057 People's Republic of China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100193 People's Republic of China
| | - Harendra S Parekh
- School of Pharmacy, The University of Queensland 20 Cornwall Street, Woolloongabba QLD 4012 Australia
| |
Collapse
|
15
|
Yang YY, Yang FQ, Gao JL. Differential proteomics for studying action mechanisms of traditional Chinese medicines. Chin Med 2019; 14:1. [PMID: 30636970 PMCID: PMC6325846 DOI: 10.1186/s13020-018-0223-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022] Open
Abstract
Differential proteomics, which has been widely used in studying of traditional Chinese medicines (TCMs) during the past 10 years, is a powerful tool to visualize differentially expressed proteins and analyzes their functions. In this paper, the applications of differential proteomics in exploring the action mechanisms of TCMs on various diseases including cancers, cardiovascular diseases, diabetes, liver diseases, kidney disorders and obesity, etc. were reviewed. Furthermore, differential proteomics in studying of TCMs identification, toxicity, processing and compatibility mechanisms were also included. This review will provide information for the further applications of differential proteomics in TCMs studies.
Collapse
Affiliation(s)
- Yi-Yao Yang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People’s Republic of China
| | - Feng-Qing Yang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing, 401331 People’s Republic of China
| | - Jian-Li Gao
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang People’s Republic of China
| |
Collapse
|
16
|
Chinese Herbal Medicine for Osteosarcoma in the Mouse: A Systematic Review and Meta-Analysis. Chin J Integr Med 2018; 25:370-377. [PMID: 30484018 DOI: 10.1007/s11655-018-2565-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2016] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To summarize and critically assess the inhibitory effects of Chinese herbal medicine (CHM) on tumor volume and tumor weight for the treatment of osteosarcoma (OS) in mouse models. METHODS PubMed, Embase, Web of Science, China Knowledge Resource Integrated Database (CNKI), Wanfang Database, VIP Database, and Chinese BioMedical (CBM) were searched since their inception dates to March 10, 2016. Two reviewers independently selected the controlled studies estimating effects of CHM on mouse OS by administration in vivo. A pair-wise meta-analysis was performed. Twenty-five studies with adequate randomization were included in the systematic review. RESULTS CHM may significantly inhibit OS growth in mice, as assessed using the tumor weight [20 studies, n=443; 290 for CHM and 153 for the control: pooled mean difference (MD)=-2.90; 95% confidence interval (Cl): -3.50 to -2.31: P<0.01], tumor volume (16 studies, n=382; 257 for CHM and 125 for the control; pooled MD =-2.57; 95% Cl: -3.33 to -1.80; P<0.01) and tumor growth inhibition rate. CONCLUSION CHM could significantly inhibit the growth of OS in mouse models, which might be supportive for the design of preclinical and clinical trials in future.
Collapse
|
17
|
Livingston JA, Wang WL, Tsai JW, Lazar AJ, Leung CH, Lin H, Advani S, Daw N, Santiago-O'Farrill J, Hollomon M, Gordon NB, Kleinerman ES. Analysis of HSP27 and the Autophagy Marker LC3B + Puncta Following Preoperative Chemotherapy Identifies High-Risk Osteosarcoma Patients. Mol Cancer Ther 2018; 17:1315-1323. [PMID: 29592877 PMCID: PMC5984702 DOI: 10.1158/1535-7163.mct-17-0901] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/11/2018] [Accepted: 03/20/2018] [Indexed: 12/30/2022]
Abstract
Chemotherapy-induced autophagy is a proposed mechanism of chemoresistance and potential therapeutic target in osteosarcoma. We evaluated heat shock protein 27 (HSP27) and autophagy-related proteins as predictors of pathologic treatment response and prognostic markers among osteosarcoma patients who received standard chemotherapy. We analyzed 394 tumor specimens (pre-treatment, post-treatment, and metastases) from 260 osteosarcoma patients by immunohistochemistry for cytoplasmic light chain 3B (LC3B)-positive puncta, sequestosome 1 (SQSTM1), high mobility group box 1 (HMGB1), and HSP27 expression. The staining percentage and intensity for each marker were scored and the extent to which marker expression was correlated with pathologic response, relapse-free survival (RFS), and overall survival (OS) was assessed. LCB3+ puncta in post-treatment primary tumors (50%) and metastases (67%) was significantly higher than in pre-treatment biopsy specimens (30%; P = 0.023 and <0.001). Among 215 patients with localized osteosarcoma, both pre-treatment [multivariate hazard ratio (HR), 26.7; 95% confidence interval (CI), 1.47-484; P = 0.026] and post-treatment HSP27 expression (multivariate HR, 1.85; 95% CI, 1.03-3.33; P = 0.039) were associated with worse OS. Lack of LC3B+ puncta at resection was an independent poor prognostic marker in both univariate (HR, 1.78; 95% CI, 1.05-3.03; P = 0.034) and multivariate models (HR, 1.75; 95% CI, 1.01-3.04; P = 0.045). Patients with LC3B+/HSP27- tumors at resection had the best 10-year OS (75%) whereas patients with LC3B-/HSP27+ tumors had the worst 10-year survival (25%). Neither HSP27 expression nor the presence of LCB3+ puncta was correlated with pathologic treatment response. Our findings establish HSP27 expression and LC3B+ puncta as independent prognostic markers in osteosarcoma patients receiving standard chemotherapy and support further investigation into strategies targeting HSP27 or modulating autophagy in osteosarcoma treatment. Mol Cancer Ther; 17(6); 1315-23. ©2018 AACR.
Collapse
Affiliation(s)
- J Andrew Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei-Lien Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jen-Wei Tsai
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cheuk Hong Leung
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shailesh Advani
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Najat Daw
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Mario Hollomon
- Department of Biology, Texas Southern University, Houston, Texas
| | - Nancy B Gordon
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eugenie S Kleinerman
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
18
|
Lan YL, Wang X, Lou JC, Xing JS, Yu ZL, Wang H, Zou S, Ma X, Zhang B. Bufalin inhibits glioblastoma growth by promoting proteasomal degradation of the Na +/K +-ATPase α1 subunit. Biomed Pharmacother 2018; 103:204-215. [PMID: 29653366 DOI: 10.1016/j.biopha.2018.04.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 01/02/2023] Open
Abstract
Chansu is a traditional Chinese medicine that is generally recognized as a specific inhibitor of Na+/K+-ATPase. Bufalin, an active component of Chansu, is an endogenous steroid hormone with great potential as a cancer treatment. However, the mechanism by which it exerts its antitumor activity requires further research. Currently, the α1 subunit of Na+/K+-ATPase (ATP1A1) is known to exert important roles in tumorigenesis, and the precise mechanisms underlying the effect of Bufalin on the Na+/K+-ATPase α1 subunit was therefore investigated in this study to determine its role in glioblastoma treatments. The effect of ATP1A1 on the sensitivity of glioblastoma cells to Bufalin was investigated using MTT assays, RT-PCR and siRNA. Western blot was also used to explore the important roles of the ubiquitin-proteasome pathway in the Bufalin-mediated inhibition of ATP1A1. Xenografted mice were used to examine the anti-tumor activity of Bufalin in vivo. LC-MS/MS analysis was performed to determine the ability of Bufalin to traverse the blood-brain barrier (BBB). The results indicated that Bufalin inhibited the expression of ATP1A1 in glioblastoma by promoting the activation of proteasomes and the subsequent protein degradation of ATP1A1, while Bufalin had no effect on ATP1A1 protein synthesis. Bufalin also inhibited the expression of ATP1A1 in xenografted mice and significantly suppressed tumor growth. These data should contribute to future basic and clinical investigations of Bufalin. In conclusion, Bufalin significantly inhibited the expression of ATP1A1 in glioblastoma cells by activating the ubiquitin-proteasome signaling pathway. Bufalin may therefore have the potential to be an effective anti-glioma drug for human glioblastoma in the future.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, China; Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China; Department of Pharmacy, Dalian Medical University, Dalian, 116044, China; Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | - Xun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, China
| | - Jia-Cheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, China
| | - Jin-Shan Xing
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, China
| | - Zhen-Long Yu
- Department of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Hongjin Wang
- Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Shuang Zou
- Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | - Xiaochi Ma
- Department of Pharmacy, Dalian Medical University, Dalian, 116044, China.
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhong Shan Road, Dalian, 116023, China.
| |
Collapse
|
19
|
Shi XJ, Qiu YY, Yu H, Liu C, Yuan YX, Yin PH, Liu T. Increasing the anticancer performance of bufalin (BUF) by introducing an endosome-escaping polymer and tumor-targeting peptide in the design of a polymeric prodrug. Colloids Surf B Biointerfaces 2018; 166:224-234. [PMID: 29602078 DOI: 10.1016/j.colsurfb.2018.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 03/15/2018] [Accepted: 03/17/2018] [Indexed: 12/26/2022]
Abstract
A well-defined multifunctional brush-type polymeric prodrug covalently linked with an anticancer drug (bufalin, BUF), a tumor-targeting peptide (RGD), and an endosome-escaping polymer, poly(N,N-diethylaminoethyl methacrylate-co-butyl methacrylate (P(DEA-co-BMA)), was developed. Its anticancer performance against colon cancer was investigated in vitro and in vivo. Reversible addition-fragmentation transfer (RAFT) polymerization of oligo(ethylene glycol) monomethyl ether methacrylate (OEGMA), 2-((3-(tert-butoxy)-3-oxopropyl)thio)ethyl methacrylate (BSTMA), and 2-(2-bromoisobutyryloxy)ethylmethacrylate (BIEM) afforded the multifunctional random copolymer, P(OEGMA-co-BSTMA-co-BIEM), in which hydrophilic POEGMA can stabilize nanoparticles in water, PBSTMA can be converted into carboxyl groups, and PBIEM can be employed as a macromolecular atom radical transfer polymerization (ATRP) initiator. The ATRP of DEA and BMA using P(OEGMA-co-BSTMA-co-BIEM) as a macromolecular ATRP initiator led to the formation of the pH-responsive brush-type copolymer, P(OEGMA-co-BSTMA)-g-P(DEA-co- BMA). After hydrolysis by trifluoroacetic acid and post-functionalization the final polymeric prodrug, P(OEGMA-co-BUF-co-RGD)-g-P(DEA-co-BMA), was obtained with a drug content of ∼7.8 wt%. P(OEGMA-co-BUF-co-RGD)-g-P(DEA-co-BMA) can be assembled into nanoparticles (BUF- NP-RGD) in aqueous solution with a diameter of 148.4 ± 0.7 nm and a zeta potential of -7.6 ± 0.4 mV. BUF-NP-RGD exhibited controlled drug release in the presence of esterase. Additionally, P(OEGMA-co- BSMA)-g-P(DEA-co-BMA) showed a significant hemolysis effect at a pH comparable to that of endosomes/lysosomes. Cell viability and a tumor-bearing nude mouse model were employed to evaluate the anticancer efficacy of BUF-NP-RGD. It was revealed that BUF-NP-RGD showed improved anticancer performance compared with that of free BUF both in vitro and in vivo. Histological and immunochemical analysis further demonstrated that BUF-NP-RGD exhibited improved cell apoptosis, angiogenesis inhibition, and an anti-proliferation effect.
Collapse
Affiliation(s)
- Xiao-Jing Shi
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yan-Yan Qiu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Hui Yu
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Cheng Liu
- Centralab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yu-Xia Yuan
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Pei-Hao Yin
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Tao Liu
- Centralab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
20
|
Fujii E, Inada Y, Kakoki M, Nishimura N, Endo S, Fujiwara S, Wada N, Kawano Y, Okuno Y, Sugimoto T, Hata H. Bufalin induces DNA damage response under hypoxic condition in myeloma cells. Oncol Lett 2018; 15:6443-6449. [PMID: 29616114 PMCID: PMC5876453 DOI: 10.3892/ol.2018.8091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/13/2018] [Indexed: 12/13/2022] Open
Abstract
Hypoxia serves a crucial role in the development of drug resistance in various cancer cells. Therefore, many attempts targeting hypoxia are underway to overcome the drug resistance mediated by hypoxia. This strategy is useful for multiple myeloma (MM) cells, as MM cells reside within the bone marrow, where oxygen concentrations are relatively low. A natural compound library was screened to identify compounds exerting cytotoxicity in MM cells under hypoxic conditions. Bufalin exhibited marked cytotoxicity to MM cells under normoxic and hypoxic conditions. No significant toxicity was observed in lymphocytes obtained from healthy donors. Under normoxic conditions, bufalin induced a DNA double strand break (DSB) response, ROS induction and apoptosis within 24 with a rapid response compared with melphalan. Interestingly, the bufalin-induced DSB response was not impaired by low oxygen concentrations while the DSB response by melphalan was reduced. Furthermore, treatment with bufalin abolished HIF-1α expression under hypoxia, suggesting that bufalin exerts cytotoxicity under hypoxia by regulating HIF-1α. These results indicate that bufalin induces apoptosis in MM cells through DSB under hypoxic conditions by inhibiting HIF-1α, suggesting that bufalin could be useful for eradication of drug-resistant MM cells in the hypoxic microenvironment.
Collapse
Affiliation(s)
- Eri Fujii
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan.,Department of Clinical Laboratory, Osaka University Hospital, Suita, Osaka 565-0871, Japan
| | - Yuki Inada
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Misaki Kakoki
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Nao Nishimura
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Shinya Endo
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Shiho Fujiwara
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Naoko Wada
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yawara Kawano
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yutaka Okuno
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Toshiya Sugimoto
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Hiroyuki Hata
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.,Division of Informative Clinical Sciences, Faculty of Medical Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| |
Collapse
|
21
|
Yu Z, Li T, Wang C, Deng S, Zhang B, Huo X, Zhang B, Wang X, Zhong Y, Ma X. Gamabufotalin triggers c-Myc degradation via induction of WWP2 in multiple myeloma cells. Oncotarget 2017; 7:15725-37. [PMID: 26894970 PMCID: PMC4941272 DOI: 10.18632/oncotarget.7398] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/05/2016] [Indexed: 12/14/2022] Open
Abstract
Deciding appropriate therapy for multiple myeloma (MM) is challenging because of the occurrence of multiple chromosomal changes and the fatal nature of the disease. In the current study, gamabufotalin (GBT) was isolated from toad venom, and its tumor-specific cytotoxicity was investigated in human MM cells. We found GBT inhibited cell growth and induced apoptosis with the IC50 values <50 nM. Mechanistic studies using functional approaches identified GBT as an inhibitor of c-Myc. Further analysis showed that GBT especially evoked the ubiquitination and degradation of c-Myc protein, thereby globally repressing the expression of c-Myc target genes. GBT treatment inhibited ERK and AKT signals, while stimulating the activation of JNK cascade. An E3 ubiquitin-protein ligase, WWP2, was upregulated following JNK activation and played an important role in c-Myc ubiquitination and degradation through direct protein-protein interaction. The antitumor effect of GBT was validated in a xenograft mouse model and the suppression of MM-induced osteolysis was verified in a SCID-hu model in vivo. Taken together, our study identified the potential of GBT as a promising therapeutic agent in the treatment of MM.
Collapse
Affiliation(s)
- Zhenlong Yu
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Tao Li
- Department of Biology, College of Chemistry and Life Sciences, Zhejiang Normal University, Zhejiang, China
| | - Chao Wang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Sa Deng
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Baojing Zhang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiaokui Huo
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaobo Wang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Yuping Zhong
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaochi Ma
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
22
|
Zhao Z, Jia Q, Wu MS, Xie X, Wang Y, Song G, Zou CY, Tang Q, Lu J, Huang G, Wang J, Lin DC, Koeffler HP, Yin JQ, Shen J. Degalactotigonin, a Natural Compound from Solanum nigrum L., Inhibits Growth and Metastasis of Osteosarcoma through GSK3β Inactivation-Mediated Repression of the Hedgehog/Gli1 Pathway. Clin Cancer Res 2017; 24:130-144. [PMID: 28951519 DOI: 10.1158/1078-0432.ccr-17-0692] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/23/2017] [Accepted: 09/20/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Agents extracted from natural sources with antitumor property have attracted considerable attention from researchers and clinicians because of their safety, efficacy, and immediate availability. Degalactotigonin (DGT), extracted from Solanum nigrum L, has anticancer properties without serious side effects. Here, we explored whether DGT can inhibit the growth and metastasis of osteosarcoma.Experimental Design: MTT, colony formation, and apoptosis assays were performed to analyze the effects of DGT on osteosarcoma cell viability in vitro The migration and invasion abilities were measured using a Transwell assay. Animal models were used to assess the roles of DGT in both tumor growth and metastasis of osteosarcoma. Gli1 expression and function were measured in osteosarcoma cells and clinical samples. After DGT treatment, Gli1 activation and the phosphorylation status of multiple cellular kinases were measured with a luciferase reporter and phospho-kinase antibody array.Results: DGT inhibited proliferation, induced apoptosis, and suppressed migration and invasion in osteosarcoma cells. DGT, injected intraperitoneally after tumor inoculation, significantly decreased the volume of osteosarcoma xenografts and dramatically diminished the occurrence of osteosarcoma xenograft metastasis to the lungs. Mechanistically, DGT inhibited osteosarcoma growth and metastasis through repression of the Hedgehog/Gli1 pathway, which maintains malignant phenotypes and is involved in the prognosis of osteosarcoma patients. DGT decreased the activity of multiple intracellular kinases that affect the survival of osteosarcoma patients, including GSK3β. In addition, DGT represses the Hedgehog/Gli1 pathway mainly through GSK3β inactivation.Conclusions: Our studies provide evidence that DGT can suppress the growth and metastasis of human osteosarcoma through modulation of GSK3β inactivation-mediated repression of the Hedgehog/Gli1 pathway. Clin Cancer Res; 24(1); 130-44. ©2017 AACR.
Collapse
Affiliation(s)
- Zhiqiang Zhao
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiang Jia
- Guangzhou City Polytechnic, Guangzhou, China.,Institute of Biology, Guizhou Academy of Sciences, Guiyang, China
| | - Man-Si Wu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China. .,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yongqian Wang
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guohui Song
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chang-Ye Zou
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Gang Huang
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jin Wang
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - De-Chen Lin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - H Phillip Koeffler
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jun-Qiang Yin
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
23
|
Bufalin attenuates cancer-induced pain and bone destruction in a model of bone cancer. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1211-1219. [PMID: 28840279 DOI: 10.1007/s00210-017-1419-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/10/2017] [Indexed: 01/01/2023]
Abstract
Bufalin is a natural anti-inflammatory small molecule. Given the close relationship between inflammation and cancer, many scholars have studied the effect of bufalin on cancer in vitro, but in vivo research is still lacking. A murine bone cancer model was used in this study. We conducted pain sensitive test on mice with bone cancer, by nocifensive behavior, mechanical allodynia, and thermal hyperalgesia. Serum levels of bone loss markers with bufalin treatment were measured by ELISA. Expressions of osteoprotegerin (OPG) and receptor activator of NF-κB ligand (RANKL) were analyzed in bufalin-treated mice by real-time PCR and Western blot. Cannabinoid 2 receptor (CB2) inverse agonist AM630 was administrated to mice with bone cancer together with bufalin. Bufalin relieved cancer-induced pain and bone destruction in the murine bone cancer model. Serum levels of bone loss markers after bufalin treatment were reduced. Bufalin upregulated OPG and downregulated RANKL. The CB2 receptor inverse agonist, AM630, reduced the pain relief of bufalin treatment in the mouse bone cancer model. This study demonstrates that bufalin relieves cancer-induced pain and bone destruction, which is mediated through the CB2 receptor.
Collapse
|
24
|
Wu SH, Bau DT, Hsiao YT, Lu KW, Hsia TC, Lien JC, Ko YC, Hsu WH, Yang ST, Huang YP, Chung JG. Bufalin induces apoptosis in vitro and has Antitumor activity against human lung cancer xenografts in vivo. ENVIRONMENTAL TOXICOLOGY 2017; 32:1305-1317. [PMID: 27444971 DOI: 10.1002/tox.22325] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 06/06/2023]
Abstract
Bufalin has been shown to be effective against a variety of cancer cells, but its role in lung cancer has never been studied in an animal model. In this study, we evaluated bufalin effects in a human lung cancer cell line NCI-H460 both in vitro and in vivo. Bufalin caused significant cytotoxicity in NCI-H460 cells at a concentration as low as 1 μM. DNA condensation was observed in bufalin-treated cells in a dose-dependent manner. Mitochondrial membrane potential (ΔΨm ) was reduced and reactive oxygen species (ROS) were increased in bufalin-treated NCI-H460 cells. Levels of several proapoptotic proteins such as Fas, Fas-ligand, cytochrome c, apoptosis protease activating factor-1, endonuclease G, caspase-3 and caspase-9 were increased after bufalin treatment. At the same time, anti-apoptotic B-cell lymphoma 2 protein levels were reduced. Bufalin decreased glucose regulated protein-78 gene expression but increased growth arrest- and DNA damage-inducible 153 gene expression. Bufalin injected intraperitoneally in a dose-dependent manner reduced tumor size in BALB/C nu/nu mice implanted with NCI-H460 cells. Bufalin injection did not produce significant drug-related toxicity in experimental animals except at a high dose (0.4 mg kg-1 ). In conclusion, low concentrations of bufalin can induce apoptosis in the human lung cancer cell line NCI-H460 in vitro. Bufalin also reduced tumor size in mice injected with NCI-H460 cells without significant drug-related toxicity. These results indicate that bufalin may have potential to be developed as an agent for treating human non-small cell lung cancer. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1305-1317, 2017.
Collapse
Affiliation(s)
- Shin-Hwar Wu
- Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan
- Division of Critical Care Medicine, Department of Medicine, Changhua Christian Hospital, Changhua, 500, Taiwan
| | - Da-Tian Bau
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, 404, Taiwan
| | - Yung-Ting Hsiao
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Kung-Wen Lu
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Te-Chun Hsia
- Department of Internal Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung, 40402, Taiwan
| | - Yang-Ching Ko
- Institute of Clinical Medical Science, China Medical University, Taichung, 404, Taiwan
| | - Wu-Huei Hsu
- Department of Internal Medicine, China Medical University, Taichung, 404, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung, 404, Taiwan
| | - Su-Tso Yang
- Department of Radiology, China Medical University Hospital, Taichung, 404, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Yi-Ping Huang
- Department of Physiology, China Medical University, Taichun, 404, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Taichung, 404, Taiwan
| |
Collapse
|
25
|
Lee CH, Shih YL, Lee MH, Au MK, Chen YL, Lu HF, Chung JG. Bufalin Induces Apoptosis of Human Osteosarcoma U-2 OS Cells through Endoplasmic Reticulum Stress, Caspase- and Mitochondria-Dependent Signaling Pathways. Molecules 2017; 22:molecules22030437. [PMID: 28287444 PMCID: PMC6155407 DOI: 10.3390/molecules22030437] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 12/22/2022] Open
Abstract
Bone cancer is one of the cancer-related diseases, and there are increased numbers of patients with bone cancer worldwide. Therefore the efficacy of treatment of bone cancer is considered extremely vital. Bufalin has been showed to have biological activities including anticancer activities in vitro and in vivo. However, the exact associated mechanisms for bufalin induced apoptosis in human bone cancer cells are still unclear. In the present study, we investigated the effect of bufalin on the cytotoxic effects in U-2 OS human osteosarcoma cells. For examining apoptotic cell deaths, we used flow cytometry assay, Annexin V/PI double staining, and TUNNEL assay. Reactive oxygen species (ROS), Ca2+, mitochondrial membrane potential (ΔΨm), and caspase-8, -9 and -3 activities were measured by flow cytometry assay. Furthermore, western blotting and a confocal laser microscopy examination were used for measuring the alterations of apoptotic associated protein expression and translocation, respectively. The results indicated that bufalin induced cell morphological changes, decreased the viable cell number, induced apoptotic cell death, and increased the apoptotic cell number, and affected apoptotic associated protein expression in U-2 OS cells. Bufalin increased apoptotic proteins such as Bak, and decreased anti-apoptotic proteins such as Bcl-2 and Bcl-x in U-2 OS cells. Furthermore, bufalin increased the protein levels of cytochrome c (Cyto c), AIF (Apoptosis inducing factor) and Endo G (Endonuclease G) in cytoplasm that were also confirmed by confocal microscopy examination. Based on those findings, bufalin induced apoptotic cell death in U-2 OS cells may be via endoplasmic reticulum (ER) stress, caspase-, and mitochondria-dependent pathways; thus, we may suggest that bufalin could be used as an anti-cancer agent for the treatment of osteosarcoma in the future, and further in vivo studies are needed.
Collapse
Affiliation(s)
- Ching-Hsiao Lee
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli Country 356, Taiwan.
| | - Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan.
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 110, Taiwan.
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan.
| | - Mei-Hui Lee
- Department of Genetic Counseling Center, Changhua Christian Hospital, Changhua 500, Taiwan.
| | - Man-Kuan Au
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei 112, Taiwan.
| | - Yung-Liang Chen
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu 300, Taiwan.
| | - Hsu-Feng Lu
- Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei City 242, Taiwan.
- Department of Clinical Pathology, Cheng Hsin General Hospital, Taipei 112, Taiwan.
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung 404, Taiwan.
- Department of Biotechnology, Asia University, Wufeng, Taichung 413, Taiwan.
| |
Collapse
|
26
|
Chaiyawat P, Settakorn J, Sangsin A, Teeyakasem P, Klangjorhor J, Soongkhaw A, Pruksakorn D. Exploring targeted therapy of osteosarcoma using proteomics data. Onco Targets Ther 2017; 10:565-577. [PMID: 28203090 PMCID: PMC5295800 DOI: 10.2147/ott.s119993] [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] [Indexed: 12/17/2022] Open
Abstract
Despite multimodal therapeutic treatments of osteosarcoma (OS), some patients develop resistance to currently available regimens and eventually end up with recurrent or metastatic outcomes. Many attempts have been made to discover effective drugs for improving outcome; however, due to the heterogeneity of the disease, new therapeutic options have not yet been identified. This study aims to explore potential targeted therapy related to protein profiles of OS. In this review of proteomics studies, we extracted data on differentially expressed proteins (DEPs) from archived literature in PubMed and our in-house repository. The data were divided into three experimental groups, DEPs in 1) OS/OB: OS vs osteoblastic (OB) cells, 2) metastasis: metastatic vs non-metastatic sublines plus fresh tissues from primary OS with and without pulmonary metastasis, and 3) chemoresistance: spheroid (higher chemoresistance) vs monolayer cells plus fresh tissues from biopsies from good and poor responders. All up-regulated protein entities in the list of DEPs were sorted and cross-referenced with identifiers of targets of US Food and Drug Administration (FDA)-approved agents and chemical inhibitors. We found that many targets of FDA-approved antineoplastic agents, mainly a group of epigenetic regulators, kinases, and proteasomes, were highly expressed in OS cells. Additionally, some overexpressed proteins were targets of FDA-approved non-cancer drugs, including immunosuppressive and antiarrhythmic drugs. The resulting list of chemical agents showed that some transferase enzyme inhibitors might have anticancer activity. We also explored common targets of OS/OB and metastasis groups, including amidophosphoribosyltransferase (PPAT), l-lactate dehydrogenase B chain (LDHB), and pyruvate kinase M2 (PKM2) as well as the common target of all categories, cathepsin D (CTSD). This study demonstrates the benefits of a text mining approach to exploring therapeutic targets related to protein expression patterns. These results suggest possible repurposing of some FDA-approved medicines for the treatment of OS and using chemical inhibitors in drug screening tests.
Collapse
Affiliation(s)
- Parunya Chaiyawat
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics
| | | | - Apiruk Sangsin
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics
| | - Pimpisa Teeyakasem
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics
| | | | | | - Dumnoensun Pruksakorn
- Orthopedic Laboratory and Research Netting Center, Department of Orthopedics; Excellence Center in Osteology Research and Training Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
27
|
Polymeric prodrug of bufalin for increasing solubility and stability: Synthesis and anticancer study in vitro and in vivo. Int J Pharm 2016; 506:382-93. [DOI: 10.1016/j.ijpharm.2016.04.041] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 04/01/2016] [Accepted: 04/15/2016] [Indexed: 11/21/2022]
|
28
|
Liu T, Jia T, Yuan X, Liu C, Sun J, Ni Z, Xu J, Wang X, Yuan Y. Development of octreotide-conjugated polymeric prodrug of bufalin for targeted delivery to somatostatin receptor 2 overexpressing breast cancer in vitro and in vivo. Int J Nanomedicine 2016; 11:2235-50. [PMID: 27284243 PMCID: PMC4883818 DOI: 10.2147/ijn.s100404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Development of polymeric prodrugs of small molecular anticancer drugs has become one of the most promising strategies to overcome the intrinsic shortcomings of small molecular anticancer drugs and improve their anticancer performance. MATERIALS AND METHODS In the current work, we fabricated a novel octreotide (Oct)-modified esterase-sensitive tumor-targeting polymeric prodrug of bufalin (BUF) and explored its anticancer performance against somatostatin receptor 2 overexpressing breast cancer. RESULTS The obtained tumor-targeting polymeric prodrug of BUF, P(oligo[ethylene glycol] monomethyl ether methacrylate [OEGMA]-co-BUF-co-Oct), showed a nanosize dimension and controlled drug release features in the presence of esterase. It was demonstrated by in vitro experiment that P(OEGMA-co-BUF-co-Oct) showed enhanced cytotoxicity, cellular uptake, and apoptosis in comparison with those of free BUF. In vivo experiment further revealed the improved accumulation of drugs in tumor tissues and enhanced anticancer performance of P(OEGMA-co-BUF-co-Oct). CONCLUSION Taken together, this study indicated that polymeric prodrug of BUF holds promising potential toward the treatment of somatostatin receptor 2 overexpressing breast cancer.
Collapse
Affiliation(s)
- Tao Liu
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Tingting Jia
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xia Yuan
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Cheng Liu
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jian Sun
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhenhua Ni
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jian Xu
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xuhui Wang
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yi Yuan
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
29
|
Ma K, Zhang C, Huang MY, Li WY, Hu GQ. Cinobufagin induces autophagy-mediated cell death in human osteosarcoma U2OS cells through the ROS/JNK/p38 signaling pathway. Oncol Rep 2016; 36:90-8. [PMID: 27176794 PMCID: PMC4899018 DOI: 10.3892/or.2016.4782] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
The main objective of this study was to explore whether autophagy could be triggered by cinobufagin, and to clarify the role of autophagy in the antitumor effects of cinobufagin on U2OS cells and the underlying mechanisms. U2OS cells were exposed to 15, 30, 60 and 120 mg/l cinobufagin for 0, 12, 24 and 48 h. An MTT assay was used to measure cell viability. FITC-Annexin Ⅴ/PI staining and flow cytometry were used to analyze the apoptotic ratio, while apoptotic morphological changes were assessed by PI and Hoechst 33258 viable cell staining. The effects of autophagy on the cells were investigated with GFP-LC3b green fluorescence plasmid transfection and transmission electron microscopy. The levels of caspase-3, -8, - 9, cleaved PARP, LC3-II/LC3-I, p62 and the activation of JNK/p-38 were detected by western blot analysis. Reactive oxygen species (ROS) fluorescence intensity was examined under fluorescence microscopy with an analysis software system. Cell proliferation was obviously inhibited by cinobufagin in a dose- and time-dependent manner. The apoptosis ratio was gradually increased with treatment time as evidenced by flow cytometric analysis and Hoechst 33258 staining. Exposure to cinobufagin resulted in the activation of caspase-3, -8, -9, as well as cleaved PARP which indicated that cinobufagin induced caspase-dependent apoptosis. Autophagy was confirmed in the cinobufagin-treated cells as evidenced by formation of autophagosomes, accumulation of GFP-LC3 fluorescence particles as well as the upregulation of LC3-II/LC3-I levels. Inhibition of autophagy diminished apoptosis as detected by the MTT assays. Moreover the percentage of apoptotic cells decreased following pretreatment with 3-MA, CQ and si-beclin-1. Cinobufagin also induced phosphorylation of the JNK and p38 signaling pathway as well as ROS generation. The JNK and p38 inhibitors significantly attenuated coexistence of apoptosis and autophagy-related proteins. The ROS scavenger also prevented phosphorylation of the JNK and p38 signaling pathway. Our research proved that cinobufagin triggered apoptosis and autophagic cell death via activation of the ROS/JNK/p-38 axis.
Collapse
Affiliation(s)
- Kun Ma
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Chuan Zhang
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Man-Yu Huang
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Wu-Yin Li
- Luoyang Orthopaedic-Traumatological Hospital and Henan Orthopaedic Hospital, Luoyang, Henan 471002, P.R. China
| | - Guo-Qiang Hu
- College of Pharmacy, Henan University, Kaifeng, Henan 475000, P.R. China
| |
Collapse
|
30
|
Zhang DM, Feng LX, Liu M, Jin WH, Luo J, Nie AY, Zhou Y, Li Y, Wu WY, Jiang BH, Yang M, Hu LH, Guo DA, Liu X. Possible target-related proteins and signal network of bufalin in A549 cells suggested by both iTRAQ-based and label-free proteomic analysis. Proteomics 2016; 16:935-45. [DOI: 10.1002/pmic.201500418] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/03/2015] [Accepted: 01/15/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Dong-Mei Zhang
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
- Department of Pharmacy; Lanzhou General Hospital of Lanzhou Military Command of Chinese PLA; Lanzhou P. R. China
| | - Li-Xing Feng
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| | - Miao Liu
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| | | | - Ji Luo
- AB Sciex; Shanghai P. R. China
| | - Ai-Ying Nie
- Thermo Fisher Scientific; Shanghai P. R. China
| | - Yue Zhou
- Thermo Fisher Scientific; Shanghai P. R. China
| | - Yin Li
- Thomson Reuters; Shanghai P. R. China
| | - Wan-Ying Wu
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| | - Bao-Hong Jiang
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| | - Min Yang
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| | - Li-Hong Hu
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| | - De-An Guo
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| | - Xuan Liu
- Shanghai Institute of Materia Medica; Chinese Academy of Sciences; Shanghai P. R. China
| |
Collapse
|
31
|
Delebinski CI, Georgi S, Kleinsimon S, Twardziok M, Kopp B, Melzig MF, Seifert G. Analysis of proliferation and apoptotic induction by 20 steroid glycosides in 143B osteosarcoma cells in vitro. Cell Prolif 2015; 48:600-10. [PMID: 26300346 DOI: 10.1111/cpr.12208] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/04/2015] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Osteosarcoma is the most common type of malignant bone tumour in children and adolescents; it has poor prognosis, is highly metastatic and is resistant to current therapeutic approaches. In this study, different herbal extracts used in phytotherapy have been screened after searching innovative natural anti-cancer components. MATERIALS AND METHODS Twenty steroid glycosides were examined for accordance to their potential of inhibiting cell proliferation and inducing apoptosis in the osteosarcoma cell line 143B. Cell proliferation was examined using a CASY counter. Effects of cardiac glycosides on induction of apoptosis were evaluated by Annexin V-APC and flow cytometry, caspase activity assay and measurement of mitochondrial membrane potential. RESULTS The study revealed that various steroid glycosides suppress cell proliferation in a concentration-dependent manner. Further investigations indicated apoptotic induction by 17 of the 20 tested cardenolides and bufadienolides. Bufadienolide proscillaridin A, arenobufagin, and cardenolides evomonoside, convallatoxol and ouabain waged strongest apoptotic induction, associated with breakdown of mitochondrial membrane potential and activation of caspases -8 and -9. In contrast, the bufadienolide resibufogenin and cardenolide uzarin had no effect on proliferation inhibition, apoptotic induction or change in mitochondrial membrane potential. CONCLUSION These results indicate that bufadienolides proscillaridin A and arenobufagin and cardenolide evomonoside, or related natural compounds might be promising new starting points for development of novel anti-cancer agents for treatment of osteosarcoma.
Collapse
Affiliation(s)
- C I Delebinski
- Department of Paediatric Oncology/Haematology, Otto-Heubner-Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitaetsmedizin Berlin, Berlin, 13353, Germany
| | - S Georgi
- FU Berlin, Institute for Pharmacy, Berlin, 14195, Germany
| | - S Kleinsimon
- Department of Paediatric Oncology/Haematology, Otto-Heubner-Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitaetsmedizin Berlin, Berlin, 13353, Germany
| | - M Twardziok
- Department of Paediatric Oncology/Haematology, Otto-Heubner-Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitaetsmedizin Berlin, Berlin, 13353, Germany
| | - B Kopp
- Department of Pharmacognosy, University of Vienna, Vienna, A-1090, Austria
| | - M F Melzig
- FU Berlin, Institute for Pharmacy, Berlin, 14195, Germany
| | - G Seifert
- Department of Paediatric Oncology/Haematology, Otto-Heubner-Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitaetsmedizin Berlin, Berlin, 13353, Germany
| |
Collapse
|
32
|
Li Y, Yuan J, Yang Q, Cao W, Zhou X, Xie Y, Tu H, Zhang Y, Wang S. Immunoliposome co-delivery of bufalin and anti-CD40 antibody adjuvant induces synergetic therapeutic efficacy against melanoma. Int J Nanomedicine 2014; 9:5683-700. [PMID: 25506218 PMCID: PMC4260685 DOI: 10.2147/ijn.s73651] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Liposomes constitute one of the most popular nanocarriers for improving the delivery and efficacy of agents in cancer patients. The purpose of this study was to design and evaluate immunoliposome co-delivery of bufalin and anti-CD40 to induce synergetic therapeutic efficacy while eliminating systemic side effects. Bufalin liposomes (BFL) conjugated with anti-CD40 antibody (anti-CD40-BFL) showed enhanced cytotoxicity compared with bufalin alone. In a mouse B16 melanoma model, intravenous injection of anti-CD40-BFL achieved smaller tumor volume than did treatment with BFL (average: 117 mm3 versus 270 mm3, respectively); the enhanced therapeutic efficacy through a caspase-dependent pathway induced apoptosis, which was confirmed using terminal deoxynucleotidyl transferase-mediated dUTP-Fluorescein nick end labeling and Western blot assay. Meanwhile, anti-CD40-BFL elicited unapparent body-weight changes and a significant reduction in serum levels of tumor necrosis factor-α, interleukin-1β, interleukin-6, interferon-γ, and hepatic enzyme alanine transaminase, suggesting minimized systemic side effects. This may be attributed to the mechanism by which liposomes are retained within the tumor site for an extended period of time, which is supported by the following biodistribution and flow cytometric analyses. Taken together, the results demonstrated a highly promising strategy for liposomal vehicle transport of anti-CD40 plus bufalin that can be used to enhance antitumor effects via synergetic systemic immunity while blocking systemic toxicity.
Collapse
Affiliation(s)
- Ying Li
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiani Yuan
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Qian Yang
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Wei Cao
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xuanxuan Zhou
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yanhua Xie
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Honghai Tu
- Institute for Drug and Instrument Control, Xinjiang Military Area Command, Urumqi, Xinjiang, People's Republic of China
| | - Ya Zhang
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Siwang Wang
- Department of Natural Medicine and Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
33
|
XU R, XIE HQ, DENG LL, ZHANG JX, YANG FM, LIU JH, HAO XJ, ZHANG YH. A new bufadienolide with cytotoxic activity from the Chinese traditional drug Ch'an Su. Chin J Nat Med 2014; 12:623-7. [DOI: 10.1016/s1875-5364(14)60095-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Indexed: 01/20/2023]
|
34
|
Yiang GT, Chou PL, Hung YT, Chen JN, Chang WJ, Yu YL, Wei CW. Vitamin C enhances anticancer activity in methotrexate‑treated Hep3B hepatocellular carcinoma cells. Oncol Rep 2014; 32:1057-63. [PMID: 24969544 PMCID: PMC4121419 DOI: 10.3892/or.2014.3289] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 04/25/2014] [Indexed: 02/06/2023] Open
Abstract
Methotrexate (MTX) has been widely used for rheumatoid arthritis therapy for a long time. MTX is also used as an anticancer drug for various tumors. However, many studies have shown that high-dose MTX treatment for cancer therapy may cause liver and renal damage. Alhough the mechanisms involved in MTX-induced liver and renal damage require further research, many studies have indicated that MTX-induced cytotoxicity is associated with increases in oxidative stress and caspase activation. In order to reduce MTX-induced side-effects and increase anticancer efficiency, currently, combination treatments of low-dose MTX and other anticancer drugs are considered and applied for various tumor treatments. The present study showed that MTX induces increases in H2O2 levels and caspase-9/-3 activation leading to cell death in hepatocellular carcinoma Hep3B cells. Importantly, this study is the first to demonstrate that vitamin C can efficiently aid low-dose MTX in inducing cell death in Hep3B cells. Therefore, the present study provides a possible powerful therapeutic method for tumors using a combined treatment of vitamin C and low-dose MTX.
Collapse
Affiliation(s)
- Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Pei-Lun Chou
- Division of Allergy-Immunology-Rheumatology, Department of Internal Medicine, Saint Mary's Hospital Luodong, Yilan 265, Taiwan, R.O.C
| | - Yu-Ting Hung
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Shalu, Taichung 433, Taiwan, R.O.C
| | - Jen-Ni Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan, R.O.C
| | - Wei-Jung Chang
- Graduate Institute of Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Yung-Luen Yu
- Graduate Institute of Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Chyou-Wei Wei
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Shalu, Taichung 433, Taiwan, R.O.C
| |
Collapse
|
35
|
Evaluating the cancer therapeutic potential of cardiac glycosides. BIOMED RESEARCH INTERNATIONAL 2014; 2014:794930. [PMID: 24895612 PMCID: PMC4033509 DOI: 10.1155/2014/794930] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 11/23/2022]
Abstract
Cardiac glycosides, also known as cardiotonic steroids, are a group of natural products that share a steroid-like structure with an unsaturated lactone ring and the ability to induce cardiotonic effects mediated by a selective inhibition of the Na+/K+-ATPase. Cardiac glycosides have been used for many years in the treatment of cardiac congestion and some types of cardiac arrhythmias. Recent data suggest that cardiac glycosides may also be useful in the treatment of cancer. These compounds typically inhibit cancer cell proliferation at nanomolar concentrations, and recent high-throughput screenings of drug libraries have therefore identified cardiac glycosides as potent inhibitors of cancer cell growth. Cardiac glycosides can also block tumor growth in rodent models, which further supports the idea that they have potential for cancer therapy. Evidence also suggests, however, that cardiac glycosides may not inhibit cancer cell proliferation selectively and the potent inhibition of tumor growth induced by cardiac glycosides in mice xenografted with human cancer cells is probably an experimental artifact caused by their ability to selectively kill human cells versus rodent cells. This paper reviews such evidence and discusses experimental approaches that could be used to reveal the cancer therapeutic potential of cardiac glycosides in preclinical studies.
Collapse
|
36
|
Abstract
Osteosarcoma (OS) is the most common primary malignant tumor of bone and the third most common cancer in childhood and adolescence. Nowadays, early diagnosis, drug resistance and recurrence of the disease represent the major challenges in OS treatment. Post-genomics, and in particular proteomic technologies, offer an invaluable opportunity to address the level of biological complexity expressed by OS. Although the main goal of OS oncoproteomics is focused on diagnostic and prognostic biomarker discovery, in this review we describe and discuss global protein profiling approaches to other aspects of OS biology and pathophysiology, or to investigate the mechanism of action of chemotherapeutics. In addition, we present proteomic analyses carried out on OS cell lines as in vitro models for studying osteoblastic cell biology and the attractive opportunity offered by proteomics of OS cancer stem cells.
Collapse
Affiliation(s)
- Giulia Bernardini
- Dipartimento di Biotecnologie, Chimica e Farmacia, via Fiorentina 1, Università degli Studi di Siena, 53100 Siena, Italy
| | | | | | | |
Collapse
|
37
|
Anti-inflammatory and antinociceptive activities of bufalin in rodents. Mediators Inflamm 2014; 2014:171839. [PMID: 24719521 PMCID: PMC3955582 DOI: 10.1155/2014/171839] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/10/2014] [Accepted: 01/12/2014] [Indexed: 12/20/2022] Open
Abstract
The aims of this study were to evaluate the anti-inflammatory and analgesic effects of bufalin, a major component of "Chan-su." We used a carrageenan-induced paw edema model to assess the anti-inflammatory activity of this compound, and Western blot analysis detected NF- κ B signaling during this effect. The antinociceptive activities were evaluated by acetic acid-induced writhing, formalin, and hot-plate tests; open-field test investigated effects on the central nervous system. Our data showed that bufalin (0.3 and 0.6 mg/kg, i.p.) potently decreased carrageenan-induced paw edema. Bufalin down regulated the expression levels of nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) during these treatments. Further studies demonstrated that bufalin significantly inhibited the activation of NF-κB signaling. Bufalin also reduced acetic acid-induced writhing and the licking time in the formalin test and increased hot-plate reaction latencies. Naloxone pretreatment (2 mg/kg, i.p.) in the early phases of the formalin test and hot-plate test significantly attenuated the bufalin-induced antinociception effects, which suggests the involvement of the opioid system. A reduction in locomotion was not observed in the open-field test after bufalin administration. Taken together, bufalin treatment resulted in in vivo anti-inflammatory and analgesic effects, and bufalin may be a novel, potential drug for the treatment of inflammatory diseases.
Collapse
|
38
|
Li M, Yu X, Guo H, Sun L, Wang A, Liu Q, Wang X, Li J. Bufalin exerts antitumor effects by inducing cell cycle arrest and triggering apoptosis in pancreatic cancer cells. Tumour Biol 2013. [PMID: 24218335 DOI: 10.1007/s13227-013-1326-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
As one of the most aggressive human malignancies, pancreatic cancer is a leading cause of cancer-related deaths worldwide and only about 4% of patients will live 5 years after diagnosis. Eighty to approximately eighty-five percent of patients are diagnosed with an unresectable or metastatic disease, which is correlated with poor prognosis and low survival rate. Therefore, it is tremendously significant to exploit novel chemicals to prevent and treat pancreatic cancer. Previous research and clinical studies have demonstrated that many natural products derived from traditional Chinese medicine (TCM) such as camptothecin derivatives and vinca alkaloids could be effective antitumor compounds, hinting that TCM is a promising source for developing new antitumor drugs. In this report, we investigated the effects of bufalin, a primary active ingredient of the traditional Chinese medicine Chan-Su, on pancreatic cancer cell lines PANC-1 and CFPAC-1 and studied the underlying molecular mechanism. We found that exposure to bufalin could suppress the proliferation of pancreatic cancer cells time and dose dependently. We used flow cytometry to study the effects of bufalin on apoptosis and cell cycle distribution in PANC-1 and CFPAC-1 cells. The results indicated that bufalin could significantly induce both apoptosis and G2/M cell cycle arrest in pancreatic cancer cells. With western blotting, we found that the expression level of an antiapoptotic protein heat shock protein 27 (Hsp27) and its partner molecule p-Akt was decreased upon the treatment with bufalin. Besides, bufalin activated pro-caspase-3 and pro-caspase-9 and modulated the expression level of Bcl-2 and Bax. These data suggested that bufalin may trigger apoptosis by targeting Hsp27, which could inhibit apoptosis by interfering with key apoptotic proteins. The influence on the level of cylinB1, CDK1, and p21 was also observed after bufalin treatment, and the relationship between Hsp27 and the cell cycle-related proteins mentioned above deserves much more research. In addition, our data showed that bufalin could enhance the growth inhibition effect of gemcitabine in above pancreatic cancer cells. Taken together, bufalin might be worthy of further study for its potential as a therapeutic agent for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Meiying Li
- Department of Medical Oncology, Cancer Center, Qilu Hospital, Shandong University, Jinan, 250012, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bufalin exerts antitumor effects by inducing cell cycle arrest and triggering apoptosis in pancreatic cancer cells. Tumour Biol 2013; 35:2461-71. [PMID: 24218335 DOI: 10.1007/s13277-013-1326-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/14/2013] [Indexed: 12/12/2022] Open
Abstract
As one of the most aggressive human malignancies, pancreatic cancer is a leading cause of cancer-related deaths worldwide and only about 4% of patients will live 5 years after diagnosis. Eighty to approximately eighty-five percent of patients are diagnosed with an unresectable or metastatic disease, which is correlated with poor prognosis and low survival rate. Therefore, it is tremendously significant to exploit novel chemicals to prevent and treat pancreatic cancer. Previous research and clinical studies have demonstrated that many natural products derived from traditional Chinese medicine (TCM) such as camptothecin derivatives and vinca alkaloids could be effective antitumor compounds, hinting that TCM is a promising source for developing new antitumor drugs. In this report, we investigated the effects of bufalin, a primary active ingredient of the traditional Chinese medicine Chan-Su, on pancreatic cancer cell lines PANC-1 and CFPAC-1 and studied the underlying molecular mechanism. We found that exposure to bufalin could suppress the proliferation of pancreatic cancer cells time and dose dependently. We used flow cytometry to study the effects of bufalin on apoptosis and cell cycle distribution in PANC-1 and CFPAC-1 cells. The results indicated that bufalin could significantly induce both apoptosis and G2/M cell cycle arrest in pancreatic cancer cells. With western blotting, we found that the expression level of an antiapoptotic protein heat shock protein 27 (Hsp27) and its partner molecule p-Akt was decreased upon the treatment with bufalin. Besides, bufalin activated pro-caspase-3 and pro-caspase-9 and modulated the expression level of Bcl-2 and Bax. These data suggested that bufalin may trigger apoptosis by targeting Hsp27, which could inhibit apoptosis by interfering with key apoptotic proteins. The influence on the level of cylinB1, CDK1, and p21 was also observed after bufalin treatment, and the relationship between Hsp27 and the cell cycle-related proteins mentioned above deserves much more research. In addition, our data showed that bufalin could enhance the growth inhibition effect of gemcitabine in above pancreatic cancer cells. Taken together, bufalin might be worthy of further study for its potential as a therapeutic agent for pancreatic cancer treatment.
Collapse
|
40
|
Niederberger E, Geisslinger G. Proteomics and NF-κB: an update. Expert Rev Proteomics 2013; 10:189-204. [PMID: 23573785 DOI: 10.1586/epr.13.5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The transcription factor NF-κB was discovered in 1986 and since then has been extensively studied in relation to cancer research and inflammatory or autoimmune diseases due to its important roles in the regulation of apoptosis and inflammation as well as innate and adaptive immunity. Although much is known about NF-κB signaling, novel NF-κB functions in different diseases are still being uncovered, together with its target proteins, interaction partners and regulators of its activation cascade. Proteomic approaches are particularly suited to the discovery of new proteins involved in distinct signal transduction cascades. This review provides an update on and extension of a recent review that summarized a number of proteomic approaches to NF-κB signaling. The studies discussed here utilized innovative techniques and offer several new hypotheses on the role of NF-κB in physiological and pathophysiological processes, which open new avenues for research on NF-κB in the future.
Collapse
Affiliation(s)
- Ellen Niederberger
- Pharmazentrum Frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt am Main, Germany
| | | |
Collapse
|
41
|
Calderón-Montaño JM, Burgos-Morón E, López-Lázaro M. The in vivo antitumor activity of cardiac glycosides in mice xenografted with human cancer cells is probably an experimental artifact. Oncogene 2013; 33:2947-8. [PMID: 23752179 DOI: 10.1038/onc.2013.229] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- J M Calderón-Montaño
- Faculty of Pharmacy, Department of Pharmacology, University of Seville, Seville, Spain
| | - E Burgos-Morón
- Faculty of Pharmacy, Department of Pharmacology, University of Seville, Seville, Spain
| | - M López-Lázaro
- Faculty of Pharmacy, Department of Pharmacology, University of Seville, Seville, Spain
| |
Collapse
|
42
|
Miao Q, Bi LL, Li X, Miao S, Zhang J, Zhang S, Yang Q, Xie YH, Zhang J, Wang SW. Anticancer effects of bufalin on human hepatocellular carcinoma HepG2 cells: roles of apoptosis and autophagy. Int J Mol Sci 2013; 14:1370-82. [PMID: 23344047 PMCID: PMC3565325 DOI: 10.3390/ijms14011370] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 12/25/2012] [Accepted: 12/25/2012] [Indexed: 01/15/2023] Open
Abstract
The traditional Chinese medicine bufalin, extracted from toad’s skin, has been demonstrated to exert anticancer activities in various kinds of human cancers. The mechanisms of action lie in its capacity to induce apoptosis, or termed type I programmed cell death (PCD). However, type II PCD, or autophagy, participates in cancer proliferation, progression, and relapse, as well. Recent studies on autophagy seem to be controversial because of the dual roles of autophagy in cancer survival and death. In good agreement with previous studies, we found that 100 nM bufalin induced extensive HepG2 cell apoptosis. However, we also noticed bufalin triggered autophagy and enhanced Beclin-1 expression, LC3-I to LC3-II conversion, as well as decreased p62 expression and mTOR signaling activation in HepG2 cells. Blockage of autophagy by selective inhibitor 3-MA decreased apoptotic ratio in bufalin-treated HepG2 cells, suggesting a proapoptotic role of bufalin-induced autophagy. Furthermore, we investigated the underlying mechanisms of bufalin-induced autophagy. Bufalin treatment dose-dependently promoted AMPK phosphorylation while AMPK inhibition by compound C significantly attenuated bufalin-induced autophagy. Taken together, we report for the first time that bufalin induces HepG2 cells PCD, especially for autophagy, and the mechanism of action is, at least in part, AMPK-mTOR dependent.
Collapse
Affiliation(s)
- Qing Miao
- Institute of Materia Medica, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Q.M.); (L.-L.B.); (S.M.); (Q.Y.); (Y.-H.X.)
| | - Lin-Lin Bi
- Institute of Materia Medica, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Q.M.); (L.-L.B.); (S.M.); (Q.Y.); (Y.-H.X.)
| | - Xin Li
- Department of Pharmacy, Chinese PLA General Hospital, Beijing 100850, China; E-Mail:
| | - Shan Miao
- Institute of Materia Medica, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Q.M.); (L.-L.B.); (S.M.); (Q.Y.); (Y.-H.X.)
| | - Jin Zhang
- Department of Hand Surgery, 401 Military Hospital, Qingdao 266071, China; E-Mail:
| | - Song Zhang
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an 710032, China; E-Mail:
| | - Qian Yang
- Institute of Materia Medica, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Q.M.); (L.-L.B.); (S.M.); (Q.Y.); (Y.-H.X.)
| | - Yan-Hua Xie
- Institute of Materia Medica, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Q.M.); (L.-L.B.); (S.M.); (Q.Y.); (Y.-H.X.)
| | - Jian Zhang
- Department of Pulmonary Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China
- Authors to whom correspondence should be addressed; E-Mails: (S.-W.W.); (J.Z.); Tel.: +86-29-8477-4748 (S.-W.W.); +86-29-8477-1132 (J.Z.); Fax: +86-29-8322-4790 (S.-W.W.); +86-29-8477-1132 (J.Z.)
| | - Si-Wang Wang
- Institute of Materia Medica, Fourth Military Medical University, Xi’an 710032, China; E-Mails: (Q.M.); (L.-L.B.); (S.M.); (Q.Y.); (Y.-H.X.)
- Authors to whom correspondence should be addressed; E-Mails: (S.-W.W.); (J.Z.); Tel.: +86-29-8477-4748 (S.-W.W.); +86-29-8477-1132 (J.Z.); Fax: +86-29-8322-4790 (S.-W.W.); +86-29-8477-1132 (J.Z.)
| |
Collapse
|