1
|
Hao Z, Zhou Y, Zhang Y, Wang D, Wei Y, Ji X, Sun WR, Wang P, Li Y, Lopez IB, Pedraz JL, Ramalingam M, Xie S, Wang R. Celastrol loaded nanocomplex for painless tumor therapy via YAP inhibition. Sci Rep 2025; 15:13133. [PMID: 40240779 PMCID: PMC12003811 DOI: 10.1038/s41598-025-97055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer-related pain is prevalent and severely impairs patients' quality of life. However, conventional cancer therapies primarily target tumor cell destruction, often overlooking the management of cancer pain. Thus, there is an immediate necessity to develop therapeutic agents that can both suppress tumor growth and alleviate cancer pain. In this study, we report a celastrol (CEL)-based nanocomposites (PDA-BSA-MnO2-CEL) for pain-less cancer immunotherapy. Results from in vitro and in vivo experiments demonstrate the efficacy and mechanism of the nanocomposites in pain-less immunotherapy. MnO2 and CEL induce immunogenic cell death (ICD), mediating immunotherapy. Additionally, CEL significantly reduces the secretion of the immunosuppressive factor Yes-associated protein (YAP) within the tumor microenvironment, thereby enhancing the efficacy of immunotherapy. The downregulation of YAP leads to reduced expression of vascular endothelial growth factor (VEGF), inhibiting tumor growth and decreasing activation of the pain-associated VEGF receptor 1 (VEGFR1), thus providing an analgesic effect. Moreover, CEL reduces inflammatory pain by lowering levels of inflammatory factors in tumors. The design of this nanocomposites system integrates immunotherapy with cancer pain inhibition, offering a novel approach to patient-centered tumor therapy.
Collapse
Affiliation(s)
- Zhaokun Hao
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai, 264000, People's Republic of China
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Yuming Zhou
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, People's Republic of China
| | - Yuqiang Zhang
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, People's Republic of China
| | - Danyang Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Yiying Wei
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Xiaopu Ji
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Wan Ru Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Pingyu Wang
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - YouJie Li
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Irene Bautista Lopez
- NanoBioCel Group, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/ EHU), 01006, Vitoria-Gasteiz, Spain
| | - José Luis Pedraz
- NanoBioCel Group, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/ EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba Health Research Institute, Jose Atxotegi, s/n, 01009, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain
- Joint Research Laboratory (JRL) on Bioprinting and Advanced Pharma Development, Centro de investigació n Lascaray Ikergunea, A Joined Venture of TECNALIA, Basque Research & Technology Alliance (BRTA), Avenida Miguel de Unamuno, 01006, Vitoria-Gasteiz, Spain
| | - Murugan Ramalingam
- NanoBioCel Group, Department of Pharmacy and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/ EHU), 01006, Vitoria-Gasteiz, Spain.
- Bioaraba Health Research Institute, Jose Atxotegi, s/n, 01009, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III (ISCIII), 28029, Madrid, Spain.
- Joint Research Laboratory (JRL) on Bioprinting and Advanced Pharma Development, Centro de investigació n Lascaray Ikergunea, A Joined Venture of TECNALIA, Basque Research & Technology Alliance (BRTA), Avenida Miguel de Unamuno, 01006, Vitoria-Gasteiz, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013, Bilbao, Spain.
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| | - Shuyang Xie
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai, 264000, People's Republic of China.
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| | - Ranran Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, 264003, People's Republic of China.
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai, 264000, People's Republic of China.
| |
Collapse
|
2
|
Su D, Wei RY, Yan ZM, Zhong GH, Qin XQ, Huang ST, Long JY, Zhang FL, He P, Chen ZJ, Yan YQ, Jiang N, Tang WZ. Design, synthesis, and evaluation of antitumor activity of novel C-6 sulfhydryl-substituted and 20-substituted derivatives of celastrol. Chem Biol Drug Des 2023; 102:316-331. [PMID: 37156601 DOI: 10.1111/cbdd.14247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/01/2023] [Accepted: 04/04/2023] [Indexed: 05/10/2023]
Abstract
Celastrol has been identified as a potential candidate for anticancer drug development. In this study, 28 novel celastrol derivatives with C-6 sulfhydryl substitution and 20-substitution were designed and synthesized, and their antiproliferative activity against human cancer cells and non-malignant human cells was evaluated, with cisplatin and celastrol being used as controls. The results showed that most of the derivatives had enhanced in vitro anticancer activity compared to the parent compound celastrol. Specifically, derivative 2f demonstrated the most potent inhibitory potential and selectivity against HOS with an IC50 value of 0.82 μM. Our study provides new insights into the structure-activity relationship of celastrol and suggests that compound 2f may be a promising drug candidate for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Di Su
- Department of Gastrointestinal Surgery, Guangxi Clinical Research Center for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, P.R. China
| | - Rong-Yuan Wei
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Zhi-Ming Yan
- College of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Guo-Hui Zhong
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, P.R. China
| | - Xiang-Qing Qin
- Department of Gastrointestinal Surgery, Guangxi Clinical Research Center for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, P.R. China
| | - Shu-Tong Huang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, P.R. China
| | - Juan-Yue Long
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, P.R. China
| | - Feng-Ling Zhang
- College of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Ping He
- College of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Zhong-Ji Chen
- College of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Ya-Qian Yan
- College of Pharmacy, Guangxi Medical University, Nanning, P.R. China
| | - Neng Jiang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, P.R. China
| | - Wei-Zhong Tang
- Department of Gastrointestinal Surgery, Guangxi Clinical Research Center for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, P.R. China
| |
Collapse
|
3
|
Zhang X, Zhou J, Zhu Y, Wong YK, Liu D, Gao P, Lin Q, Zhang J, Chen X, Wang J. Quantitative chemical proteomics reveals anti-cancer targets of Celastrol in HCT116 human colon cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154096. [PMID: 35452923 DOI: 10.1016/j.phymed.2022.154096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Celastrol (Cel) is a naturally-derived compound with anti-cancer properties and exerts beneficial effects against various diseases. Although an extensive body of research already exists for Cel, the vast majority are inductive studies with limited validation of specific pathways and functions. The cellular targets that bind to Cel remain poorly characterized, which limits attempts to uncover its mechanism of action. PURPOSE The present study aims to comprehensively identify the protein targets of Cel in HCT116 cells in an unbiased manner, and elucidate the mechanism of the anti-cancer activity of Cel based on target information. METHODS A comprehensive analysis of protein targets that bind to Cel was performed in HCT116 colon cancer cells using a quantitative chemical biology method. A Cel probe (Cel-P) was synthesized to allow in situ monitoring of treatment in living HCT116 cells, and specific targets were identified with a quantitative chemical biology method (isobaric tags for relative and absolute quantitation) using mass spectrometry. RESULTS In total, 100 protein targets were identified as specific targets of Cel. Pathways associated with the targets were investigated. Multiple pathways were demonstrated to be potential effectors of Cel. These pathways included the suppression of protein synthesis, deregulation of cellular reactive oxygen species, and suppression of fatty acid metabolism, and they were validated with in vitro experiments. CONCLUSION The extensive information on the protein targets of Cel and their functions uncovered by this study will enhance the current understanding of the mechanism of action of Cel and serve as a valuable knowledge base for future studies.
Collapse
Affiliation(s)
- Xing Zhang
- Institute of Chinese Materia Medica and Artemisinin Research Center, Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jing Zhou
- Department of physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530022, China; Department of Epidemiology, School of Public Health, Guangxi Medical University, Nanning 530022, China
| | - Yongping Zhu
- Institute of Chinese Materia Medica and Artemisinin Research Center, Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yin Kwan Wong
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore; Department of Urology, the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, China
| | - Dandan Liu
- Institute of Chinese Materia Medica and Artemisinin Research Center, Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Peng Gao
- Institute of Chinese Materia Medica and Artemisinin Research Center, Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Jianbin Zhang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China.
| | - Xiao Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Jigang Wang
- Institute of Chinese Materia Medica and Artemisinin Research Center, Academy of Chinese Medical Sciences, Beijing 100700, China; Department of physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning 530022, China; Department of Epidemiology, School of Public Health, Guangxi Medical University, Nanning 530022, China; Department of Urology, the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, China; Center for Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Southern Medical University, Dongguan 523125, China; Central People's Hospital of Zhanjiang, Zhanjiang 524037, China; Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
4
|
Lei ZC, Li N, Yu NR, Ju W, Sun XN, Zhang XL, Dong HJ, Sun JB, Chen L. Design and Synthesis of Novel Celastrol Derivatives as Potential Anticancer Agents against Gastric Cancer Cells. JOURNAL OF NATURAL PRODUCTS 2022; 85:1282-1293. [PMID: 35536757 DOI: 10.1021/acs.jnatprod.1c01236] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Gastric cancer (GC) is a common malignant disease worldwide, and finding novel agents and strategies for the treatment of GC are of urgent need. Celastrol (CEL) is a well-known natural product with antineoplastic activity. In this study, pyrazole analogues were introduced at the C-29 position of CEL. A total of 24 new derivatives were designed, synthesized, and evaluated for their mechanism and antitumor activity in vitro and in vivo. Among them, compound 21 exhibited the best activity against BGC-823 cells (IC50 = 0.21 ± 0.01 μM). Further biological studies showed that 21 significantly raised the reactive oxygen species (ROS) levels to activate the apoptotic pathway, causing mitochondrial dysfunction in BGC-823 cells. In addition, 21 also arrested cells in the G2/M phase to induce tumor cell apoptosis. In a nude mouse tumor xenograft model, 21 exhibited a better tumor inhibition rate (89.85%) than CEL (inhibition rate 76.52%). Taken together, the present study has provided an anticancer lead compound candidate, 21, and has revealed that increased ROS generation may be an effective strategy in the treatment of GC.
Collapse
Affiliation(s)
- Zhi-Chao Lei
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Na Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science & Technology, 26 Yuxiang Street, Hebei 050018, People's Republic of China
| | - Nai-Rong Yu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Wei Ju
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Xiao-Na Sun
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Xue-Ling Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Hai-Juan Dong
- The Public Laboratory Platform, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Jian-Bo Sun
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
| | - Li Chen
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, People's Republic of China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, 132 Lanhei Road, Kunming 650201, Yunnan, People's Republic of China
| |
Collapse
|
5
|
Youns M, Askoura M, Abbas HA, Attia GH, Khayyat AN, Goda RM, Almalki AJ, Khafagy ES, Hegazy WAH. Celastrol Modulates Multiple Signaling Pathways to Inhibit Proliferation of Pancreatic Cancer via DDIT3 and ATF3 Up-Regulation and RRM2 and MCM4 Down-Regulation. Onco Targets Ther 2021; 14:3849-3860. [PMID: 34194230 PMCID: PMC8238076 DOI: 10.2147/ott.s313933] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/13/2021] [Indexed: 12/17/2022] Open
Abstract
Background Pancreatic cancer is one of the most serious and lethal human cancers with a snowballing incidence around the world. The natural product celastrol has also been widely documented as a potent anti-inflammatory, anti-angiogenic, and anti-oxidant. Purpose To elucidate the antitumor effect of celastrol on pancreatic cancer cells and its modulatory role on whole genome expression. Methods The antitumor activity of celastrol on a panel of pancreatic cancer cells has been evaluated by Sulforhodamine B assay. Caspase 3/7 and histone-associated DNA fragments assays were done for apoptosis measurement. Additionally, prostaglandin (PGE2) inhibition was evaluated. Moreover, a microarray gene expression profiling was carried out to detect possible key players that modulate the antitumor effects of celastrol on cells of pancreatic cancer. Results Our findings indicated that celastrol suppresses the cellular growth of pancreatic cancer cells, induces apoptosis, and inhibits PGE2 production. Celastrol modulated many signaling genes and its cytotoxic effect was mainly mediated via over-expression of ATF3 and DDIT3, and down-expression of RRM2 and MCM4. Conclusion The current study aims to be a starting point to generate a hypothesis on the most significant regulatory genes and for a full dissection of the celastrol possible effects on each single gene to prevent the pancreatic cancer growth.
Collapse
Affiliation(s)
- Mahmoud Youns
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo, Egypt.,Department of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
| | - Momen Askoura
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Hisham A Abbas
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Gouda H Attia
- Department of Pharmacognosy, Faculty of Pharmacy, Kafr El-Shiekh University, Kafr El-Shiekh, Egypt.,Department of Pharmacognosy, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Ahdab N Khayyat
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Reham M Goda
- Department of Microbiology and Biotechnology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Mansoura, Egypt
| | - Ahmad J Almalki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj, 11942, Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia, 41552, Egypt
| | - Wael A H Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
6
|
Lactoferrin-dual drug nanoconjugate: Synergistic anti-tumor efficacy of docetaxel and the NF-κB inhibitor celastrol. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111422. [PMID: 33255023 DOI: 10.1016/j.msec.2020.111422] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/27/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Despite the progress in cancer nanotherapeutics, some obstacles still impede the success of nanocarriers and hinder their clinical translation. Low drug loading, premature drug release, off-target toxicity and multi-drug resistance are among the most difficult challenges. Lactoferrin (LF) has demonstrated a great tumor targeting capacity via its high binding affinity to low density lipoprotein (LDL) and transferrin (Tf) receptors overexpressed by various cancer cells. Herein, docetaxel (DTX) and celastrol (CST) could be successfully conjugated to LF backbone for synergistic breast cancer therapy. Most importantly, the conjugate self-assembled forming nanoparticles of 157.8 nm with elevated loading for both drugs (6.94 and 5.98% for DTX and CST, respectively) without risk of nanocarrier instability. Moreover, the nanoconjugate demonstrated enhanced in vivo anti-tumor efficacy in breast cancer-bearing mice, as reflected by a reduction in tumor volume, prolonged survival rate and significant suppression of NF-κB p65, TNF-α, COX-2 and Ki-67 expression levels compared to the group given free combined DTX/CST therapy and to positive control. This study demonstrated the proof-of-principle for dual drug coupling to LF as a versatile nanoplatform that could augment their synergistic anticancer efficacy.
Collapse
|
7
|
Hou W, Liu B, Xu H. Celastrol: Progresses in structure-modifications, structure-activity relationships, pharmacology and toxicology. Eur J Med Chem 2020; 189:112081. [DOI: 10.1016/j.ejmech.2020.112081] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 12/13/2022]
|
8
|
Niemelä E, Desai D, Lundsten E, Rosenholm JM, Kankaanpää P, Eriksson JE. Quantitative bioimage analytics enables measurement of targeted cellular stress response induced by celastrol-loaded nanoparticles. Cell Stress Chaperones 2019; 24:735-748. [PMID: 31079284 PMCID: PMC6629742 DOI: 10.1007/s12192-019-00999-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 04/12/2019] [Accepted: 04/17/2019] [Indexed: 10/26/2022] Open
Abstract
The cellular stress response, which provides protection against proteotoxic stresses, is characterized by the activation of heat shock factor 1 and the formation of nuclear stress bodies (nSBs). In this study, we developed a computerized method to quantify the formation and size distribution of nSBs, as stress response induction is of interest in cancer research, neurodegenerative diseases, and in other pathophysiological processes. We employed an advanced bioimaging and analytics workflow to enable quantitative detailed subcellular analysis of cell populations even down to single-cell level. This type of detailed analysis requires automated single cell analysis to allow for detection of both size and distribution of nSBs. For specific induction of nSB we used mesoporous silica nanoparticles (MSNs) loaded with celastrol, a plant-derived triterpene with the ability to activate the stress response. To enable specific targeting, we employed folic acid functionalized nanoparticles, which yields targeting to folate receptor expressing cancer cells. In this way, we could assess the ability to quantitatively detect directed and spatio-temporal nSB induction using 2D and 3D confocal imaging. Our results demonstrate successful implementation of an imaging and analytics workflow based on a freely available, general-purpose software platform, BioImageXD, also compatible with other imaging modalities due to full 3D/4D and high-throughput batch processing support. The developed quantitative imaging analytics workflow opens possibilities for detailed stress response examination in cell populations, with significant potential in the analysis of targeted drug delivery systems related to cell stress and other cytoprotective cellular processes.
Collapse
Affiliation(s)
- Erik Niemelä
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Diti Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Emine Lundsten
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Pasi Kankaanpää
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - John E. Eriksson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| |
Collapse
|
9
|
Abstract
Celastrol is a highly investigated anticancer moiety. It is a pentacyclic triterpenoid, isolated several decades ago with promising role in chemoprevention. Celastrol has been found to target multiple proinflammatory, angiogenic and metastatic proteins. Inhibition of these targets results in significant reduction of cancer growth, survival and metastasis. This review summarizes the varied molecular targets of celastrol along with insight into the various recently published clinical, preclinical and industrial patents (2011-2017).
Collapse
|
10
|
Maysinger D, Moquin A, Choi J, Kodiha M, Stochaj U. Gold nanourchins and celastrol reorganize the nucleo- and cytoskeleton of glioblastoma cells. NANOSCALE 2018; 10:1716-1726. [PMID: 29308473 DOI: 10.1039/c7nr07833a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The physicochemical properties and cytotoxicity of diverse gold nanoparticle (AuNP) morphologies with smooth surfaces have been examined extensively. Much less is known about AuNPs with irregular surfaces. This study focuses on the effects of gold nanourchins in glioblastoma cells. With limited success of monotherapies for glioblastoma, multimodal treatment has become the preferred regimen. One possible example for such future therapeutic applications is the combination of AuNPs with the natural cytotoxic agent celastrol. Here, we used complementary physical, chemical and biological methods to characterize AuNPs and investigate their impact on glioblastoma cells. Our results show that gold nanourchins altered glioblastoma cell morphology and reorganized the nucleo- and cytoskeleton. These changes were dependent on gold nanourchin surface modification. PEGylated nanourchins had no significant effect on glioblastoma cell morphology or viability, unless they were combined with celastrol. By contrast, CTAB-nanourchins adversely affected the nuclear lamina, microtubules and filamentous actin. These alterations correlated with significant glioblastoma cell death. We identified several mechanisms that contributed to the impact of AuNPs on the cytoskeleton and cell survival. Specifically, CTAB-nanourchins caused a significant increase in the abundance of Rock1. This protein kinase is a key regulator of the cytoskeleton. In addition, CTAB-nanourchins led to a marked decline in pro-survival signaling via the PI3 kinase-Akt pathway. Taken together, our study provides new insights into the molecular pathways and structural components altered by gold nanourchins and their implications for multimodal glioblastoma therapy.
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.
| | | | | | | | | |
Collapse
|
11
|
Das J, Sarkar A, Ghosh P. Friedelane triterpenoids: transformations toward A-ring modifications including 2-homoderivatives. NEW J CHEM 2018. [DOI: 10.1039/c8nj00009c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Common reaction strategies were employed on suitable substrates to achieve a series of C2,C3-; C3,C4- and C2,C3,C4-functionalized (including 2-homo-) friedelane triterpenoids with just one to four efficient steps.
Collapse
Affiliation(s)
- Jayanta Das
- Natural Products and Polymer Chemistry Laboratory
- Department of Chemistry
- North Bengal University
- Darjeeling-734013
- India
| | - Antara Sarkar
- Natural Products and Polymer Chemistry Laboratory
- Department of Chemistry
- North Bengal University
- Darjeeling-734013
- India
| | - Pranab Ghosh
- Natural Products and Polymer Chemistry Laboratory
- Department of Chemistry
- North Bengal University
- Darjeeling-734013
- India
| |
Collapse
|
12
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. Cytotoxic effect of celastrol alone or in combination with paclitaxel on anaplastic thyroid carcinoma cells. Tumour Biol 2017; 39:1010428317698369. [DOI: 10.1177/1010428317698369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The influence of celastrol alone or in combination with paclitaxel on survival of anaplastic thyroid carcinoma cells was investigated. In 8505C and SW1736 cells, after treatment of celastrol, cell viability decreased, and cytotoxic activity increased. The protein levels of heat shock protein (hsp) 90, hsp70, Bax, death receptor 5, cleaved caspase-3, cleaved poly (ADP-ribose) polymerase, phospho-extracellular signal-regulated kinase 1/2 (ERK1/2), and phospho-c-Jun N-terminal kinase (JNK) were elevated, and those of Bcl2, phospho-nuclear factor-kappaB (NF-κB), and total and phospho-Akt were reduced. The endoplasmic reticulum stress markers expression and reactive oxygen species production were enhanced. In celastrol-treated cells, N-acetylcysteine increased cell viability and phospho-NF-κB protein levels, and decreased reactive oxygen species production and cytotoxic activity. The protein levels of cyclooxygenase 2, phospho-ERK1/2, phospho-JNK and Bip were diminished. After treatment of both celastrol and paclitaxel, compared with paclitaxel alone, cell viability and the percentage of viable cells were reduced, and death rate and cytotoxic activity were elevated. The protein levels of phospho-ERK1/2, phospho-JNK, Bip, and cyclooxygenase 2, and reactive oxygen species production were enhanced. All of the Combination Index values calculated by Chou–Talalay equation were lower than 1.0, implying the synergism between celastrol and paclitaxel in induction of cell death. In conclusion, our results suggest that celastrol induces cytotoxicity through involvement of Bcl2 family proteins and death receptor, and modulation of phospho-NF-κB, Akt, and mitogen-activated protein kinase in association with endoplasmic reticulum stress and reactive oxygen species production in anaplastic thyroid carcinoma cells. Moreover, celastrol synergizes with paclitaxel in induction of cytotoxicity in anaplastic thyroid carcinoma cells.
Collapse
Affiliation(s)
- Si Hyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jun Goo Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Chul Sik Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Sung-Hee Ihm
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Moon Gi Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Hyung Joon Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Seong Jin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
13
|
Zhu L, Chen Y, Wei C, Yang X, Cheng J, Yang Z, Chen C, Ji Z. Anti-proliferative and pro-apoptotic effects of cinobufagin on human breast cancer MCF-7 cells and its molecular mechanism. Nat Prod Res 2017; 32:493-497. [PMID: 28412840 DOI: 10.1080/14786419.2017.1315575] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Ling Zhu
- Department of Oncology, Wannan Medical College, Wuhu, P. R. China
| | - Yuxin Chen
- Department of Oncology, Wannan Medical College, Wuhu, P. R. China
| | - Cheng Wei
- Department of Oncology, Wannan Medical College, Wuhu, P. R. China
| | - Xi Yang
- Department of Oncology, Wannan Medical College, Wuhu, P. R. China
| | - Jing Cheng
- The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Zhimin Yang
- The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| | - Chuanrong Chen
- Department of Oncology, Wuhu NO.2 People’s Hospital, Wuhu, P. R. China
| | - Zhaoning Ji
- The Cancer Center, Yijishan Hospital of Wannan Medical College, Wuhu, P. R. China
| |
Collapse
|
14
|
Hernandes C, Pereira AMS, Severino P. Compounds From Celastraceae Targeting Cancer Pathways and Their Potential Application in Head and Neck Squamous Cell Carcinoma: A Review. Curr Genomics 2016; 18:60-74. [PMID: 28503090 PMCID: PMC5321769 DOI: 10.2174/1389202917666160803160934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/28/2015] [Accepted: 11/29/2015] [Indexed: 12/13/2022] Open
Abstract
Squamous cell carcinoma of the head and neck is one of the most common cancer types worldwide. It initiates on the epithelial lining of the upper aerodigestive tract, at most instances as a consequence of tobacco and alcohol consumption. Treatment options based on conventional therapies or targeted therapies under development have limited efficacy due to multiple genetic alterations typically found in this cancer type. Natural products derived from plants often possess biological activities that may be valuable in the development of new therapeutic agents for cancer treatment. Several genera from the family Celastraceae have been studied in this context. This review reports studies on chemical constituents isolated from species from the Celastraceae family targeting cancer mechanisms studied to date. These results are then correlated with molecular characteristics of head and neck squamous cell carcinoma in an attempt to identify constituents with potential application in the treatment of this complex disease at the molecular level.
Collapse
Affiliation(s)
- Camila Hernandes
- aAlbert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo, Brazil; bDepartment of Biotechnology, Universidade de Ribeirão Preto, Ribeirão Preto, Brazil
| | - Ana Maria Soares Pereira
- aAlbert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo, Brazil; bDepartment of Biotechnology, Universidade de Ribeirão Preto, Ribeirão Preto, Brazil
| | - Patricia Severino
- aAlbert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, Sao Paulo, Brazil; bDepartment of Biotechnology, Universidade de Ribeirão Preto, Ribeirão Preto, Brazil
| |
Collapse
|
15
|
Zhang Y, Xu YY, Sun WJ, Zhang MH, Zheng YF, Shen HM, Yang J, Zhu XQ. FBS or BSA Inhibits EGCG Induced Cell Death through Covalent Binding and the Reduction of Intracellular ROS Production. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5013409. [PMID: 27830147 PMCID: PMC5088332 DOI: 10.1155/2016/5013409] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/05/2016] [Accepted: 08/24/2016] [Indexed: 02/04/2023]
Abstract
Previously we have shown that (-)-epigallocatechin gallate (EGCG) can induce nonapoptotic cell death in human hepatoma HepG2 cells only under serum-free condition. However, the underlying mechanism for serum in determining the cell fate remains to be answered. The effects of fetal bovine serum (FBS) and its major component bovine serum albumin (BSA) on EGCG-induced cell death were investigated in this study. It was found that BSA, just like FBS, can protect cells from EGCG-induced cell death in a dose-dependent manner. Detailed analysis revealed that both FBS and BSA inhibited generation of ROS to protect against toxicity of EGCG. Furthermore, EGCG was shown to bind to certain cellular proteins including caspase-3, PARP, and α-tubulin, but not LC3 nor β-actin, which formed EGCG-protein complexes that were inseparable by SDS-gel. On the other hand, addition of FBS or BSA to culture medium can block the binding of EGCG to these proteins. In silico docking analysis results suggested that BSA had a stronger affinity to EGCG than the other proteins. Taken together, these data indicated that the protective effect of FBS and BSA against EGCG-induced cell death could be due to (1) the decreased generation of ROS and (2) the competitive binding of BSA to EGCG.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, Zhejiang 310058, China
| | - Yu-Ying Xu
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, Zhejiang 310058, China
| | - Wen-Jie Sun
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, Zhejiang 310058, China
| | - Mo-Han Zhang
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, Zhejiang 310058, China
| | - Yi-Fan Zheng
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, Zhejiang 310058, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Jun Yang
- Department of Toxicology, School of Public Health, Hangzhou Normal University, 16 Xue Lin Street, Hangzhou, Zhejiang 310036, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, National Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xin-Qiang Zhu
- Department of Toxicology, Zhejiang University School of Public Health, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
16
|
Lange I, Moschny J, Tamanyan K, Khutsishvili M, Atha D, Borris RP, Koomoa DL. Scrophularia orientalis extract induces calcium signaling and apoptosis in neuroblastoma cells. Int J Oncol 2016; 48:1608-16. [PMID: 26848085 PMCID: PMC4777595 DOI: 10.3892/ijo.2016.3373] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/15/2015] [Indexed: 12/26/2022] Open
Abstract
Effective neuroblastoma (NB) treatments are still limited despite treatment options available today. Therefore, this study attempted to identify novel plant extracts that have anticancer effects. Cytotoxicity and increased intracellular calcium levels were determined using the Sulforhodamine B (SRB) assay and Fluo4-AM (acetoxymethyl) staining and fluorescence microscopy in NB cells in order to screen a library of plant extracts. The current study examined the anticancer effects of a dichloromethane extract from Scrophularia orientalis L. (Scrophulariaceae), a plant that has been used in Traditional Chinese Medicine. This extract contained highly potent agents that significantly reduced cell survival and increased calcium levels in NB cells. Further analysis revealed that cell death induced by this extract was associated with intracellular calcium release, opening of the MPTP, caspase 3- and PARP-cleavage suggesting that this extract induced aberrant calcium signaling that resulted in apoptosis via the mitochondrial pathway. Therefore, agents from Scrophularia orientalis may have the potential to lead to new chemo therapeutic anticancer drugs. Furthermore, targeting intracellular calcium signaling may be a novel strategy to develop more effective treatments for NB.
Collapse
Affiliation(s)
- Ingo Lange
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| | - Julia Moschny
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| | - Kamilla Tamanyan
- Institute of Botany, National Academy of Sciences, Yerevan, Armenia
| | - Manana Khutsishvili
- National Herbarium of Georgia, Institute of Botany, Ilia State University, Tbilisi, Georgia
| | - Daniel Atha
- The New York Botanical Garden, Bronx, NY, USA
| | - Robert P Borris
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Dana-Lynn Koomoa
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI, USA
| |
Collapse
|
17
|
Kibble M, Saarinen N, Tang J, Wennerberg K, Mäkelä S, Aittokallio T. Network pharmacology applications to map the unexplored target space and therapeutic potential of natural products. Nat Prod Rep 2015; 32:1249-66. [PMID: 26030402 DOI: 10.1039/c5np00005j] [Citation(s) in RCA: 310] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is widely accepted that drug discovery often requires a systems-level polypharmacology approach to tackle problems such as lack of efficacy and emerging resistance of single-targeted compounds. Network pharmacology approaches are increasingly being developed and applied to find new therapeutic opportunities and to re-purpose approved drugs. However, these recent advances have been relatively slow to be translated into the field of natural products. Here, we argue that a network pharmacology approach would enable an effective mapping of the yet unexplored target space of natural products, hence providing a systematic means to extend the druggable space of proteins implicated in various complex diseases. We give an overview of the key network pharmacology concepts and recent experimental-computational approaches that have been successfully applied to natural product research, including unbiased elucidation of mechanisms of action as well as systematic prediction of effective therapeutic combinations. We focus specifically on anticancer applications that use in vivo and in vitro functional phenotypic measurements, such as genome-wide transcriptomic response profiles, which enable a global modelling of the multi-target activity at the level of the biological pathways and interaction networks. We also provide representative examples of other disease applications, databases and tools as well as existing and emerging resources, which may prove useful for future natural product research. Finally, we offer our personal view of the current limitations, prospective developments and open questions in this exciting field.
Collapse
Affiliation(s)
- Milla Kibble
- Institute for Molecular Medicine Finland (FIMM), Biomedicum Helsinki 2U, 00014 University of Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
18
|
Sugar-decorated mesoporous silica nanoparticles as delivery vehicles for the poorly soluble drug celastrol enables targeted induction of apoptosis in cancer cells. Eur J Pharm Biopharm 2015; 96:11-21. [DOI: 10.1016/j.ejpb.2015.07.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 11/22/2022]
|
19
|
Celastrol stimulates hypoxia-inducible factor-1 activity in tumor cells by initiating the ROS/Akt/p70S6K signaling pathway and enhancing hypoxia-inducible factor-1α protein synthesis. PLoS One 2014; 9:e112470. [PMID: 25383959 PMCID: PMC4226555 DOI: 10.1371/journal.pone.0112470] [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: 04/10/2014] [Accepted: 10/06/2014] [Indexed: 11/19/2022] Open
Abstract
Celastrol, a tripterine derived from the traditional Chinese medicine plant Tripterygium wilfordii Hook F. (“Thunder of God Vine”), has been reported to have multiple effects, such as anti-inflammation, suppression of tumor angiogenesis, inhibition of tumor growth, induction of apoptosis and protection of cells against human neurodegenerative diseases. However, the mechanisms that underlie these functions are not well defined. In this study, we reported for the first time that Celastrol could induce HIF-1α protein accumulation in multiple cancer cell lines in an oxygen-independent manner and that the enhanced HIF-1α protein entered the nucleus and promoted the transcription of the HIF-1 target genes VEGF and Glut-1. Celastrol did not influence HIF-1α transcription. Instead, Celastrol induced the accumulation of the HIF-1α protein by inducing ROS and activating Akt/p70S6K signaling to promote HIF-1α translation. In addition, we found that the activation of Akt by Celastrol was transient. With increased exposure time, inhibition of Hsp90 chaperone function by Celastrol led to the subsequent depletion of the Akt protein and thus to the suppression of Akt activity. Moreover, in HepG2 cells, the accumulation of HIF-1α increased the expression of BNIP3, which induced autophagy. However, HIF-1α and BNIP3 did not influence the cytotoxicity of Celastrol because the main mechanism by which Celastrol kills cancer cells is through stimulating ROS-mediated JNK activation and inducing apoptosis. Furthermore, our data showed that the dose required for Celastrol to induce HIF-1α protein accumulation and enhance HIF-1α transcriptional activation was below its cytotoxic threshold. A cytotoxic dose of Celastrol for cancer cells did not display cytotoxicity in LO2 normal human liver cells, which indicated that the novel functions of Celastrol in regulating HIF-1 signaling and inducing autophagy might be used in new applications, such as in anti-inflammation and protection of cells against human neurodegenerative diseases. Future studies regarding these applications are required.
Collapse
|
20
|
Jo H, Loison F, Luo HR. Microtubule dynamics regulates Akt signaling via dynactin p150. Cell Signal 2014; 26:1707-16. [PMID: 24726838 DOI: 10.1016/j.cellsig.2014.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/06/2014] [Indexed: 01/22/2023]
Abstract
Following activation at the plasma membrane, Akt is subsequently deactivated in the cytoplasm. Although activation and deactivation of Akt must sometimes be separated in order to elicit and control cellular responses, the exact details of the spatiotemporal organization of Akt signaling are incompletely understood. Here we show that microtubule dynamics specifically modulate the deactivation phase of Akt signaling. Localization of Akt to microtubules sustains its activity, while disruption of microtubules attenuates Akt signaling independent of its initial activation. Conversely, stabilization of microtubules elevates Akt signaling both in vitro and in muscle tissues in vivo. Localization of Akt to microtubules is mediated by the microtubule binding protein dynactin p150, which is shown to be a direct target of Akt. Finally, microtubule disruption-induced Akt deactivation contributes to delayed cell cycle progression and accelerated cell death. Taken together, we revealed that, after initiation, the overall intensity and duration of oncogenic Akt signaling are determined by microtubule dynamics, a mechanism that could be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Hakryul Jo
- Environmental Health Sciences, Yale University School of Public Health, New Haven, CT, 06520, USA.
| | - Fabien Loison
- Department of Pathology and Laboratory medicine, Harvard Medical School and Children's Hospital Boston, Boston, MA 02115 USA; Department of Microbiology, Faculty of Science, Mahidol University, Phayathai Campus, 272 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Hongbo R Luo
- Department of Pathology and Laboratory medicine, Harvard Medical School and Children's Hospital Boston, Boston, MA 02115 USA.
| |
Collapse
|
21
|
Shan WG, Zhang LW, Xiang JG, Zhan ZJ. Natural Friedelanes. Chem Biodivers 2013; 10:1392-434. [DOI: 10.1002/cbdv.201100256] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Indexed: 01/09/2023]
|
22
|
Yin F, Liu X, Li D, Wang Q, Zhang W, Li L. Bioinformatic analysis of chemokine (C-C motif) ligand 21 and SPARC-like protein 1 revealing their associations with drug resistance in ovarian cancer. Int J Oncol 2013; 42:1305-16. [PMID: 23404140 DOI: 10.3892/ijo.2013.1819] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/14/2013] [Indexed: 11/06/2022] Open
Abstract
Chemokine (C-C motif) ligand 21 (CCL21) and SPARC-like protein 1 (SPARCL1/MAST9/hevin/SC-1) are associated with various biological behavior in the development of cancers. Although the expression of CCL21 and SPARCL1 is downregulated in many solid tumors, their roles in ovarian cancer and their associations with drug resistance have rarely been studied. We performed a comprehensive bioinformatic analysis consisting of motif analysis, literature co-occurrence, gene/protein-gene/protein interaction network, protein-small molecule interaction network, and microRNAs enrichments which revealed that CCL21 and SPARCL1 directly or indirectly interact with a number of genes, proteins, small molecules and pathways associated with drug resistance in ovarian and other cancers. These results suggested that CCL21 and SPARCL1 may contribute to drug resistance in ovarian cancer. This study provided important information for further investigation of drug resistance-related functions of CCL21 and SPARCL1 in ovarian cancer.
Collapse
Affiliation(s)
- Fuqiang Yin
- Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | | | | | | | | | | |
Collapse
|
23
|
Jantas D, Roman A, Kuśmierczyk J, Lorenc-Koci E, Konieczny J, Lenda T, Lasoń W. The extent of neurodegeneration and neuroprotection in two chemical in vitro models related to Parkinson's disease is critically dependent on cell culture conditions. Neurotox Res 2013; 24:41-54. [PMID: 23307753 DOI: 10.1007/s12640-012-9374-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 12/27/2012] [Accepted: 12/28/2012] [Indexed: 01/21/2023]
Abstract
The proteasome inhibition and mitochondrial dysfunction are involved in pathomechanism of Parkinson's disease. The main aim of this study was to assess how particular culture conditions of human dopaminergic neuroblastoma SH-SY5Y cells could affect the extent of neurodegeneration induced by proteasome inhibitor-lactacystin (LC) and mitochondrial toxin-rotenone (Rot). This study revealed that induction of neuronal differentiation of SH-SY5Y cells with retinoic acid (RA-SH-SY5Y) caused a higher resistance of these cells to LC-evoked cell death when compared to undifferentiated cells (UN-SH-SY5Y). In contrast, RA-SH-SY5Y cells were more vulnerable than the UN-SH-SY5Y to Rot-induced cell damage. Furthermore, we found that a prolonged incubation of the cells under low serum condition (PLSC) significantly increased the LC toxicity in both differentiated and undifferentiated cells. Next, the effects of combined treatment with LC and Rot on cell viability were studied in RA-SH-SY5Y cells under PLSC and normal low serum condition (NLSC). At a low concentration, Rot (0.001-1 μM) attenuated the LC-evoked cell death in RA-SH-SY5Y cells exposed to NLSC. In contrast, under PLSC low concentrations of Rot lacked neuroprotective action while its higher levels (10 μM) enhanced the LC toxicity. Further, we showed that low concentrations of celastrol (Cel; 0.001 μM), a putative neuroprotective agent with antioxidant and anti-inflammatory properties, were able to partially attenuate the Rot-evoked toxicity under both PLSC and NLSC. On the other hand, Cel (0.001 and 0.01 μM) attenuated the LC-induced cell damage only under PLSC. Interestingly, higher concentrations of Cel (>1 μM) reduced cell viability in both UN- and RA-SH-SY5Y but only in UN-SH-SY5Y cells the effect was enhanced under PLSC. The obtained data indicate that toxicity of LC and Rot in SH-SY5Y cell line depends on the stage of cell differentiation and is enhanced in cells cultured for a longer time in low serum medium. Moreover, the neuroprotective properties of Rot and Cel against the LC-induced cell damage can be observed only under particular low serum conditions.
Collapse
Affiliation(s)
- D Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343, Kraków, Poland.
| | | | | | | | | | | | | |
Collapse
|
24
|
Jinwal UK, Akoury E, Abisambra JF, O'Leary JC, Thompson AD, Blair LJ, Jin Y, Bacon J, Nordhues BA, Cockman M, Zhang J, Li P, Zhang B, Borysov S, Uversky VN, Biernat J, Mandelkow E, Gestwicki JE, Zweckstetter M, Dickey CA. Imbalance of Hsp70 family variants fosters tau accumulation. FASEB J 2012; 27:1450-9. [PMID: 23271055 DOI: 10.1096/fj.12-220889] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Dysfunctional tau accumulation is a major contributing factor in tauopathies, and the heat-shock protein 70 (Hsp70) seems to play an important role in this accumulation. Several reports suggest that Hsp70 proteins can cause tau degradation to be accelerated or slowed, but how these opposing activities are controlled is unclear. Here we demonstrate that highly homologous variants in the Hsp70 family can have opposing effects on tau clearance kinetics. When overexpressed in a tetracycline (Tet)-based protein chase model, constitutive heat shock cognate 70 (Hsc70) and inducible Hsp72 slowed or accelerated tau clearance, respectively. Tau synergized with Hsc70, but not Hsp72, to promote microtubule assembly at nearly twice the rate of either Hsp70 homologue in reconstituted, ATP-regenerating Xenopus extracts supplemented with rhodamine-labeled tubulin and human recombinant Hsp72 and Hsc70. Nuclear magnetic resonance spectroscopy with human recombinant protein revealed that Hsp72 had greater affinity for tau than Hsc70 (I/I0 ratio difference of 0.3), but Hsc70 was 30 times more abundant than Hsp72 in human and mouse brain tissue. This indicates that the predominant Hsp70 variant in the brain is Hsc70, suggesting that the brain environment primarily supports slower tau clearance. Despite its capacity to clear tau, Hsp72 was not induced in the Alzheimer's disease brain, suggesting a mechanism for age-associated onset of the disease. Through the use of chimeras that blended the domains of Hsp72 and Hsc70, we determined that the reason for these differences between Hsc70 and Hsp72 with regard to tau clearance kinetics lies within their C-terminal domains, which are essential for their interactions with substrates and cochaperones. Hsp72 but not Hsc70 in the presence of tau was able to recruit the cochaperone ubiquitin ligase CHIP, which is known to facilitate the ubiquitination of tau, describing a possible mechanism of how the C-termini of these homologous Hsp70 variants can differentially regulate tau triage. Thus, efforts to promote Hsp72 expression and inhibit Hsc70 could be therapeutically relevant for tauopathies.
Collapse
Affiliation(s)
- Umesh K Jinwal
- Department of Pharmaceutical Sciences, University of South Florida Health Byrd Alzheimer's Institute, University of South Florida, Tampa, Florida 33613, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Klaić L, Morimoto RI, Silverman RB. Celastrol analogues as inducers of the heat shock response. Design and synthesis of affinity probes for the identification of protein targets. ACS Chem Biol 2012; 7:928-37. [PMID: 22380712 DOI: 10.1021/cb200539u] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The natural product celastrol (1) possesses numerous beneficial therapeutic properties and affects numerous cellular pathways. The mechanism of action and cellular target(s) of celastrol, however, remain unresolved. While a number of studies have proposed that the activity of celastrol is mediated through reaction with cysteine residues, these observations have been based on studies with specific proteins or by in vitro analysis of a small fraction of the proteome. In this study, we have investigated the spatial and structural requirements of celastrol for the design of suitable affinity probes to identify cellular binding partners of celastrol. Although celastrol has several potential sites for modification, some of these were not synthetically amenable or yielded unstable analogues. Conversion of the carboxylic acid functionality to amides and long-chain analogues, however, yielded bioactive compounds that induced the heat shock response (HSR) and antioxidant response and inhibited Hsp90 activity. This led to the synthesis of biotinylated celastrols (23 and 24) that were used as affinity reagents in extracts of human Panc-1 cells to identify Annexin II, eEF1A, and β-tubulin as potential targets of celastrol.
Collapse
Affiliation(s)
- Lada Klaić
- Department of Chemistry, ‡Department of Molecular Biosciences, §Rice Institute for Biomedical Research, and ∥Chemistry of
Life Processes Institute and Center for Molecular Innovation and Drug
Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Richard I. Morimoto
- Department of Chemistry, ‡Department of Molecular Biosciences, §Rice Institute for Biomedical Research, and ∥Chemistry of
Life Processes Institute and Center for Molecular Innovation and Drug
Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Richard B. Silverman
- Department of Chemistry, ‡Department of Molecular Biosciences, §Rice Institute for Biomedical Research, and ∥Chemistry of
Life Processes Institute and Center for Molecular Innovation and Drug
Discovery, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
26
|
Agarwal N, Sonpavde G, Sartor O. Cabazitaxel for the treatment of castration-resistant prostate cancer. Future Oncol 2011; 7:15-24. [PMID: 21174534 DOI: 10.2217/fon.10.168] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Treatment with docetaxel-based chemotherapy results in improved survival in patients with metastatic castration-resistant prostate cancer. However, all patients eventually develop progressive disease associated with poor outcomes. In this article, we discuss the available second-line therapeutic options following docetaxel, with a special focus on cabazitaxel, which is the first agent to yield extended survival as second-line therapy following docetaxel. Cabazitaxel, a novel semi-synthetic taxane, is effective even in docetaxel-resistant model systems. Recently, results of the Phase III TROPIC trial demonstrated improved survival with cabazitaxel plus prednisone compared with mitoxantrone and prednisone in patients with progressive metastatic castration-resistant prostate cancer, following prior docetaxel, which led to approval by the US FDA.
Collapse
|
27
|
Management of cytoskeleton architecture by molecular chaperones and immunophilins. Cell Signal 2011; 23:1907-20. [PMID: 21864675 DOI: 10.1016/j.cellsig.2011.07.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/22/2011] [Accepted: 07/26/2011] [Indexed: 11/20/2022]
Abstract
Cytoskeletal structure is continually remodeled to accommodate normal cell growth and to respond to pathophysiological cues. As a consequence, several cytoskeleton-interacting proteins become involved in a variety of cellular processes such as cell growth and division, cell movement, vesicle transportation, cellular organelle location and function, localization and distribution of membrane receptors, and cell-cell communication. Molecular chaperones and immunophilins are counted among the most important proteins that interact closely with the cytoskeleton network, in particular with microtubules and microtubule-associated factors. In several situations, heat-shock proteins and immunophilins work together as a functionally active heterocomplex, although both types of proteins also show independent actions. In circumstances where homeostasis is affected by environmental stresses or due to genetic alterations, chaperone proteins help to stabilize the system. Molecular chaperones facilitate the assembly, disassembly and/or folding/refolding of cytoskeletal proteins, so they prevent aberrant protein aggregation. Nonetheless, the roles of heat-shock proteins and immunophilins are not only limited to solve abnormal situations, but they also have an active participation during the normal differentiation process of the cell and are key factors for many structural and functional rearrangements during this course of action. Cytoskeleton modifications leading to altered localization of nuclear factors may result in loss- or gain-of-function of such factors, which affects the cell cycle and cell development. Therefore, cytoskeletal components are attractive therapeutic targets, particularly microtubules, to prevent pathological situations such as rapidly dividing tumor cells or to favor the process of cell differentiation in other cases. In this review we will address some classical and novel aspects of key regulatory functions of heat-shock proteins and immunophilins as housekeeping factors of the cytoskeletal network.
Collapse
|
28
|
Geng F, Tang L, Li Y, Yang L, Choi KS, Kazim AL, Zhang Y. Allyl isothiocyanate arrests cancer cells in mitosis, and mitotic arrest in turn leads to apoptosis via Bcl-2 protein phosphorylation. J Biol Chem 2011; 286:32259-67. [PMID: 21778226 DOI: 10.1074/jbc.m111.278127] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Allyl isothiocyanate (AITC) occurs in many commonly consumed cruciferous vegetables and exhibits significant anti-cancer activities. Available data suggest that it is particularly promising for bladder cancer prevention and/or treatment. Here, we show that AITC arrests human bladder cancer cells in mitosis and also induces apoptosis. Mitotic arrest by AITC was associated with increased ubiquitination and degradation of α- and β-tubulin. AITC directly binds to multiple cysteine residues of the tubulins. AITC induced mitochondrion-mediated apoptosis, as shown by cytochrome c release from mitochondria to cytoplasm, activation of caspase-9 and caspase-3, and formation of TUNEL-positive cells. Inhibition of caspase-9 blocked AITC-induced apoptosis. Moreover, we found that apoptosis induction by AITC depended entirely on mitotic arrest and was mediated via Bcl-2 phosphorylation at Ser-70. Pre-arresting cells in G(1) phase by hydroxyurea abrogated both AITC-induced mitotic arrest and Bcl-2 phosphorylation. Overexpression of a Bcl-2 mutant prevented AITC from inducing apoptosis. We further showed that AITC-induced Bcl-2 phosphorylation was caused by c-Jun N-terminal kinase (JNK), and AITC activates JNK. Taken together, this study has revealed a novel anticancer mechanism of a phytochemical that is commonly present in human diet.
Collapse
Affiliation(s)
- Feng Geng
- Department of Cancer Prevention and Control, California Institute of Technology, Pasadena, California 91125, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
The main anticancer bullets of the Chinese medicinal herb, thunder god vine. Molecules 2011; 16:5283-97. [PMID: 21701438 PMCID: PMC6264543 DOI: 10.3390/molecules16065283] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 06/17/2011] [Accepted: 06/20/2011] [Indexed: 11/17/2022] Open
Abstract
The thunder god vine or Tripterygium wilfordii Hook. F. is a representative Chinese medicinal herb which has been used widely and successfully for centuries in treating inflammatory diseases. More than 100 components have been isolated from this plant, and most of them have potent therapeutic efficacy for a variety of autoimmune and inflammatory diseases. In the past four decades, the anticancer activities of the extracts from this medicinal herb have attracted intensive attention by researchers worldwide. The diterpenoid epoxide triptolide and the quinone triterpene celastrol are two important bioactive ingredients that show a divergent therapeutic profile and can perturb multiple signal pathways. Both compounds promise to turn traditional medicines into modern drugs. In this review, we will mainly address the anticancer activities and mechanisms of action of these two agents and briefly describe some other antitumor components of the thunder god vine.
Collapse
|
30
|
Lu KW, Chen JC, Lai TY, Yang JS, Weng SW, Ma YS, Lin HY, Wu RSC, Wu KC, Wood WG, Chung JG. Gypenosides suppress growth of human oral cancer SAS cells in vitro and in a murine xenograft model: the role of apoptosis mediated by caspase-dependent and caspase-independent pathways. Integr Cancer Ther 2011; 11:129-40. [PMID: 21665877 DOI: 10.1177/1534735411403306] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Gypenosides (Gyp) are the major components of Gynostemma pentaphyllum Makino. The authors investigated the effects of Gyp on cell morphology, viability, cell cycle distribution, and induction of apoptosis in human oral cancer SAS cells and the determination of murine SAS xenograft model in vivo. EXPERIMENTAL DESIGN Flow cytometry was used to quantify the percentage of viable cells; cell cycle distribution; sub-G1 phase (apoptosis); caspase-3, -8, and -9 activity; reactive oxygen species (ROS) production, intracellular Ca(2+) determination; and the level of mitochondrial membrane potential (ΔΨ(m)). Western blotting was used to examine levels of apoptosis-associated proteins, and confocal laser microscopy was used to examine the translocation of proteins in cells. RESULTS Gyp induced morphological changes, decreased the percentage of viable cells, caused G0/G1 phase arrest, and triggered apoptotic cell death in SAS cells. Cell cycle arrest induced by Gyp was associated with apoptosis. The production of ROS, increased intracellular Ca(2+) levels, and the depolarization of ΔΨ(m) were observed. Gyp increased levels of the proapoptotic protein Bax but inhibited the levels of the antiapoptotic proteins Bcl-2 and Bcl-xl. Gyp also stimulated the release of cytochrome c and Endo G. Translocation of GADD153 to the nucleus was stimulated by Gyp. Gyp in vivo attenuated the size and volume of solid tumors in a murine xenograft model of oral cancer. CONCLUSIONS Gyp-induced cell death occurs through caspase-dependent and caspase-independent apoptotic signaling pathways, and the compound reduced tumor size in a xenograft nu/nu mouse model of oral cancer.
Collapse
Affiliation(s)
- Kung-Wen Lu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Deactivation of Akt by a small molecule inhibitor targeting pleckstrin homology domain and facilitating Akt ubiquitination. Proc Natl Acad Sci U S A 2011; 108:6486-91. [PMID: 21464312 DOI: 10.1073/pnas.1019062108] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The phosphatidylinositol-3,4,5-triphosphate (PIP3) binding function of pleckstrin homology (PH) domain is essential for the activation of oncogenic Akt/PKB kinase. Following the PIP3-mediated activation at the membrane, the activated Akt is subjected to other regulatory events, including ubiquitination-mediated deactivation. Here, by identifying and characterizing an allosteric inhibitor, SC66, we show that the facilitated ubiquitination effectively terminates Akt signaling. Mechanistically, SC66 manifests a dual inhibitory activity that directly interferes with the PH domain binding to PIP3 and facilitates Akt ubiquitination. A known PH domain-dependent allosteric inhibitor, which stabilizes Akt, prevents the SC66-induced Akt ubiquitination. A cancer-relevant Akt1 (e17k) mutant is unstable, making it intrinsically sensitive to functional inhibition by SC66 in cellular contexts in which the PI3K inhibition has little inhibitory effect. As a result of its dual inhibitory activity, SC66 manifests a more effective growth suppression of transformed cells that contain a high level of Akt signaling, compared with other inhibitors of PIP3/Akt pathway. Finally, we show the anticancer activity of SC66 by using a soft agar assay as well as a mouse xenograft tumor model. In conclusion, in this study, we not only identify a dual-function Akt inhibitor, but also demonstrate that Akt ubiquitination could be chemically exploited to effectively facilitate its deactivation, thus identifying an avenue for pharmacological intervention in Akt signaling.
Collapse
|