1
|
Ye H, Wang Y, Zhang X, Yang L, Cai B, Zhang D, Peng B. Characterization of global research trends and prospects on celastrol, a principal bioactive ingredient of Tripterygium wilfordii Hook F: bibliometric analysis. PHARMACEUTICAL BIOLOGY 2025; 63:15-26. [PMID: 39745069 DOI: 10.1080/13880209.2024.2443424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
CONTEXT Celastrol, acknowledged as a prominent exemplar of the potential for transforming traditional medicinal compounds into contemporary pharmaceuticals, has garnered considerable attention owing to its extensive pharmacological activities. The increasing volume of publications concerning celastrol highlights its importance in current scientific inquiry. Despite the growing interest in this compound, a bibliometric analysis focused on this subject remains to be undertaken. OBJECTIVE Our study explored a bibliometric approach to identify and characterize global research trends and frontiers related to celastrol, including mapping research outputs, influential contributors, and thematic areas, as well as highlighting gaps and opportunities for future investigations. MATERIALS AND METHODS In this study, we utilized the Web of Science Core Collection (WoSCC) to source and review articles related to celastrol published from 1997 to 2023. The bibliometric analysis was conducted using the R package 'Bibliometrix,' supplemented by visualization tools including CiteSpace, VOSviewer, and GraphPad Prism 10. RESULTS Celastrol related research papers have exhibited an upward trend annually and can be categorized into three distinct phases, each highlighting different areas of focus. China, the United States, and South Korea rank as the top three nations for publication volume, with varied research interests across these countries. Several prolific research teams have emerged, each with distinct areas of interest. Examining the primary research domains of celastrol (anti-inflammatory, anticancer, and toxicity) reveals a notable intersection between the first two domains. DISCUSSION AND CONCLUSIONS The scope and depth of celastrol research have been steadily expanding, with regional and team-specific variations. Key research areas include anti-inflammatory, anticancer, and toxicity studies. Future research is expected to focus on enhancing the effectiveness and reducing the toxicity of celastrol. Meanwhile, given the multi-target characteristics of celastrol's effects, integrating methods such as network biology and molecular simulation will provide a novel perspective for celastrol research.
Collapse
Affiliation(s)
- Huizi Ye
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Yufang Wang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Xue Zhang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lin Yang
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Banglan Cai
- Postgraduate training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
- School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Denghai Zhang
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Bin Peng
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| |
Collapse
|
2
|
Qi M, Zhang K, Zhang X, Zhu Y, Cai B, Wang C, Zhao G, Zhang D, Zhang J. Arginine tagged liposomal carrier for the delivery of celastrol for ferroptosis-induced hepatocellular carcinoma therapy. Colloids Surf B Biointerfaces 2025; 250:114546. [PMID: 39919344 DOI: 10.1016/j.colsurfb.2025.114546] [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: 10/10/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025]
Abstract
Hepatocellular carcinoma (HCC) is a predominant malignant liver tumor that cannot be efficiently treated because of poor response, toxicity, and drug resistance. Ferroptosis is an iron-dependent way of cell death associated with abnormal intracellular lipid metabolism. Celastrol (Cel) has the ability to inhibit the progression of HCC by regulating multiple signaling pathways and induce ferroptosis. However, Cel exists the limitations of low water solubility, low oral bioavailability, and high organ toxicity. Cel was encapsulated in polyethylene glycol-based liposomes modified with L-arginine (Cel@Lip-Arg). Cel@Lip-Arg has a uniform size distribution (∼100 nm), high drug loading (80 %), and excellent ability to target liver cancer cells. In vitro experiments demonstrated that Cel@Lip-Arg considerably suppressed the activity of HuH7 (hepatoma) cells but had a negligible effect on L02 (normal) cells. Cel@Lip-Arg induced ferroptosis in hepatoma cells by promoting transferrin receptor expression, inhibiting system xc- and glutathione peroxidase 4, and favoring intracellular peroxide accumulation. In vivo experiments revealed that Cel@Lip-Arg plays a therapeutic role by inducing ferroptosis. Compared to Cel, Cel@Lip-Arg had a higher anti-hepatoma activity and effectively reduced the toxicity of Cel in mice. Cel@Lip-Arg-induced ferroptosis was concluded to be an attractive strategy for the precise treatment of HCC.
Collapse
Affiliation(s)
- Manman Qi
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Kai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China
| | - Xue Zhang
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Yuzhao Zhu
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Banglan Cai
- School of Basic Medicine, Ningxia Medical University, Ningxia 750004, PR China
| | - Chao Wang
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China
| | - Gang Zhao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Denghai Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Jian Zhang
- School of Medicine, Shanghai University, Shanghai 200444, PR China; Shanghai Universal Medical Imaging Diagnostic Center, Shanghai University, Shanghai 200233, PR China.
| |
Collapse
|
3
|
Quadros M, Goyal M, Chauhan G, Gadhave D, Gupta V. An Inhaled Nanoemulsion Encapsulating a Herbal Drug for Non-Small Cell Lung Cancer (NSCLC) Treatment. Pharmaceutics 2025; 17:540. [PMID: 40430833 PMCID: PMC12115302 DOI: 10.3390/pharmaceutics17050540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Celastrol (Cela), a phytochemical extracted from Tripterygium wilfordii, has been extensively investigated for its potential anti-inflammatory, anti-psoriatic, antioxidant, neuroprotective, and antineoplastic properties. However, its clinical translation is limited due to poor bioavailability, low solubility, and nonspecific toxicity. This study aimed to develop and evaluate an inhalable Cela-loaded nanoemulsion (NE) formulation to enhance targeted drug delivery and therapeutic efficacy in non-small cell lung cancer (NSCLC). Methods: The NE formulation was optimized using Capmul MCM (25%), Tween 80 (20%), Transcutol HP (5%), and water (50%) as the oil, surfactant, co-surfactant, and aqueous phase, respectively. Physicochemical characterization included globule size, zeta potential, and drug release in simulated lung fluid. In vitro aerosolization performance, cytotoxicity in NSCLC cell lines (A549), scratch and clonogenic assays, and 3D tumor spheroid models were employed to assess therapeutic potential. Results: The NE showed a globule size of 201.4 ± 3.7 nm and a zeta potential of -15.7 ± 0.2 mV. Drug release was sustained, with 20.4 ± 5.5%, 29.1 ± 10%, 64.6 ± 4.1%, and 88.1 ± 5.2% released at 24, 48, 72, and 120 h, respectively. In vitro aerosolization studies indicated a median aerodynamic particle size of 4.8 ± 0.2 μm, confirming its respirability in the lung. Cell culture studies indicated higher toxicity of NE-Cela in NSCLC cells. NE-Cela significantly reduced A549 cell viability, showing a ~6-fold decrease in IC50 (0.2 ± 0.1 μM) compared to Cela alone (1.2 ± 0.2 μM). Migration and clonogenic assays demonstrated reduced cell proliferation, and 3D spheroid models supported its therapeutic activity in tumor-like environments. Conclusions: The inhalable NE-Cela formulation improved Cela's physicochemical limitations and demonstrated enhanced anti-cancer efficacy in NSCLC models. These findings support its potential as a targeted, well-tolerated therapeutic option for lung cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, NY 11439, USA
| |
Collapse
|
4
|
Gu Y, Yang M, Wang W, Li L, Ma Y, Liu W, Zhao Q. YX-112, a novel celastrol-derived PROTAC, inhibits the development of triple-negative breast cancer by targeting the degradation of multiple proteins. Front Pharmacol 2025; 16:1571135. [PMID: 40303918 PMCID: PMC12037497 DOI: 10.3389/fphar.2025.1571135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Background Celastrol is an effective component of the plant Tripterygium wilfordii Hook. f., which has a high inhibitory effect on triple-negative breast cancer. However, the therapeutic window of celastrol is narrow, and as a multi-target drug, its mechanism of action in triple-negative breast cancer is not very clear. Therefore, developing new celastrol derivatives has become an urgent task. Method In this work, we apply the PROTAC strategy to design and synthesis novel celastrol derivative. The antiproliferative activity of compound YX-112 against various types of cells was detected by CCK8 method. DIA-based quantitative proteomics, Western blot was used to explore the mechanism of compound YX-112 on triple-negative breast cancer MDA-MB-231 cells. Finally, the binding mode between compound YX-112 and target protein was predicted through molecular docking. Results We developed a novel PROTAC YX-112 of celastrol, which was extremely sensitive to the triple-negative breast cancer MDA-MB-231 cells, with an IC50 value of 0.32 ± 0.04 μM, and its antiproliferative activity was 3 times that of celastrol. Subsequently, through DIA-based quantitative proteomics and Western blot validation experiments, it was found that YX-112 could target the degradation of CHEK1 and PIK3R2 proteins in MDA-MB-231 cells in a ubiquitin-proteasome dependent manner, indicating that it could be used as a degrader of CHEK1 and PIK3R2 proteins. Additionally, YX-112 could effectively inhibit the expression levels of CDK4 and p-AKT, and its inhibitory effect was stronger than that of celastrol. Finally, molecular docking predicted the binding mode between celastrol and CHEK1, showing that celastrol could form hydrogen bond interaction with the key residue GLN13. Conclusion This study provides new insights into the derivation of celastrol and its molecular mechanisms of action.
Collapse
Affiliation(s)
- Yongxue Gu
- Thyroid and Breast Medical Center, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, China
| | - Mengmeng Yang
- Thyroid and Breast Medical Center, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, China
| | - Wenbin Wang
- Thyroid and Breast Medical Center, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, China
| | - Lihua Li
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Ying Ma
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wenshan Liu
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
- Shandong Key Laboratory of Medicine and Health (Clinical Applied Pharmacology), Department of Pharmacy, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Qiang Zhao
- Department of Anesthesiology, Weifang People’s Hospital, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
5
|
Wang X, Abu Bakar MH, Liqun S, Kassim MA, Shariff KA, Karunakaran T. Targeting metabolic diseases with celastrol: A comprehensive review of anti-inflammatory mechanisms and therapeutic potential. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119560. [PMID: 40015541 DOI: 10.1016/j.jep.2025.119560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii is a traditional Chinese medicine used to treat rheumatic diseases, with properties such as clearing heat, detoxifying, dispelling wind, and relieving pain. In recent years, its active compound, celastrol, garnered significant attention for its potential therapeutic effects on metabolic diseases. Celastrol exhibits bioactivities such as regulating metabolic functions and anti-inflammatory effects, positioning it as a promising candidate for the treatment of obesity, diabetes, atherosclerosis (AS), and non-alcoholic fatty liver disease (NAFLD). AIM OF THE REVIEW This review aims to explore the pharmacological mechanisms of celastrol in metabolic diseases, focusing on its anti-inflammatory mechanisms and metabolic regulation effects, providing theoretical support for further investigation of its therapeutic potential in metabolic diseases. METHODS Literature was retrieved from PubMed, Web of Science, Scopus, Cochrane, and Google Scholar. This review primarily focuses on anti-inflammatory mechanisms of celastrol, its metabolic regulation, and toxicity studies, by systematically analyzing its effects in obesity, diabetes, AS, and NAFLD, providing scientific evidence for its potential clinical applications. RESULTS Celastrol regulates multiple signaling pathways, particularly inhibiting NF-κB and activating AMPK, reducing the production of pro-inflammatory cytokines and improving insulin sensitivity, enhancing its therapeutic potential in metabolic diseases. Additionally, celastrol regulates adipogenesis and energy metabolism by influencing key transcription factors such as PPARγ and SREBP-1c. Numerous studies highlight its role in alleviating oxidative stress and improving mitochondrial function, further enhancing its metabolic benefits. CONCLUSION In summary, celastrol holds great promise as a multi-target therapeutic agent for metabolic diseases, offering anti-inflammatory, metabolic regulatory, and antioxidative benefits. Despite these, challenges remain for the clinical application of celastrol due to its poor bioavailability and potential toxicity. Advanced formulation strategies and targeted delivery systems are urgently needed to overcome challenges related to bioavailability and clinical translation.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia; Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Song Liqun
- Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohd Asyraf Kassim
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia
| | - Khairul Anuar Shariff
- School of Materials & Mineral Resources Engineering, Universiti Sains Malaysia, Nibong Tebal, 14300, Penang, Malaysia
| | | |
Collapse
|
6
|
Zou L, Hou Y, Nie X, Wang S, Tian S, Sun Z, Sun Z, Xu X, Li G, Ma G, Liu H. All-Small-Molecule Supramolecular Hydrogel Combining Self-Delivery and ROS-Responsive Release for Inhibiting Tumor Growth and Postoperative Recurrence. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13494-13512. [PMID: 39993162 DOI: 10.1021/acsami.4c20852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Supramolecular hydrogels show unprecedented advantages and have attracted widespread attention in biomedical sciences. However, it is challenging for bioactive star molecules, such as celastrol, to meet ideal formation conditions. Here, we report a dynamic covalent method to construct a dihydrol-type celastrol-phenylenediboronic acid-guanosine (DHcelPBG) supramolecular hydrogel. The DHcelPBG hydrogel can effectively accelerate 4T1 cell apoptosis by modulating the PI3K/Akt signaling pathway. Especially, the DHcelPBG hydrogel can serve as a self-delivery platform for reactive oxygen species (ROS)-facilitated self-release. An excessive ROS-containing tumor microenvironment can promote the obtained DHcelPBG hydrogel to kill more 4T1 tumor cells. Meanwhile, the hydrogel also exhibits distinguished degradability and biocompatibility. Subsequently, the orthotopic 4T1 tumor model results further demonstrate that the DHcelPBG hydrogel remarkably inhibits tumor growth and does not damage healthy tissue. In the postoperative recurrence 4T1 tumor model, the DHcelPBG hydrogel also effectively prevents postoperative tumor recurrence and lung metastasis without causing adverse side effects, resulting in an extended lifetime. The DHcelPBG hydrogel also exhibits distinguished degradability and biocompatibility. The DHcelPBG hydrogel integrates ROS-responsiveness, localized self-delivery, and antitumor activity into one system for breast cancer treatment with fewer side effects, showing great potential for clinical transformation in cancer therapy.
Collapse
Affiliation(s)
- Linjun Zou
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Yong Hou
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Xueqiang Nie
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Shengchen Wang
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Sichao Tian
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Zhaocui Sun
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Zhonghao Sun
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xudong Xu
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Guang Li
- Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan Branch, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Jinghong 666100, China
| | - Guoxu Ma
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Haitao Liu
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education; State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
7
|
Yu Y, Wang J, Ruan L, Chen L, Khan MZ, You A, Wang C, Li L, Ren H, Wang T, Liu W. Evaluation of Celastrol Antiviral Activity Against Equid Alphaherpesvirus Type 8 Infection. Viruses 2025; 17:347. [PMID: 40143276 PMCID: PMC11945448 DOI: 10.3390/v17030347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Equid alphaherpesvirus type 8 (EHV-8) is a contagious pathogen that causes reproductive disorders, respiratory diseases, and viral encephalitis in equids, resulting in significant economic losses for the global horse and donkey industries. Currently, there are no approved antiviral drugs or vaccines available for EHV-8 control. In this study, we investigated the antiviral efficacy of celastrol against EHV-8 both in vitro and in vivo. Our results demonstrated that celastrol significantly inhibited EHV-8 infection in Rabbit kidney (RK-13) and equine dermal cells (NBL-6) in a dose-dependent manner. Mechanistic studies revealed that celastrol interfered with viral replication at multiple stages of the infection cycle. Furthermore, we found that celastrol induced an antiviral interferon response through activation of the Nrf2/HO-1 signaling pathway. Importantly, celastrol treatment significantly reduced EHV-8 replication and ameliorated lung pathology in a mouse model. These findings suggest that celastrol may represent a promising therapeutic agent for the treatment of EHV-8 infections.
Collapse
Affiliation(s)
- Yue Yu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Jiayu Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Lian Ruan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Li Chen
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Muhammad Zahoor Khan
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Anrong You
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Changfa Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Liangliang Li
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Huiying Ren
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Tongtong Wang
- Liaocheng Research Institute of Donkey High-Efficiency Breeding and Ecological Feeding, Liaocheng University, Liaocheng 252000, China (M.Z.K.)
| | - Wenhua Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
8
|
Liu Y, Song J, Guo Y, Li S, Yuan M, Tang J, Wang Y, Li M, Guo Y, Guo L. Synergistic therapy with celastrol-curcumin multifunctional nanomedicine: Anti-hepatocellular carcinoma and reduced hepatotoxicity. Int J Pharm 2025; 671:125289. [PMID: 39880142 DOI: 10.1016/j.ijpharm.2025.125289] [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: 10/16/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Hepatocellular carcinoma is one of the leading causes of cancer deaths globally and a key hindrance to extending life expectancy. Celastrol (CEL) demonstrates excellent antitumor activity, but faces challenges like low solubility and a narrow therapeutic window, limiting its clinical application. To address these limitations, drug combinations and nano-delivery systems have emerged as effective solutions. Curcumin (CUR), known for its antitumor and hepatoprotective effects, also exhibits good biocompatibility and the ability to mitigate drug-induced liver injury. Considering the complementary properties of CEL and CUR, including CEL's potent antitumor activity and CUR's hepatoprotective effects, we developed a novel self-assembling nanodrug delivery system (CCPN) for the co-loading of both compounds. CCPN nanoparticles were constructed through non-covalent interactions, including hydrogen bonding, π-π stacking, and electrostatic forces, which confer good stability and significantly enhance the solubility and bioavailability of CEL and CUR. Extensive in vitro and in vivo experiments demonstrated that CCPN effectively reduced CEL-induced hepatotoxicity in zebrafish and mouse models, exhibiting good biosafety. Additionally, CUR's fluorescence provides a unique advantage for real-time monitoring of drug distribution and release, facilitating the tracking of therapeutic progress. Furthermore, CCPN nanoparticles enhanced delivery efficiency in HepG2 cells, exhibiting superior anti-liver tumor outcomes, which are associated with the promotion of apoptosis in tumor cells. This study presents CCPN as a promising therapeutic strategy for hepatocellular carcinoma, integrating reduced hepatotoxicity, self-monitoring capabilities, and superior therapeutic efficacy.
Collapse
Affiliation(s)
- Yushi Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China; Department of Pharmacy, Chengdu Seventh People's Hospital, Chengdu 610213 China
| | - Jiawen Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yurou Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Meifeng Li
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China.
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China.
| |
Collapse
|
9
|
Liu X, Su YX, Yang YM, Li RT, Zhang ZJ. The Small Molecules of Plant Origin with Anti-Glioma Activity. Int J Mol Sci 2025; 26:1942. [PMID: 40076568 PMCID: PMC11900624 DOI: 10.3390/ijms26051942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Gliomas, originating from glial cells, are prevalent and aggressive brain tumors with high recurrence rates and poor prognosis. Despite advancements in surgical, radiation, and chemotherapeutic treatments, the survival rates remain low. Current standard therapies, such as Temozolomide, have limitations due to cytotoxicity, restricted effectiveness, and severe side effects. So, the development of safer anti-glioma agents is the need of the hour. Bioactive compounds of plant origin, either natural or synthetic, have potential implications due to them actively attacking different targets with a wide range of bioactivities, including anti-glioma activities. In this review, for the first time, there is an overall overview of 51 small molecules of plant origin and seven of their synthetic derivatives, represented as anti-glioma agents in the past decades. The goal of the present review is to provide a summary to comprehend the anti-glioma effects of these compounds in addition to providing a reference for preclinical research into novel anti-glioma agents for future clinical application.
Collapse
Affiliation(s)
| | | | | | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (X.L.); (Y.-X.S.); (Y.-M.Y.)
| | - Zhi-Jun Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (X.L.); (Y.-X.S.); (Y.-M.Y.)
| |
Collapse
|
10
|
Xue R, Qin C, Li L, Huang L, Tang K, Chen J, Liang H, Xu H, Qin X, Yang C, Tan Q. SRF/SLC31A1 signaling promotes cuproptosis induced by celastrol in NSCLC. Int Immunopharmacol 2025; 148:114165. [PMID: 39930648 DOI: 10.1016/j.intimp.2025.114165] [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: 10/31/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 05/08/2025]
Abstract
Faced with the highly malignant challenge of lung cancer, traditional chemotherapeutic agents, although predominantly inducing apoptosis, are severely limited in their therapeutic effect by the overexpression of anti-apoptotic proteins in lung cancer. Recently discovered copper-induced non-apoptotic cell death, known as cuproptosis, represents a novel mechanism for regulating cell death. Whether Celastrol (Cel), a potential anti-tumor drug, can counter non-small cell lung cancer (NSCLC) through inducing cuproptosis remains to be thoroughly investigated. This study demonstrates that the copper chelator tetrathiomolybdate (TTM) is more effective in rescuing Cel-induced NSCLC cell death compared to other inhibitors. RNA sequencing revealed that Cel significantly upregulates the copper transporter protein SLC31A1. In addition, Cel also promotes intracellular copper accumulation, reduces GSH levels, and exhibits features of cuproptosis, including loss of iron-sulfur cluster proteins (FDX1, SDHB, POLD1), increased HSP70, and DLAT oligomerization. Experiments also found that Cel significantly increases reactive oxygen species (ROS) levels, reduces mitochondrial membrane potential, and lowers ATP levels. It was predicted through online databases that SRF may be the transcription factor for SLC31A1, and this was validated through overexpression experiments. In vivo data demonstrated that Cel significantly inhibits tumor growth without damaging the heart, liver, or kidneys of mice. This study first reveals that celastrol disrupts intracellular copper homeostasis through the SRF/SLC31A1 pathway, promoting cuproptosis in NSCLC cells, providing support for Cel as a potential safe and effective chemotherapeutic agent.
Collapse
Affiliation(s)
- Rui Xue
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Chuling Qin
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Lanyu Li
- Guilin Medical University Laboratory Animal Center, Guilin 541001 China
| | - Lingyue Huang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Ke Tang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Jianning Chen
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Huihui Liang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Huimin Xu
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Xuanjie Qin
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Chang Yang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Qinyou Tan
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China; Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001 China; China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001 China.
| |
Collapse
|
11
|
Deng X, He J, Deng W, Deng W, Zhu X, Luo H, Wang D. Celastrol ameliorates lipopolysaccharide (LPS)-induced acute lung injury by improving mitochondrial function through AMPK/PGC-1α/Nrf1-dependent mechanism. Free Radic Biol Med 2025; 227:210-220. [PMID: 39643138 DOI: 10.1016/j.freeradbiomed.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/16/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Acute lung injury (ALI) is a devastating clinical syndrome without effective therapy. Celastrol, as a natural anti-inflammatory compound, has showed therapeutic potential against inflammatory diseases. In this study, we have investigated the potential effect of Celastrol on lipopolysaccharide (LPS)-induced ALI. C57BL/6J mice, Nrf1-knockout mice and A549 (human alveolar epithelial cell line) cells were used to investigate the protective role of Celastrol in LPS-induced ALI. Our data showed that administration of Celastrol significantly alleviated lung pathologic injury and increased the survival rate, which was associated with the improvement of mitochondrial function in the injured lung. Moreover, Celastrol enhanced phosphorylation of AMP-activated protein kinase (AMPK) and expression of peroxisome proliferator-activated receptor coactivator protein-1α (PGC-1α), thereby increasing the nuclear translocation of nuclear respiratory factor 1 (Nrf1) and subsequent up-regulation of its downstream mitochondria electron transport chain complex I (NDUF) gene expression, which induced an increase in mitochondrial complex Ⅰ activity. The beneficial effects of Celastrol on regulation of Nrf1 were abolished by inhibition of AMPK and PGC-1α. Finally, in Nrf1 deficient mice, the protective effects of Celastrol on LPS-induced ALI were largely vanished. Our data indicated that Celastrol can prevent LPS-induced ALI by improving mitochondrial function through AMPK/PGC-1α/Nrf1-dependent mechanism, suggesting that Celastrol may represent a novel therapeutic potential for LPS-induced ALI.
Collapse
Affiliation(s)
- Xinyu Deng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing He
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenpeng Deng
- Department of Laboratory and Blood Transfusion of Jiangbei Campus, The First Affiliated Hospital of Army Medical University (The 958th hospital of Chinese People's Liberation Army), Chongqing, China
| | - Wang Deng
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingyu Zhu
- Faculty of Foresty, University of British Columbia, Vancouver, BC, Canada
| | - Hao Luo
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Daoxin Wang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
A Yehia FAZ, Abbas HA, Ibrahim TM, Mansour B, Awan ZA, Al-Rabia MW, Abdulaal WH, Zeyadi MA, Okbazghi SZ, Ibrahim TS, Hegazy WAH, Gomaa SE. Celastrol boosts fluconazole efficacy against vaginal candidiasis: in vitro and in vivo evidence. AMB Express 2025; 15:18. [PMID: 39881021 PMCID: PMC11780038 DOI: 10.1186/s13568-025-01824-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/12/2025] [Indexed: 01/31/2025] Open
Abstract
Candida albicans is a commensal fungus that naturally inhabits the vagina. However, overgrowth of C. albicans can result in vulvovaginal candidiasis (VVC), one of the most prevalent fungal infections affecting women. The rapid emergence of azole resistance in C. albicans, in addition to the limited available antifungal agents, complicates the treatment and emphasizes the urgent need for novel therapeutic options. Efflux-mediated azole resistance is a common resistance mechanism in fluconazole (FLZ)-resistant C. albicans. Combination therapy using natural compounds is a potential approach that can restore fluconazole's antifungal activity in azole-resistant isolates via efflux pump inhibition. This study aimed to evaluate the ability of celastrol, a natural triterpene, to retrieve FLZ antifungal activity against azole-resistant C. albicans in vitro and in vivo. Celastrol did not exhibit antifungal activity against the tested clinical isolates; however, the sub-MIC of celastrol inhibited rhodamine 6G (R6G) efflux and increased R6G accumulation inside celastrol-treated C. albicans cells. Synergy was spotted between celastrol and FLZ via a checkerboard assay. Quantification of m-RNA levels of efflux-mediated azole resistance genes within azole-resistant C. albicans demonstrated CDR1 overexpression. Upon celastrol treatment, a significant decline in ABC transporters transcript levels were detected. Moreover, molecular docking demonstrated that celastrol is a potential ABC efflux transporters blocker that successfully fits into target binding pockets. A negligible hemolytic effect of celastrol against human erythrocytes was observed. In the in vivo model of VVC, the combination of FLZ and celastrol in vaginal gel revealed a drastic reduction in the fungal burden with apparently normal vaginal tissue. Celastrol promising in vitro and in vivo findings strengthen its future use for the treatment of azole-resistant C. albicans.
Collapse
Affiliation(s)
- Fatma Al-Zahraa A Yehia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Hisham A Abbas
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Tarek M Ibrahim
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Basem Mansour
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Belqas, 11152, Egypt
- Department of Pharmaceutical Chemistry, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Zuhier A Awan
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed W Al-Rabia
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Mohamed Saeed Tamer Chair for Pharmaceutical Industries, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Wesam H Abdulaal
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mustafa Adnan Zeyadi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Solomon Z Okbazghi
- Global Analytical and Pharmaceutical Development, Alexion Pharmaceuticals, New Haven, Connecticut, 06510, USA
| | - Tarek S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Wael A H Hegazy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
- Pharmacy Program, Department of Pharmaceutical Sciences, College of Health Sciences, 113, Muscat, Oman.
| | - Salwa E Gomaa
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
13
|
Ding Q, Chen H, Zhang Y, Yang J, Li M, He Q, Mei L. Innovative integration of nanomedicines and phototherapy to modulate autophagy for enhanced tumor eradication. J Control Release 2025; 377:855-879. [PMID: 39631701 DOI: 10.1016/j.jconrel.2024.11.071] [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: 10/24/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Nanomedicines, by significantly enhancing the solubility, stability, and targeted delivery of therapeutic agents, have emerged as transformative tools in light-induced therapies, particularly in the context of oncology. These advancements are attributed to their ability to mediate autophagy through light activation, thereby revolutionizing cancer treatment paradigms. This review provides a comprehensive analysis of the state-of-the-art integration of nanomedicines with phototherapy techniques, emphasizing their role in modulating autophagy within cancer cells. It delineates the potential of light-responsive nanomaterials to induce selective tumor cell death by precisely regulating over-activated autophagy pathways. Additionally, it discusses innovative strategies for combining nanomedicines with phototherapy and other clinical modalities for tumor treatment, as well as integrating autophagy with various forms of programmed cell death to address challenges related to drug resistance and therapeutic efficacy. By synthesizing recent advancements and delineating future research directions, this review offers a thorough perspective on the optimization of light-induced autophagy through nanomedicines, highlighting novel strategies for enhancing cancer treatment efficacy.
Collapse
Affiliation(s)
- Qihang Ding
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Haiyan Chen
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yifan Zhang
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Junbin Yang
- Hainan Academy of Inspection and Testing, Hainan 570203, PR China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| | - Ling Mei
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
14
|
Yang C, Xue R, Qin C, Huang L, Nie R, Luo Y, Xu S, Tang K, Chen J, Jia L, Tan Q. Celastrol Induces Ferroptosis by Regulating CERKL to Exert Anti-Gastric Cancer Effect. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:931-949. [PMID: 40374372 DOI: 10.1142/s0192415x25500351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Gastric cancer is a significant global health issue. Celastrol, a natural compound, has shown antitumor potential, but its molecular mechanism in gastric cancer remains unclear. In this study, we treated HGC-27 cells with celastrol and employed CCK8, colony formation, and Transwell assays, revealing its inhibitory effect on cell proliferation and migration. Flow cytometry assay results showed that celastrol could elevate the level of reactive oxygen species (ROS) in HGC-27 cells. By using the iron ion and malondialdehyde (MDA) detection kits, it was found that celastrol promoted the accumulation of iron ions (Fe[Formula: see text] in HGC-27 cells, increased the MDA content, and simultaneously decreased the glutathione (GSH) content. Additionally, Western blot analysis indicated that celastrol exerts an inhibitory effect on the expression of ferroptosis-marker proteins GPX4 and SLC7A11. PCR array and further experiments identified CERKL as a key factor, whose downregulation by celastrol was associated with enhanced ferroptosis. In vivo, celastrol inhibited tumor growth without affecting body weight or organ histology. Our findings suggest that celastrol may inhibit gastric cancer via CERKL-regulated ferroptosis, providing a potential therapeutic strategy.
Collapse
Affiliation(s)
- Chang Yang
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Rui Xue
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Chuling Qin
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Lingyue Huang
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Rongrong Nie
- Rehabilitation Department, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Yuqin Luo
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Siyuan Xu
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Ke Tang
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Jianning Chen
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Lulu Jia
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Qinyou Tan
- Department of Clinical Pharmacy, School of Pharmacy, Guilin Medical University, Guilin 541001, Guangxi, China
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi, China
| |
Collapse
|
15
|
Wu L, Pi W, Huang X, Yang L, Zhang X, Lu J, Yao S, Lin X, Tan X, Wang Z, Wang P. Orchestrated metal-coordinated carrier-free celastrol hydrogel intensifies T cell activation and regulates response to immune checkpoint blockade for synergistic chemo-immunotherapy. Biomaterials 2025; 312:122723. [PMID: 39121732 DOI: 10.1016/j.biomaterials.2024.122723] [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: 04/18/2024] [Revised: 07/01/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024]
Abstract
The challenges generated by insufficient T cell activation and infiltration have constrained the application of immunotherapy. Making matters worse, the complex tumor microenvironment (TME), resistance to apoptosis collectively poses obstacles for cancer treatment. The carrier-free small molecular self-assembly strategy is a current research hotspot to overcome these challenges. This strategy can transform multiple functional agents into sustain-released hydrogel without the addition of any excipients. Herein, a coordination and hydrogen bond mediated tricomponent hydrogel (Cel hydrogel) composed of glycyrrhizic acid (GA), copper ions (Cu2+) and celastrol (Cel) was initially constructed. The hydrogel can regulate TME by chemo-dynamic therapy (CDT), which increases reactive oxygen species (ROS) in conjunction with GA and Cel, synergistically expediting cellular apoptosis. What's more, copper induced cuproptosis also contributes to the anti-tumor effect. In terms of regulating immunity, ROS generated by Cel hydrogel can polarize tumor-associated macrophages (TAMs) into M1-TAMs, Cel can induce T cell proliferation as well as activate DC mediated antigen presentation, which subsequently induce T cell proliferation, elevate T cell infiltration and enhance the specific killing of tumor cells, along with the upregulation of PD-L1 expression. Upon co-administration with aPD-L1, this synergy mitigated both primary and metastasis tumors, showing promising clinical translational value.
Collapse
Affiliation(s)
- Linying Wu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Wenmin Pi
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xuemei Huang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Luping Yang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiang Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jihui Lu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Shuchang Yao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiaoyu Lin
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xinru Tan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhixia Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Penglong Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
16
|
Shen P, Zhang L, Jiang X, Yu B, Zhang J. Targeting HMGB1 and Its Interaction with Receptors: Challenges and Future Directions. J Med Chem 2024; 67:21671-21694. [PMID: 39648929 DOI: 10.1021/acs.jmedchem.4c01912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
High mobility group box 1 (HMGB1) is a nonhistone chromatin protein predominantly located in the nucleus. However, under pathological conditions, HMGB1 can translocate from the nucleus to the cytoplasm and subsequently be released into the extracellular space through both active secretion and passive release mechanisms. The distinct cellular locations of HMGB1 facilitate its interaction with various endogenous and exogenous factors, allowing it to perform diverse functions across a range of diseases. This Perspective provides a comprehensive overview of the structure, release mechanisms, and multifaceted roles of HMGB1 in disease contexts. Furthermore, it introduces the development of both small molecule and macromolecule inhibitors targeting HMGB1 and its interaction with receptors. A detailed analysis of the predicted pockets is also presented, aiming to establish a foundation for the future design and development of HMGB1 inhibitors.
Collapse
Affiliation(s)
- Pingping Shen
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Libang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xuewa Jiang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jian Zhang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
17
|
Hoblos R, Khalil K, Karam M, Bazzi S. The role of NF-κB transcription factor in the regulation of cytokine induced thermal hyperalgesia in a Leishmania major model in BALB/c mice. Exp Parasitol 2024; 267:108864. [PMID: 39577554 DOI: 10.1016/j.exppara.2024.108864] [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: 07/24/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Cutaneous leishmaniasis caused mainly by Leishmania major (L. major) is one of the trending models used to investigate induced hyperalgesia and the involved cytokines. Previous studies approached the role of several cytokines in the observed hyperalgesia, but the molecular mechanisms orchestrating such a response still needed to be addressed. In this study, we inspect the role of the NF-κB in the modulation of L. major-prompted hyperalgesia and cytokine expression in BALB/c mice by administering celastrol, a potent blocker of this transcription factor. Intraperitoneal injection of 0.5 mg/kg and 1 mg/kg of celastrol attenuated the L. major-induced thermal hyperalgesia in BALB/c mice for 15 days and 21 days, respectively, as detected by hot plate and tail flick behavioral assessments. Cytokine levels were quantified in the infected paws of BALB/c mice using Sandwich ELISA. The administration of 1 mg/kg celastrol decreased TNF-α levels in L. major infected mice for 23 days, and IL-1β expression declined significantly for 23 days using both celastrol dosages. However, no significant change was observed in the levels of IL-10 in our experimental groups. The activation of NF-κB was detected by observing the phosphorylation levels of the p65 subunit using PathScan phospho-ELISA. The level of NF-κB phosphorylation was elevated in L. major infected BALB/c mice. Only administering 1 mg/kg celastrol suppressed the phosphorylation of p65, thus inactivating NF-kB. In conclusion, our results provide new insights into the correlation between the activation of NF-kB, the induction of thermal hyperalgesia, and the expression of TNF-α and IL-1β in the L. major-induced hyperalgesia model.
Collapse
Affiliation(s)
- Reem Hoblos
- University of Balamand, Faculty of Arts and Sciences, Lebanon
| | - Karl Khalil
- Lebanese American University, School of Medicine, Lebanon.
| | - Marc Karam
- University of Balamand, Faculty of Arts and Sciences, Lebanon
| | - Samer Bazzi
- University of Balamand, Faculty of Arts and Sciences, Lebanon
| |
Collapse
|
18
|
Bu X, Guo H, Gao W, Zhang L, Hou J, Li B, Xia Z, Wang W. Neuroprotection of celastrol against postoperative cognitive dysfunction through dampening cGAS-STING signaling. Exp Neurol 2024; 382:114987. [PMID: 39369806 DOI: 10.1016/j.expneurol.2024.114987] [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: 07/14/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Neuroinflammation is a central player in postoperative cognitive dysfunction (POCD), an intractable and highly confounding neurological complication with finite therapeutic options. Celastrol, a quinone methide triterpenoid, is a bioactive ingredient extracted from Tripterygium wilfordii with talented anti-inflammatory capacity. However, it is unclear whether celastrol can prevent anesthesia/surgery-evoked cognitive deficits in an inflammation-specific manner. The STING agonist 5,6-dimethylxanthenone-4-acetic acid (DMXAA) was used to determine whether celastrol possesses neuroprotection dependent on the STING pathway in vivo and in vitro. Isoflurane and laparotomy triggered cGAS-STING activation, caspase-3/GSDME-dependent pyroptosis, and enhanced Iba-1 immunoreactivity. Celastrol improved cognitive performance and decreased the levels of cGAS, 2'3'-cGAMP, STING, NF-κB phosphorylation, Iba-1, TNF-α, IL-6, and IFN-β. Downregulation of cleaved caspase-3 and N-GSDME was observed in the hippocampus of POCD mice and HT22 cells after celastrol administration, accompanied by limited secretion of pyroptosis-pertinent pro-inflammatory cytokines IL-1β and IL-18. DMXAA neutralized the favorable influences of celastrol on cognitive function, as confirmed by the activation of the STING/caspase-3/GSDME axis. These findings implicate celastrol as a therapeutic agent for POCD through anti-inflammation and anti-pyroptosis.
Collapse
Affiliation(s)
- Xueshan Bu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Hui Guo
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, Hubei Province 430070, China
| | - Wenwei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Lei Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Bixi Li
- Department of Anesthesiology, General Hospital of Central Theater Command of PLA, Wuhan, Hubei Province 430070, China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China.
| | - Wei Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China.
| |
Collapse
|
19
|
He C, Zhou J, Wu X, Zhou Y, Wang S, Liu B, Luo T, Chen Y, Yuan J, Wang D, Zhang C, Shi J. Hyaluronic acid-zein shell-core biopolymer nanoparticles enhance hepatocellular carcinoma therapy of celastrol via CD44-mediated cellular uptake. Int J Biol Macromol 2024; 281:136096. [PMID: 39353524 DOI: 10.1016/j.ijbiomac.2024.136096] [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: 07/27/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Low concentrations or limited residence times in tumor tissues, making celastrol (Cel) difficult to exert significant therapeutic effects. Thus, we developed Zein/hyaluronic acid core-shell nanoparticles (Cel/Zein@HA NPs) for active targeted delivery of Cel via CD44 receptor over-expression on cancer cells, which may strengthen the therapeutic efficacy of Cel and improve delivery targeting. Cel-loaded Zein nanoparticles (core), are elegantly enveloped by a hydrophilic HA coating that forms the shell, resulting in significantly improved encapsulation efficiency and ensured good stability. The cellular uptake of Cel/Zein@HA NPs in HepG2 cells was 1.57-fold higher than nontargeting Cel/Zein NPs. Near-infrared fluorescence imaging confirmed the accumulation of Cel/Zein@HA NPs in H22 liver cancer tumors in mice, resulting in effective antitumor effects and good biosafety. Besides, in vitro and in vivo experiments showed that compared with Cel/Zein NPs, Cel/Zein@HA NPs had more efficient inhibitory effect on tumor proliferation and lower systemic toxicity. Further studies revealed that Cel/Zein@HA NPs induced apoptosis in hepatocellular carcinoma cells by modulating Bax and Bcl-2 expression, while also inhibiting tumor angiogenesis by decreasing CD31 and VEGF levels. Overall, this study presents a promising strategy for enhancing targeted liver cancer therapy through the utilization of biopolymer nanoparticle-based nano-pharmaceuticals that facilitate CD44-mediated cellular uptake.
Collapse
Affiliation(s)
- Congjian He
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Jiahui Zhou
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Xixi Wu
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Yujin Zhou
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Siya Wang
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Bo Liu
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Ting Luo
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Ying Chen
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Jia Yuan
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Di Wang
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| | - Chen Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jinfeng Shi
- College of Pharmacy, Chengdu Medical College, Chengdu 610500, China.
| |
Collapse
|
20
|
Zhao Y, Miettinen K, Kampranis SC. Celastrol: A century-long journey from the isolation to the biotechnological production and the development of an antiobesity drug. CURRENT OPINION IN PLANT BIOLOGY 2024; 81:102615. [PMID: 39128271 DOI: 10.1016/j.pbi.2024.102615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/13/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Celastrol, a triterpenoid found in the root of the traditional medicinal plant Tripterygium wilfordii, is a potent anti-inflammatory and antiobesity agent. However, pharmacological exploitation of celastrol has been hindered by the limited accessibility of plant material, the co-existence of other toxic compounds in the same plant tissue, and the lack of an efficient chemical synthesis method. In this review, we highlight recent progress in elucidating celastrol biosynthesis and discuss how this knowledge can facilitate its scalable bioproduction using cell factories and its further development as an antiobesity and anti-inflammatory drug.
Collapse
Affiliation(s)
- Yong Zhao
- Biochemical Engineering Group, Plant Biochemistry Section, Department of Plant and Environment Sciences, Faculty of Science, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark
| | - Karel Miettinen
- Biochemical Engineering Group, Plant Biochemistry Section, Department of Plant and Environment Sciences, Faculty of Science, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark
| | - Sotirios C Kampranis
- Biochemical Engineering Group, Plant Biochemistry Section, Department of Plant and Environment Sciences, Faculty of Science, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark.
| |
Collapse
|
21
|
Liu M, Li N, Wang Z, Wang S, Ren S, Li X. Synthesis of a celastrol derivative as a cancer stem cell inhibitor through regulation of the STAT3 pathway for treatment of ovarian cancer. RSC Med Chem 2024; 15:d4md00468j. [PMID: 39246745 PMCID: PMC11376026 DOI: 10.1039/d4md00468j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Accumulating evidence suggests that the root of drug chemoresistance in ovarian cancer is tightly associated with subpopulations of cancer stem cells (CSCs), whose activation is largely associated with signal transducer and activator of transcription 3 (STAT3) signaling. Recently, celastrol has shown a significant anti-cancer effect on ovarian cancer, but its clinical translation is very challenging due to its oral bioavailability and high organ toxicity. In this study, a celastrol derivative (Cel-N) was synthesized to augment the overall efficacy, and its underlying mechanisms were also explored. Different ovarian cancer cells, SKOV3 and A2780, were used to evaluate and compare the anticancer effects. Cel-N displayed potent activities against all the tested ovarian cancer cells, with the lowest IC50 value of 0.14-0.25 μM. Further studies showed that Cel-N effectively suppressed the colony formation and sphere formation ability, decreased the percentage of CD44+CD24- and ALDH+ cells, and induced ROS production. Furthermore, western blot analysis indicated that Cel-N significantly inhibited both Tyr705 and Ser727 phosphorylation and reduced the protein expression of STAT3. In addition, Cel-N could dramatically induce apoptosis and cell cycle arrest, and inhibit migration and invasion. Importantly, Cel-N showed a potent antitumor efficacy with no or limited systemic toxicity in mice xenograft models. The anticancer effect of Cel-N is stronger than celastrol. Cel-N attenuates cancer cell stemness, inhibits the STAT3 pathway, and exerts anti-ovarian cancer effects in cell and mouse models. Our data support that Cel-N is a potent drug candidate for ovarian cancer.
Collapse
Affiliation(s)
- Meijuan Liu
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University Liaocheng 252059 China
| | - Na Li
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University Liaocheng 252059 China
| | - Zhaoxue Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University Liaocheng 252059 China
| | - Shuo Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University Liaocheng 252059 China
| | - Shaoda Ren
- Liaocheng Tumor Hospital Liaocheng Shandong 252000 China
| | - Xiaojing Li
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University Liaocheng 252059 China
| |
Collapse
|
22
|
Coronell-Tovar A, Pardo JP, Rodríguez-Romero A, Sosa-Peinado A, Vásquez-Bochm L, Cano-Sánchez P, Álvarez-Añorve LI, González-Andrade M. Protein tyrosine phosphatase 1B (PTP1B) function, structure, and inhibition strategies to develop antidiabetic drugs. FEBS Lett 2024; 598:1811-1838. [PMID: 38724486 DOI: 10.1002/1873-3468.14901] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 08/13/2024]
Abstract
Tyrosine protein phosphatase non-receptor type 1 (PTP1B; also known as protein tyrosine phosphatase 1B) is a member of the protein tyrosine phosphatase (PTP) family and is a soluble enzyme that plays an essential role in different physiological processes, including the regulation of metabolism, specifically in insulin and leptin sensitivity. PTP1B is crucial in the pathogenesis of type 2 diabetes mellitus and obesity. These biological functions have made PTP1B validated as an antidiabetic and anti-obesity, and potentially anticancer, molecular target. Four main approaches aim to inhibit PTP1B: orthosteric, allosteric, bidentate inhibition, and PTPN1 gene silencing. Developing a potent and selective PTP1B inhibitor is still challenging due to the enzyme's ubiquitous expression, subcellular location, and structural properties. This article reviews the main advances in the study of PTP1B since it was first isolated in 1988, as well as recent contextual information related to the PTP family to which this protein belongs. Furthermore, we offer an overview of the role of PTP1B in diabetes and obesity, and the challenges to developing selective, effective, potent, bioavailable, and cell-permeable compounds that can inhibit the enzyme.
Collapse
Affiliation(s)
- Andrea Coronell-Tovar
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Juan P Pardo
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | - Alejandro Sosa-Peinado
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Luz Vásquez-Bochm
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Patricia Cano-Sánchez
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Laura Iliana Álvarez-Añorve
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Martin González-Andrade
- Laboratorio de Biosensores y Modelaje molecular, Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
23
|
Li H, Li Y, Zhang L, Wang N, Lu D, Tang D, Lv Y, Zhang J, Yan H, Gong H, Zhang M, Nie K, Hou Y, Yu Y, Xiao H, Liu C. Prodrug-inspired adenosine triphosphate-activatable celastrol-Fe(III) chelate for cancer therapy. SCIENCE ADVANCES 2024; 10:eadn0960. [PMID: 38996025 PMCID: PMC11244545 DOI: 10.1126/sciadv.adn0960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 06/06/2024] [Indexed: 07/14/2024]
Abstract
Celastrol (CEL), an active compound isolated from the root of Tripterygium wilfordii, exhibits broad anticancer activities. However, its poor stability, narrow therapeutic window and numerous adverse effects limit its applications in vivo. In this study, an adenosine triphosphate (ATP) activatable CEL-Fe(III) chelate was designed, synthesized, and then encapsulated with a reactive oxygen species (ROS)-responsive polymer to obtain CEL-Fe nanoparticles (CEL-Fe NPs). In normal tissues, CEL-Fe NPs maintain structural stability and exhibit reduced systemic toxicity, while at the tumor site, an ATP-ROS-rich tumor microenvironment, drug release is triggered by ROS, and antitumor potency is restored by competitive binding of ATP. This intelligent CEL delivery system improves the biosafety and bioavailability of CEL for cancer therapy. Such a CEL-metal chelate strategy not only mitigates the challenges associated with CEL but also opens avenues for the generation of CEL derivatives, thereby expanding the therapeutic potential of CEL in clinical settings.
Collapse
Affiliation(s)
- Hanrong Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yifan Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lingpu Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physical and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
| | - Nan Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Dong Lu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physical and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
- University of Chinese Academy of Science, Beijing 100049, China
| | - Yitong Lv
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jinbo Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Heben Yan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - He Gong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing 102206,China
| | - Kaili Nie
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Science, State Key Laboratory of Polymer Physical and Chemistry, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
- University of Chinese Academy of Science, Beijing 100049, China
| | - Chaoyong Liu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
24
|
Guo Y, Peng X, Liu F, Zhang Q, Ding L, Li G, Qiu F. Potential of natural products in inflammation: biological activities, structure-activity relationships, and mechanistic targets. Arch Pharm Res 2024; 47:377-409. [PMID: 38739203 DOI: 10.1007/s12272-024-01496-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 04/23/2024] [Indexed: 05/14/2024]
Abstract
A balance between the development and suppression of inflammation can always be found in the body. When this balance is disturbed, a strong inflammatory response can damage the body. It sometimes is necessary to use drugs with a significant anti-inflammatory effect, such as nonsteroidal anti-inflammatory drugs and steroid hormones, to control inflammation in the body. However, the existing anti-inflammatory drugs have many adverse effects, which can be deadly in severe cases, making research into new safer and more effective anti-inflammatory drugs necessary. Currently, numerous types of natural products with anti-inflammatory activity and distinct structural features are available, and these natural products have great potential for the development of novel anti-inflammatory drugs. This review summarizes 260 natural products and their derivatives with anti-inflammatory activities in the last two decades, classified by their active ingredients, and focuses on their structure-activity relationships in anti-inflammation to lay the foundation for subsequent new drug development. We also elucidate the mechanisms and pathways of natural products that exert anti-inflammatory effects via network pharmacology predictions, providing direction for identifying subsequent targets of anti-inflammatory natural products.
Collapse
Affiliation(s)
- Yajing Guo
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Xuling Peng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Fanfei Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Qi Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Liqin Ding
- State Key Laboratory of Component-Based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Gen Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China.
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China.
- State Key Laboratory of Component-Based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China.
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
25
|
Lai Y, Guo S, Tang Q, Chang G, Zhang H, Li B, Feng Q, Hu K, Xu Z, Gao X, Zhang Q, Yi H, Song D, Zhang Y, Peng Y, Cai H, Zhu W, Shi J. Dihydrocelastrol induces cell death and suppresses angiogenesis through BCR/AP-1/junb signalling in diffuse large B cell lymphoma. Arch Biochem Biophys 2024; 754:109929. [PMID: 38367794 DOI: 10.1016/j.abb.2024.109929] [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: 12/07/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 02/19/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma. Although treatment options have improved, a large proportion of patients show low survival rates, highlighting an urgent need for novel therapeutic strategies. The aim of this study was to investigate the efficacy of the new small-molecule compound dihydrocelastrol (DHCE), acquired through the structural modification of celastrol (CE), in the treatment of DLBCL. DHCE showed potent anti-lymphoma efficacy and synergistic effects with doxorubicin. DHCE triggered DLBCL cell apoptosis and G0/G1-phase blockade, thereby hindering angiogenesis. DHCE inhibited B-cell receptor cascade signalling and Jun B and p65 nuclear translocation, thereby suppressing pro-tumourigenic signalling. Finally, DHCE exerted lower toxicity than CE, which showed severe hepatic, renal, and reproductive toxicity in vivo. Our findings support further investigation of the clinical efficacy of DHCE against DLBCL.
Collapse
Affiliation(s)
- Yue Lai
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Shushan Guo
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qiongwei Tang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Gaomei Chang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hui Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Bo Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qilin Feng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ke Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xuejie Gao
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qikai Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hongfei Yi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Dongliang Song
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yifei Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Peng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Haiyan Cai
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
26
|
Pan W, Yang B, He D, Chen L, Fu C. Functions and targets of miRNAs in pharmacological and toxicological effects of major components of Tripterygium wilfordii Hook F. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1997-2019. [PMID: 37831113 DOI: 10.1007/s00210-023-02764-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
Tripterygium wilfordii Hook F (TwHF) has a long history of use as a traditional Chinese medicine and has been widely administered to treat various inflammatory and autoimmune diseases. MicroRNAs (miRNAs) are endogenous, short, non-coding RNAs that regulate gene expression post-transcriptionally. They participate in the efficacies and even toxicities of the components of TwHF, rendering miRNAs an appealing therapeutic strategy. This review summarizes the recent literature related to the roles and mechanisms of miRNAs in the pharmacological and toxicological effects of main components of TwHF, focusing on two active compounds, triptolide (TP) and celastrol (CEL). Additionally, the prospects for the "You Gu Wu Yun" theory regarding TwHF nephrotoxicity are presented.
Collapse
Affiliation(s)
- Wei Pan
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Bo Yang
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Dongxiu He
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
| | - Chengxiao Fu
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China.
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
27
|
Zhang X, Chen Y, Li X, Xu H, Yang J, Wang C, Zhang C, Deng Y, Lu A, Zheng C, Lu J. Carrier-free self-assembled nanomedicine based on celastrol and galactose for targeting therapy of hepatocellular carcinoma via inducing ferroptosis. Eur J Med Chem 2024; 267:116183. [PMID: 38354520 DOI: 10.1016/j.ejmech.2024.116183] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024]
Abstract
Triggering ferroptosis is a potential therapeutic pathway and strategy for the prospective treatment of lethal hepatocellular carcinoma (HCC). The asialo-glycoprotein receptor (ASGPR) is an over-expressed receptor on the membranes of hepatocellular carcinoma cells (HCCs) and binds specifically to galactose (Gal) ligand. Celastrol (CE) is a potent anticancer natural product, but its poor water solubility and severe toxicity restrict its clinical application. In this study, a carrier-free self-assembled nanoparticles, CE-Gal-NPs, were designed and prepared by nanoprecipitation method, which could recognize ASGPR receptor by active targeting (Gal ligand) and passive targeting (EPR effect), access to the cell through the clathrin pathway and finally internalize to lysosomes. CE-Gal-NPs triggered reactive oxygen species (ROS)-mediated ferroptosis pathway and exerted anti-HCC effects in vitro and in vivo by down-regulating GPX4 and up-regulating COX-2 expression, depleting glutathione (GSH) levels, and increasing lipid peroxidation levels in cells and tumor tissues. In the H22 xenograft mouse model, the CE-Gal-NPs group exhibited dramatically superior tumor inhibition than the CE group, while Gal conjugating diminished the systemic toxicity of CE. Consequently, this study presented a promising strategy for CE potentiation and toxicity reduction, as well as a potential guideline for the development of clinically targeted therapeutic agents for HCC.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yao Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiao Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hong Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jirui Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chuanqi Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chaozheng Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yun Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone&Joint-Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
| | - Chuan Zheng
- Sichuan Key Laboratory of TCM Regulating Metabolic Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Jun Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Institute for Advancing Translational Medicine in Bone&Joint-Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
| |
Collapse
|
28
|
Zhang X, Xu Y, Fan M, Lv X, Long J, Yang R, Zhang R, Liu Z, Gu J, Wu P, Wang C. Ponicidin-induced conformational changes of HSP90 regulates the MAPK pathway to relieve ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117483. [PMID: 38008273 DOI: 10.1016/j.jep.2023.117483] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/05/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is a recurring chronic intestinal disease that can be debilitating and in severe cases, may further lead to cancer. However, all these treatment techniques still suffer from drug dependence, adverse effects and poor patient compliance. Therefore, there is an urgent need to seek new therapeutic strategies. In traditional Chinese medicine, Rabdosia rubescens (Hemsl.) H.Hara has the effects of clearing heat-toxin and promoting blood circulation to relieve pain, it is wildly used for treating inflammatory diseases such as sore throats and tonsillitis. Ponicidin is an important molecule for the anti-inflammatory effects of Rabdosia rubescens, but it has not been studied in the treatment of colitis. HSP90 is the most critical regulator in the development and progression of inflammatory diseases such as UC. AIM OF THE STUDY The aim of this study was to explore the anti-inflammatory activity of ponicidin and its mechanism of effect in vitro and in vivo, as well as to identify the target proteins on which ponicidin may interact. MATERIAL AND METHODS 2.5% (w/v) dextran sulfate sodium (DSS) was used to induce C57BL/6 mice to form an ulcerative colitis model, and then 5 mg/kg and 10 mg/kg ponicidin was given for treatment, while the Rabdosia rubescens extract group and Rabdosia rubescens diterpene extract group were set up for comparison of the efficacy of ponicidin. At the end of modeling and drug administration, mouse colon tissues were taken, and the length of colon was counted, inflammatory factors and inflammatory signaling pathways were detected. RAW264.7 cells were induced to form cell inflammation model with 1 μg/mL Lipopolysaccharide (LPS) for 24 h. 1 μM, 2 μM and 4 μM ponicidin were given at the same time, and after the end of the modeling and administration of the drug, the inflammatory factors and inflammatory signaling pathways were detected by qRT-PCR, western blotting, immunofluorescence and other methods. In vitro, target angling and combined with mass spectrometry were used to search for relevant targets of ponicidin, while isothermal titration calorimetry (ITC), protease degradation experiments and molecular dynamics simulations were used for further confirmation of the mode of action and site of action between ponicidin and target proteins. RESULTS Ponicidin can alleviate DSS and LPS-induced inflammation by inhibiting the MAPK signaling pathway at the cellular and animal levels. In vitro, we confirmed that ponicidin can interact with the middle domain of HSP90 and induce the conformational changes in the N-terminal domain. CONCLUSION These innovative efforts identified the molecular target of ponicidin in the treatment of UC and revealed the molecular mechanism of its interaction with HSP90.
Collapse
Affiliation(s)
- Xuerong Zhang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yuanhang Xu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Minqi Fan
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Xueqing Lv
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jiachan Long
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Rong Yang
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Rong Zhang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Jiangyong Gu
- Research Center of Integrative Medicine, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Peng Wu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Caiyan Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
29
|
Gao P, Wang J, Tang H, Pang H, Liu J, Wang C, Xia F, Chen H, Xu L, Zhang J, Yuan L, Han G, Wang J, Liu G. Chemoproteomics-based profiling reveals potential antimalarial mechanism of Celastrol by disrupting spermidine and protein synthesis. Cell Commun Signal 2024; 22:139. [PMID: 38378659 PMCID: PMC10877925 DOI: 10.1186/s12964-023-01409-5] [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: 10/12/2023] [Accepted: 11/24/2023] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Malaria remains a global health burden, and the emergence and increasing spread of drug resistance to current antimalarials poses a major challenge to malaria control. There is an urgent need to find new drugs or strategies to alleviate this predicament. Celastrol (Cel) is an extensively studied natural bioactive compound that has shown potentially promising antimalarial activity, but its antimalarial mechanism remains largely elusive. METHODS We first established the Plasmodium berghei ANKA-infected C57BL/6 mouse model and systematically evaluated the antimalarial effects of Cel in conjunction with in vitro culture of Plasmodium falciparum. The potential antimalarial targets of Cel were then identified using a Cel activity probe based on the activity-based protein profiling (ABPP) technology. Subsequently, the antimalarial mechanism was analyzed by integrating with proteomics and transcriptomics. The binding of Cel to the identified key target proteins was verified by a series of biochemical experiments and functional assays. RESULTS The results of the pharmacodynamic assay showed that Cel has favorable antimalarial activity both in vivo and in vitro. The ABPP-based target profiling showed that Cel can bind to a number of proteins in the parasite. Among the 31 identified potential target proteins of Cel, PfSpdsyn and PfEGF1-α were verified to be two critical target proteins, suggesting the role of Cel in interfering with the de novo synthesis of spermidine and proteins of the parasite, thus exerting its antimalarial effects. CONCLUSIONS In conclusion, this study reports for the first time the potential antimalarial targets and mechanism of action of Cel using the ABPP strategy. Our work not only support the expansion of Cel as a potential antimalarial agent or adjuvant, but also establishes the necessary theoretical basis for the development of potential antimalarial drugs with pentacyclic triterpenoid structures, as represented by Cel. Video Abstract.
Collapse
Affiliation(s)
- Peng Gao
- Department of rehabilitation medicine, Shunde Hospital, Southern Medical University, Foshan, 528300, China
| | - Jianyou Wang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Huan Tang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huanhuan Pang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiemei Liu
- Department of rehabilitation medicine, Shunde Hospital, Southern Medical University, Foshan, 528300, China
| | - Chen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Fei Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Honglin Chen
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Liting Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lixia Yuan
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China.
| | - Guang Han
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China.
| | - Jigang Wang
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China.
| | - Gang Liu
- Department of rehabilitation medicine, Shunde Hospital, Southern Medical University, Foshan, 528300, China.
| |
Collapse
|
30
|
Fadaei MS, Fadaei MR, Kheirieh AE, Rahmanian-Devin P, Dabbaghi MM, Nazari Tavallaei K, Shafaghi A, Hatami H, Baradaran Rahimi V, Nokhodchi A, Askari VR. Niosome as a promising tool for increasing the effectiveness of anti-inflammatory compounds. EXCLI JOURNAL 2024; 23:212-263. [PMID: 38487088 PMCID: PMC10938253 DOI: 10.17179/excli2023-6868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/16/2024] [Indexed: 03/17/2024]
Abstract
Niosomes are drug delivery systems with widespread applications in pharmaceutical research and the cosmetic industry. Niosomes are vesicles of one or more bilayers made of non-ionic surfactants, cholesterol, and charge inducers. Because of their bilayer characteristics, similar to liposomes, niosomes can be loaded with lipophilic and hydrophilic cargos. Therefore, they are more stable and cheaper in preparation than liposomes. They can be classified into four categories according to their sizes and structures, namely small unilamellar vesicles (SUVs), large unilamellar vesicles (LUVs,), multilamellar vesicles (MLVs), and multivesicular vesicles (MVVs). There are many methods for niosome preparation, such as thin-film hydration, solvent injection, and heating method. The current study focuses on the preparation methods and pharmacological effects of niosomes loaded with natural and chemical anti-inflammatory compounds in kinds of literature during the past decade. We found that most research was carried out to load anti-inflammatory agents like non-steroidal anti-inflammatory drugs (NSAIDs) into niosome vesicles. The studies revealed that niosomes could improve anti-inflammatory agents' physicochemical properties, including solubility, cellular uptake, stability, encapsulation, drug release and liberation, efficiency, and oral bioavailability or topical absorption. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Mohammad Saleh Fadaei
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Emad Kheirieh
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Rahmanian-Devin
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Abouzar Shafaghi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hooman Hatami
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Nokhodchi
- Lupin Pharmaceutical Research Center, 4006 NW 124th Ave., Coral Springs, Florida, FL 33065, USA
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Brighton BN1 9QJ, UK
| | - Vahid Reza Askari
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
31
|
Long C, He P, Tu R, Song X, Li H, Huang W, Liu J, Zhang L, Guo Y. Subchronic toxicity evaluation of Huobahuagen extract and plasma metabolic profiling analysis combined with conventional pathology methods. J Appl Toxicol 2024; 44:201-215. [PMID: 37697829 DOI: 10.1002/jat.4532] [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: 07/02/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 09/13/2023]
Abstract
Huobahua, namely, Tripterygium hypoglaucum (Levl.) Hutch, known as a traditional Chinese herbal medicine, especially its underground parts, has been widely developed into several Tripterygium agents for the treatment of rheumatoid arthritis and other autoimmune diseases. It has sparked wide public concern about its safety, such as multi-organ toxicity. However, the toxic characteristics and damage mechanism of Huobahuagen extract (HBHGE) remain unclear. In the present study, subchronic oral toxicity study of HBHGE (10.0 g crude drug/kg/day for 12 weeks) was performed in male rats. Hematological, serum biochemical, and histopathological parameters, urinalysis, and plasma metabolic profiling were assessed. The single-dose subchronic toxicity results related to HBHGE exhibited obvious toxicity to the testis and epididymis of male rats. Furthermore, plasma metabolomics analysis suggested that a series of metabolic disorders were induced by oral administration of HBHGE, mainly focusing on amino acid (glutamate, phenylalanine, and tryptophan) metabolisms, pyrimidine metabolism, glutathione metabolism, and steroid hormone biosynthesis. Moreover, it appeared that serum testosterone in male rats treated with HBHGE for 12 weeks, decreased significantly, and was susceptible to the toxic effects of HBHGE. Taken together, conventional pathology and plasma metabolomics for preliminarily exploring subchronic toxicity and underlying mechanism can provide useful information about the reduction of toxic risks from HBHGE and new insights into the development of detoxification preparations.
Collapse
Affiliation(s)
- Chengyan Long
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Peilin He
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Ruxia Tu
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiaoxian Song
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Henghua Li
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Wentao Huang
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Jianyi Liu
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Li Zhang
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Yanlei Guo
- Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Safety Evaluation of Drugs, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| |
Collapse
|
32
|
Hu SJ, Cheng G, Chen GC, Zhou H, Zhang Q, Zhao QM, Lian CX, Zhao ZH, Zhang QL, Han T, Zhang QY, Qin LP. Cannabinoid receptors type 2: Function and development in agonist discovery from synthetic and natural sources with applications for the therapy of osteoporosis. ARAB J CHEM 2024; 17:105536. [DOI: 10.1016/j.arabjc.2023.105536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
33
|
Zhao XY, Wang JQ, Neely GG, Shi YC, Wang QP. Natural compounds as obesity pharmacotherapies. Phytother Res 2024; 38:797-838. [PMID: 38083970 DOI: 10.1002/ptr.8083] [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: 08/05/2023] [Revised: 10/20/2023] [Accepted: 11/22/2023] [Indexed: 02/15/2024]
Abstract
Obesity has become a serious global public health problem, affecting over 988 million people worldwide. Nevertheless, current pharmacotherapies have proven inadequate. Natural compounds have garnered significant attention due to their potential antiobesity effects. Over the past three decades, ca. 50 natural compounds have been evaluated for the preventive and/or therapeutic effects on obesity in animals and humans. However, variations in the antiobesity efficacies among these natural compounds have been substantial, owing to differences in experimental designs, including variations in animal models, dosages, treatment durations, and administration methods. The feasibility of employing these natural compounds as pharmacotherapies for obesity remained uncertain. In this review, we systematically summarized the antiobesity efficacy and mechanisms of action of each natural compound in animal models. This comprehensive review furnishes valuable insights for the development of antiobesity medications based on natural compounds.
Collapse
Affiliation(s)
- Xin-Yuan Zhao
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ji-Qiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - G Gregory Neely
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Yan-Chuan Shi
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Qiao-Ping Wang
- Laboratory of Metabolism and Aging, School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
- Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
34
|
Bai Y, Liang C, Gao L, Han T, Wang F, Liu Y, Zhou J, Guo J, Wu J, Hu D. Celastrol Pyrazine Derivative Alleviates Silicosis Progression via Inducing ROS-Mediated Apoptosis in Activated Fibroblasts. Molecules 2024; 29:538. [PMID: 38276616 PMCID: PMC10820882 DOI: 10.3390/molecules29020538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/27/2023] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Silicosis is a complex occupational disease without recognized effective treatment. Celastrol, a natural product, has shown antioxidant, anti-inflammatory, and anti-fibrotic activities, but the narrow therapeutic window and high toxicity severely limit its clinical application. Through structural optimization, we have identified a highly efficient and low-toxicity celastrol derivative, CEL-07. In this study, we systematically investigated the therapeutic potential and underlying mechanisms of CEL-07 in silicosis fibrosis. By constructing a silicosis mouse model and analyzing with HE, Masson, Sirius Red, and immunohistochemical staining, CEL-07 significantly prevented the progress of inflammation and fibrosis, and it effectively improved the lung respiratory function of silicosis mice. Additionally, CEL-07 markedly suppressed the expression of inflammatory factors (IL-6, IL-1α, TNF-α, and TNF-β) and fibrotic factors (α-SMA, collagen I, and collagen III), and promoted apoptosis of fibroblasts by increasing ROS accumulation. Moreover, bioinformatics analysis combined with experimental validation revealed that CEL-07 inhibited the pathways associated with inflammation (PI3K-AKT and JAK2-STAT3) and the expression of apoptosis-related proteins. Overall, these results suggest that CEL-07 may serve as a potential candidate for the treatment of silicosis.
Collapse
Affiliation(s)
- Ying Bai
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Chao Liang
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Lu Gao
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Tao Han
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Fengxuan Wang
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
| | - Jing Wu
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan 232001, China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
| | - Dong Hu
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China; (Y.B.); (C.L.); (L.G.); (T.H.); (F.W.); (Y.L.); (J.Z.); (J.G.)
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan 232001, China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institute, Huainan 232001, China
- Key Laboratory of Industrial Dust Prevention and Control and Occupational Safety and Health Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
| |
Collapse
|
35
|
Liu S, Liu M, Xiu J, Zhang T, Zhang B, Cun D, Yang C, Li K, Zhang J, Zhao X. Celastrol-loaded bovine serum albumin nanoparticles target inflamed neutrophils for improved rheumatoid arthritis therapy. Acta Biomater 2024; 174:345-357. [PMID: 38013018 DOI: 10.1016/j.actbio.2023.11.028] [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: 07/30/2023] [Revised: 10/25/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Inflammatory neutrophils (INEs), motivated by cytokines, continue to migrate into the inflamed joints, driving the development of RA. Hence, inducing apoptosis of INEs to reduce recruitment at inflamed joints is an effective strategy for the treatment of RA. However, simply apoptotic INEs may trigger the release of neutrophil extracellular traps (NETs) and accelerate the inflammatory process. To overcome these drawbacks, an RGD-modified bovine serum albumin (BSA) nanoparticles (CBR NPs) was fabricated to selectively target INEs in situ for intracellular delivery of CLT. Studies have demonstrated that CBR NPs can selectively target circulating INEs and induce INEs apoptosis. Meanwhile, CBR NPs inhibited the activation of NETs via NF-κB pathway and the release of Cit-H3 thereby blocking the release process of NETs. In collagen-induced arthritis (CIA) mouse model, CBR NPs suppressed the inflammatory response, and reduced the toxic effects of CLT. In summary, this study shed light on an innovative approach to treat RA by inducing apoptosis of circulating INEs and inhibiting NETs. STATEMENT OF SIGNIFICANCE: RGD-modified bovine serum albumin (BSA) nanoparticles for delivering celastrol, abbreviated as CBR NPs, were constructed to inhibit the infiltration of circulating inflammatory neutrophils (INEs) into inflamed joints while inhibiting the release of NETs to alleviate tissue damage. CBR NPs were prepared for the first time to induce apoptosis of INEs; CBR NPs could inhibit the release of NETs while inducing apoptosis of INEs in vivo and vitro cellular experiments; CBR NPs had favorable anti-inflammatory effects and low toxicity side-effects in collagen-induced arthritis (CIA) mouse models. The application of nanotechnology to induce apoptosis of INEs while inhibiting the release of NETs was a promising approach for the treatment of RA.
Collapse
Affiliation(s)
- Siyi Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Min Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Jingya Xiu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Tian Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Bowen Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Dongyun Cun
- Department of Hepatopancreatobiliary Surgery, The Second Afliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Chunrong Yang
- Department of Pharmacy, Shantou University Medical College, Shantou 515000, PR China
| | - Kexin Li
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China
| | - Jiulong Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China.
| | - Xiuli Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110000, PR China.
| |
Collapse
|
36
|
Wilczak M, Surman M, Przybyło M. The Role of Intracellular and Extracellular Vesicles in the Development of Therapy Resistance in Cancer. Curr Pharm Des 2024; 30:2765-2784. [PMID: 39113303 DOI: 10.2174/0113816128326325240723051625] [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: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 10/22/2024]
Abstract
Cancer is the second leading cause of global mortality and claims approximately 10 million lives annually. Despite advances in treatments such as surgery, chemotherapy, and immunotherapy, resistance to these methods has emerged. Multidrug resistance (MDR), where cancer cells resist diverse treatments, undermines therapy effectiveness, escalating mortality rates. MDR mechanisms include, among others, drug inactivation, reduced drug uptake, enhanced DNA repair, and activation of survival pathways such as autophagy. Moreover, MDR mechanisms can confer resistance to other therapies like radiotherapy. Understanding these mechanisms is crucial for improving treatment efficacy and identifying new therapeutic targets. Extracellular vesicles (EVs) have gathered attention for their role in cancer progression, including MDR development through protein transfer and genetic reprogramming. Autophagy, a process balancing cellular resources, also influences MDR. The intersection of EVs and autophagy further complicates the understanding of MDR. Both extracellular (exosomes, microvesicles) and intracellular (autophagic) vesicles contribute to therapy resistance by regulating the tumor microenvironment, facilitating cell communication, and modulating drug processing. While much is known about these pathways, there is still a need to explore their potential for predicting treatment responses and understanding tumor heterogeneity.
Collapse
Affiliation(s)
- Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
37
|
Kazakova O, Ma X, Tretyakova E, Smirnova I, Slita A, Sinegubova E, Zarubaev V, Jin H, Zhou D, Xiao S. Evaluation of A-ring hydroxymethylene-amino- triterpenoids as inhibitors of SARS-CoV-2 spike pseudovirus and influenza H1N1. J Antibiot (Tokyo) 2024; 77:39-49. [PMID: 38001284 DOI: 10.1038/s41429-023-00677-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 10/24/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023]
Abstract
A set of triterpene A-ring hydroxymethylene-amino-derivatives was synthesized and their antiviral activity was studied. The synthesized compounds were tested for their potential inhibition of SARS-CoV-2 pseudovirus in BHK-21-hACE2 cells and influenza A/PuertoRico/8/34 (H1N1) virus in MDCK cell culture. Compounds 6, 8 and 19 showed significant anti-SARS-CoV-2 pseudovirus activity with EC50 value of 3.20-11.13 µM, which is comparable to the positive control amodiaquine (EC50 3.17 µM). Among them, 28-O-imidazolyl-azepano-betulin 6 and C3-hydroxymethylene-amino-glycyrrhetol-11,13-diene 19 were identified as the lead compounds with SI values of 7 and 10. The binding mode of compound 6 into the RBD domain of SARS-CoV-2 spike glycoprotein (PDB code: 7DK3) by docking and molecular dynamics simulation was investigated.
Collapse
Affiliation(s)
- Oxana Kazakova
- Ufa Institute of Chemistry UFRC RAS, pr. Oktyabrya 71, 450054, Ufa, Russia.
| | - Xinyuan Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Elena Tretyakova
- Ufa Institute of Chemistry UFRC RAS, pr. Oktyabrya 71, 450054, Ufa, Russia
| | - Irina Smirnova
- Ufa Institute of Chemistry UFRC RAS, pr. Oktyabrya 71, 450054, Ufa, Russia
| | - Alexander Slita
- Department of Virology, St. Petersburg Pasteur Institute of Epidemiology and Microbiology, Experimental Virology Laboratory, 14 Mira St., St. Petersburg, 197001, Russia
| | - Ekaterina Sinegubova
- Department of Virology, St. Petersburg Pasteur Institute of Epidemiology and Microbiology, Experimental Virology Laboratory, 14 Mira St., St. Petersburg, 197001, Russia
| | - Vladimir Zarubaev
- Department of Virology, St. Petersburg Pasteur Institute of Epidemiology and Microbiology, Experimental Virology Laboratory, 14 Mira St., St. Petersburg, 197001, Russia
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Sulong Xiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
38
|
Zhao Z, Pei X, Li Q, Zhang H, Wang Y, Qin J, He Y. Pectin-based double network hydrogels as local depots of celastrol for enhanced antitumor therapy. Int J Biol Macromol 2024; 256:128442. [PMID: 38035968 DOI: 10.1016/j.ijbiomac.2023.128442] [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: 07/31/2023] [Revised: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023]
Abstract
In this study, A double-network (DN) hydrogel composed of a physical glycyrrhizic acid (GA) network and a chemically crosslinked pectin-based network was fabricated as a local depot of celastrol (CEL) for cancer treatment. The obtained DN hydrogel possessed excellent mechanical performance, flexibility, biocompatibility, biodegradability and self-healing property. Furthermore, the release profile of CEL loaded DN hydrogel maintained a controlled and sustained release of CEL for a prolonged period. Finally, in vivo animal experiments demonstrated that the DN hydrogel could significantly enhance the therapeutic efficiency of CEL in CT-26 tumor-bearing mice upon intratumoral injection while effectively alleviate the toxicity of the CEL. In summary, this injectable pectin-based double network hydrogels are ideal delivery vehicle for tumor therapy.
Collapse
Affiliation(s)
- Zihao Zhao
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050200, China
| | - Xiaocui Pei
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050200, China
| | - Qiushuai Li
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050200, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Yong Wang
- Key Laboratory of Pathogenesis mechanism and control of inflammatory-autoimmune diseases in Hebei Province, Hebei University, Baoding City, Hebei Province 071002, China
| | - Jianglei Qin
- College of Chemistry and Environmental Science, Hebei University, Baoding City, Hebei Province 071002, China.
| | - Yingna He
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050200, China; Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Shijiazhuang, Hebei Province 050200, China.
| |
Collapse
|
39
|
Zhao Y, Yao Y, Fan S, Shen X, Chai X, Li Z, Zeng J, Pi J, Zhou Z, Huang G, Jin H. Oral delivery of pH-sensitive nanoparticles loaded Celastrol targeting the inflammatory colons to treat ulcerative colitis. J Tissue Eng 2024; 15:20417314241265892. [PMID: 39130681 PMCID: PMC11316965 DOI: 10.1177/20417314241265892] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/18/2024] [Indexed: 08/13/2024] Open
Abstract
The incidence of ulcerative colitis (UC) is rapidly rising worldwide. Oral drug delivery system is a promising approach for treating UC, but it often fails to accumulate to the inflammatory lesions, thus, it is impressive to develop a colon-targeted oral delivery system for preventing systemic toxicity and maintaining UC therapeutics. Here, a negative-charged PLGA nanoparticle system was designed to encapsulate celastrol (Cel), and then chitosan and mannose were coated on the surface of the nanoparticles (MC@Cel-NPs) to endow these nanoparticles with the mucosal adsorption and macrophage targeting abilities. MC@Cel-NPs demonstrate excellent resist decomposition ability against the strong acidic gastrointestinal environment, and accumulates in the specific inflammatory sites through the affinity of electrostatic reaction. After releasing the payload, MC@Cel-NPs could remarkably alleviate the colon inflammation, which was evidenced by the decrease in pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 in both blood and colon sections, and scavenging reactive oxygen species (ROS) in colon cells, including macrophage, neutrophil, T cell, and B cell. This nanoparticle system provided a new approach for treating UC through a Chinese herbal ingredient-related oral delivery manner.
Collapse
Affiliation(s)
- Yue Zhao
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Yinlian Yao
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Shilong Fan
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Xin Shen
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Xingxing Chai
- Laboratory Animal Center, Guangdong Medical University, Dongguan, China
| | - Zimin Li
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Jiachun Zeng
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Zhikun Zhou
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Hua Jin
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
40
|
Vilaboa N, Voellmy R. Withaferin A and Celastrol Overwhelm Proteostasis. Int J Mol Sci 2023; 25:367. [PMID: 38203539 PMCID: PMC10779417 DOI: 10.3390/ijms25010367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Withaferin A (WA) and celastrol (CEL) are major bioactive components of plants that have been widely employed in traditional medicine. The pleiotropic activities of plant preparations and the isolated compounds in vitro and in vivo have been documented in hundreds of studies. Both WA and CEL were shown to have anticancer activity. Although WA and CEL belong to different chemical classes, our synthesis of the available information suggests that the compounds share basic mechanisms of action. Both WA and CEL bind covalently to numerous proteins, causing the partial unfolding of some of these proteins and of many bystander proteins. The resulting proteotoxic stress, when excessive, leads to cell death. Both WA and CEL trigger the activation of the unfolded protein response (UPR) which, if the proteotoxic stress persists, results in apoptosis mediated by the PERK/eIF-2/ATF4/CHOP pathway or another UPR-dependent pathway. Other mechanisms of cell death may play contributory or even dominant roles depending on cell type. As shown in a proteomic study with WA, the compounds appear to function largely as electrophilic reactants, indiscriminately modifying reachable nucleophilic amino acid side chains of proteins. However, a remarkable degree of target specificity is imparted by the cellular context.
Collapse
Affiliation(s)
- Nuria Vilaboa
- Hospital Universitario La Paz-IdiPAZ, 28046 Madrid, Spain
- CIBER de Bioingenieria, Biomateriales y Nanomedicina, CIBER-BBN, 28046 Madrid, Spain
| | | |
Collapse
|
41
|
Jiang X, Yuan C, Ding R, Lu D, Peng X, Dong Z, Zhu C, Lin Y, Wu C, Xie Q. Toxic metabolites and metabolic soft spots of celastrol based on glutathione metabolic capture and high-resolution mass spectrometry. Expert Opin Drug Metab Toxicol 2023; 19:1023-1032. [PMID: 38145500 DOI: 10.1080/17425255.2023.2294042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/06/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND Celastrol is known as one of the most medicinally valuable compounds. However, the pharmaceutical application of celastrol is significantly limited due to high toxicity, while there are few reports on the mechanism of toxicity. METHODS This study searched for possible toxic metabolites through phase I in vitro metabolism and glutathione capture experiments. Then in vivo metabolism experiments in mice and rats were conducted to look for metabolites in vivo. Finally, mice in vivo toxicity experiment was conducted to verify the toxicity of different doses of celastrol to mice. RESULTS In the in vivo and in vitro metabolism experiments, we found 7 phase I metabolites in vitro, 9 glutathione conjugation metabolites in vitro, and 20 metabolites in vivo. The metabolic soft points of celastrol could be the quinone methyl structure at C3-OH and C6. In vivo toxicity experiments show that celastrol causes weight loss, diarrhea, gastrointestinal tract and liver inflammation in mice. CONCLUSIONS This study analyzed the metabolites and possible metabolic soft spots of celastrol, and its hepatotoxicity and gastrointestinal toxicity were demonstrated through in vivo studies for the first time. The results might provide an important basis for potential structural modification to increase the druggability of celastrol.
Collapse
Affiliation(s)
- Xiaojuan Jiang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Caixia Yuan
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Rong Ding
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Di Lu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xiaoyu Peng
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Zhihao Dong
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chunyan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yihua Lin
- Department of Respiratory and Critical Care Medicine, The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cell Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qiang Xie
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
42
|
Grudzińska M, Stachnik B, Galanty A, Sołtys A, Podolak I. Progress in Antimelanoma Research of Natural Triterpenoids and Their Derivatives: Mechanisms of Action, Bioavailability Enhancement and Structure Modifications. Molecules 2023; 28:7763. [PMID: 38067491 PMCID: PMC10707933 DOI: 10.3390/molecules28237763] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Melanoma is one of the most dangerous forms of skin cancer, characterized by early metastasis and rapid development. In search for effective treatment options, much attention is given to triterpenoids of plant origin, which are considered promising drug candidates due to their well described anticancer properties and relatively low toxicity. This paper comprehensively summarizes the antimelanoma potential of natural triterpenoids, that are also used as scaffolds for the development of more effective derivatives. These include betulin, betulinic acid, ursolic acid, maslinic acid, oleanolic acid, celastrol and lupeol. Some lesser-known triterpenoids that deserve attention in this context are 22β-hydroxytingenone, cucurbitacins, geoditin A and ganoderic acids. Recently described mechanisms of action are presented, together with the results of preclinical in vitro and in vivo studies, as well as the use of drug delivery systems and pharmaceutical technologies to improve the bioavailability of triterpenoids. This paper also reviews the most promising structural modifications, based on structure-activity observations. In conclusion, triterpenoids of plant origin and some of their semi-synthetic derivatives exert significant cytotoxic, antiproliferative and chemopreventive effects that can be beneficial for melanoma treatment. Recent data indicate that their poor solubility in water, and thus low bioavailability, can be overcome by complexing with cyclodextrins, or the use of nanoparticles and ethosomes, thus making these compounds promising antimelanoma drug candidates for further development.
Collapse
Affiliation(s)
- Marta Grudzińska
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Łazarza 16, 31-530 Kraków, Poland
| | - Bogna Stachnik
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| | - Agnieszka Galanty
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| | - Agnieszka Sołtys
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| | - Irma Podolak
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| |
Collapse
|
43
|
Pan Y, Zhang Y, Ouyang H, Gong T, Zhang Z, Cao X, Fu Y. Targeted Delivery of Celastrol via Chondroitin Sulfate Derived Hybrid Micelles for Alleviating Symptoms in Nonalcoholic Fatty Liver Disease. ACS APPLIED BIO MATERIALS 2023; 6:4877-4893. [PMID: 37890075 DOI: 10.1021/acsabm.3c00612] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is caused by an accumulation of excess fat in the liver leading to oxidative stress and liver cell injury, as well as overproduction of inflammatory cytokines. CD44 has been identified as a potential therapeutic target in the development of NAFLD to nonalcoholic steatohepatitis. Here, chondroitin sulfate (CS) is selected to construct a CD44-targeted delivery system for the treatment of NAFLD. Specifically, two CS-derived amphiphilic materials including CS conjugated with either 4-aminophenylboronic acid pinacol ester (CS-PBE) or phenformin (CS-PFM) were synthesized, respectively. The presence of PBE moieties on CS-PBE rendered the vehicle with enhanced loading capacity and scavenging potential against reactive oxygen species, while the presence of guanidine moieties on CS-PFM enhanced the internalization of vehicles in the differentiated hepatocytes. Next, celastrol (CLT) was encapsulated in the hybrid micelle to afford CS-Hybrid/CLT, which demonstrates sufficient stability, enhanced cellular uptake efficiencies in differentiated HepG2 cells, and therapeutic potential to alleviate lipid accumulation in differentiated HepG2 cells. In a high-fat-diet-induced NAFLD rat model, CS-Hybrid/CLT micelles demonstrated the capacity to dramatically decrease hepatic lipid accumulation and free fatty acid levels with greatly improved pathologic liver histology and downregulated hepatic inflammation levels. These results suggest that CS-based amphiphilic micelles may offer a promising strategy to effectively deliver therapeutic cargos to the liver for the treatment of NAFLD.
Collapse
Affiliation(s)
- Yi Pan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yunxiao Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hongling Ouyang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xi Cao
- Department of Pharmacy, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Grade 3 Pharmaceutical Chemistry Laboratory of State Administrate of Traditional Chinese Medicine, Hefei 230022, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
44
|
Shan Y, Zhao J, Wei K, Jiang P, Xu L, Chang C, Xu L, Shi Y, Zheng Y, Bian Y, Zhou M, Schrodi SJ, Guo S, He D. A comprehensive review of Tripterygium wilfordii hook. f. in the treatment of rheumatic and autoimmune diseases: Bioactive compounds, mechanisms of action, and future directions. Front Pharmacol 2023; 14:1282610. [PMID: 38027004 PMCID: PMC10646552 DOI: 10.3389/fphar.2023.1282610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Rheumatic and autoimmune diseases are a group of immune system-related disorders wherein the immune system mistakenly attacks and damages the body's tissues and organs. This excessive immune response leads to inflammation, tissue damage, and functional impairment. Therapeutic approaches typically involve medications that regulate immune responses, reduce inflammation, alleviate symptoms, and target specific damaged organs. Tripterygium wilfordii Hook. f., a traditional Chinese medicinal plant, has been widely studied in recent years for its application in the treatment of autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, and multiple sclerosis. Numerous studies have shown that preparations of Tripterygium wilfordii have anti-inflammatory, immunomodulatory, and immunosuppressive effects, which effectively improve the symptoms and quality of life of patients with autoimmune diseases, whereas the active metabolites of T. wilfordii have been demonstrated to inhibit immune cell activation, regulate the production of inflammatory factors, and modulate the immune system. However, although these effects contribute to reductions in inflammatory responses and the suppression of autoimmune reactions, as well as minimize tissue and organ damage, the underlying mechanisms of action require further investigation. Moreover, despite the efficacy of T. wilfordii in the treatment of autoimmune diseases, its toxicity and side effects, including its potential hepatotoxicity and nephrotoxicity, warrant a thorough assessment. Furthermore, to maximize the therapeutic benefits of this plant in the treatment of autoimmune diseases and enable more patients to utilize these benefits, efforts should be made to strengthen the regulation and standardized use of T. wilfordii.
Collapse
Affiliation(s)
- Yu Shan
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Jiang
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Xu
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linshuai Xu
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Shi
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yixin Zheng
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqin Bian
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Mi Zhou
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
| | - Steven J. Schrodi
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI. United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI. United States
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
45
|
Zhu F, Huang C, Lin Y, Li Y, Tu R, Lu W. Self-delivery of a metal-coordinated anti-angiogenic nanodrug with GSH depleting ability for synergistic chemo-phototherapy. Biomater Sci 2023; 11:7132-7145. [PMID: 37706659 DOI: 10.1039/d3bm00994g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Synergistic chemo-phototherapy has offered tremendous potential in cancer treatment. Nevertheless, nanosystems usually suffer from the complexity of multicomponents (polymeric or inorganic materials), which results in carrier-related toxicity issues. Moreover, the GSH over-expression of tumor cells seriously compromises ROS therapeutic efficiency. Herein, we designed a self-delivered nanodrug via Cu(II) coordination-driven co-self-assembly of celastrol (CST, a chemo-drug with anti-angiogenesis activity) and indocyanine green (ICG, a photosensitizer) for synergistic chemo-phototherapy with GSH depletion. The nanodrug was further cloaked by an erythrocyte membrane (RBC) to prolong the circulation time. Within the tumor microenvironment, the nanodrug would be disassembled upon intracellular GSH triggering. Moreover, the released Cu(II) could efficiently deplete the GSH, thus damaging the ROS-scavenging system and amplifying the phototherapeutic efficiency upon laser irradiation. The in vivo experiments validated the highly effective accumulation at tumor sites, potent tumor growth inhibition, and inappreciable systemic toxicity. The tumor microenvironment-responsive coordination-driven self-assembled biomimetic nanodrug may hold potential applications in tumor theranostics.
Collapse
Affiliation(s)
- Fukai Zhu
- Department of Gynecology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China.
- Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou, Fujian 363000, China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, China
| | - Cailin Huang
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, China
| | - YanLing Lin
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yang Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Department of Translational Medicine & Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare-Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, China
| | - Ruiqin Tu
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weihong Lu
- Department of Gynecology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, China.
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, 361015, China
| |
Collapse
|
46
|
Nie X, Fu L, Cheng Y, Wu X, Lv K, Li R, Wu Y, Leung GPH, Fu C, Lee SMY, Seto SW, Zhang J, Li J. Garcinone E suppresses breast cancer growth and metastasis by modulating tumor-associated macrophages polarization via STAT6 signaling. Phytother Res 2023; 37:4442-4456. [PMID: 37259475 DOI: 10.1002/ptr.7909] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Cancer metastasis remains the most common cause of death in breast cancer patients. Tumor-associated macrophages (TAMs) are a novel therapeutic target for the treatment of metastatic breast cancer. Despite the good anti-cancer activity of garcinone E (GE), there are no reports on its therapeutic effects on breast cancer metastasis. The objective of this study was to examine the anti-cancer effects of GE on metastatic breast cancer. RAW 264.7 and THP-1 cells were polarized to M2 macrophages by IL-4/IL-13 in vitro. A 4T1 mouse breast cancer model and the tail vein breast cancer metastasis model were used to explore the effect of GE on breast cancer growth and metastasis in vivo. In vitro studies showed that GE dose-dependently suppressed IL-4 + IL-13-induced expression of CD206 in both RAW 264.7 cells and differentiated THP-1 macrophages. However, GE did not affect the LPS + IFN-γ-induced polarization to the M1-like macrophages in vitro. GE inhibited the expression of the M2 macrophage specific genes in RAW 264.7 cells, and simultaneously impaired M2 macrophage-induced breast cancer cell proliferation and migration, and angiogenesis. In animal studies, GE significantly suppressed tumor growth, angiogenesis, and lung metastasis in 4T1 tumor-bearing mice, without causing toxicity. In both tumor and lung tissues, the proportion of M2-like TAMs was significantly decreased while the proportion of M1-like TAMs was markedly increased by GE treatment. Mechanistically, GE inhibited phosphorylation of STAT6 in vitro and in vivo. Our results demonstrate for the first time that GE suppresses breast cancer growth and pulmonary metastasis by modulating M2-like macrophage polarization through the STAT6 signaling pathway.
Collapse
Affiliation(s)
- Xin Nie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, SAR, China
| | - Li Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanfen Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoping Wu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Kongpeng Lv
- Department of Interventional Radiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, SAR, China
| | - Sai-Wang Seto
- Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hong Kong, China
- The Research Center for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingjing Li
- The Research Center for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hong Kong, SAR, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong, SAR, China
| |
Collapse
|
47
|
Zhou M, Liao J, Lai W, Xu R, Liu W, Xie D, Wang F, Zhang Z, Huang J, Zhang R, Li G. A celastrol-based nanodrug with reduced hepatotoxicity for primary and metastatic cancer treatment. EBioMedicine 2023; 94:104724. [PMID: 37480625 PMCID: PMC10393547 DOI: 10.1016/j.ebiom.2023.104724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Cancer is the world's leading cause of death and a key hindrance to extending life expectancy. Celastrol, a bioactive compound derived from Tripterygium wilfordii, has been shown to have excellent antitumor activity, but its poor solubility and severe organ toxicity side effects have hampered its clinical application. METHODS In this study, a self-assembled nanodrug (PLC-NP) was designed to deliver celastrol to tumor sites while efficiently reducing its side effects by conjugating celastrol with the bioactive material LMWH and P-selectin targeting peptide (PSN). Extensive in vitro and in vivo experiments were performed to investigate both therapeutic efficacy and adverse effects. Furthermore, the specific mechanism of the antitumor activity has also been explored. FINDING The PLC-NP nanodrugs were spherical in shape, with a mean particle size of 115.83 ± 6.93 nm. PLC-NP was sufficiently stable during blood circulation, with a selective target to P-selectin-highly expressed tumor cells, followed by releasing the containing celastrol under acidic environment and high levels of esterase in tumor cells. Both in vitro and in vivo results confirmed that celastrol's antitumor and anti-metastatic abilities were not attenuated and were actually strengthened after being formed into nanodrugs. More importantly, the organ toxicities of the modified celastrol nanodrug were dramatically reduced. Mechanistic study indicated that the inactivation of PI3K/Akt/mTOR signaling pathway and ROS-mediated mitochondrial dysfunction play critical roles in celastrol-mediated autophagy and apoptosis. INTERPRETATION Our findings could offer a potential strategy for the translation of toxic compounds into clinical therapeutic nanomedicine. FUNDING See a detailed list of funding bodies in the Acknowledgements section at the end of the manuscript.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Jiaxing Liao
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Wenjing Lai
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Rufu Xu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Wuyi Liu
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Dandan Xie
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Fengling Wang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Zhe Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Jingbin Huang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China.
| | - Guobing Li
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, 183 Xinqiao Road, Chongqing, 400037, China.
| |
Collapse
|
48
|
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
|
49
|
Wu Y, Zhou M, Lin R, Yu L, Zhang X, Xie J. Acid-Responsive Macroporous Silica Nanoparticles for Bcl-2-Functional-Converting Peptide Release and Synergism with Celastrol for Enhanced Therapy against Resistant Cancer. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37312263 DOI: 10.1021/acsami.3c03670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Combination of chemotherapeutics with polypeptide/protein drugs has been demonstrated to be an effective approach for treatment against cancer multidrug resistance. However, due to the low biostability and weak cell penetrating ability of biomacromolecules, intracellular delivery and release of biomacromolecules in a spatiotemporally controllable manner in target sites in vivo face great challenges, and synergistic effects will not be achieved as expected just by simple drug combination. Here, we conceived an inspired strategy to combat the drug-resistant tumors by fabricating multiarm PEG-gated large pore-sized mesoporous silica nanoparticles for the Bcl-2-functional-converting peptide (denoted as N9@M-CA∼8P) payload and controlled release and realizing synergistic effects with celastrol integration at a low dosage as a curative sensitizer. Our results demonstrated that the N9 peptide could be pH-responsively released from the macropores of the M-CA∼8P nanosystem both in simulated physiological environments and in cancer cells and at tumor sites. Biosafe and enhanced therapeutic outcomes (90% tumor inhibition) were obtained by combination of the N9@M-CA∼8P nanosystem with celastrol coordinatively inducing mitochondrion-mediated cell apoptosis in resistant cancer cell lines and in the corresponding xenografted mice models. Overall, this study provides convincing evidence for effective and safe resistant cancer treatment through a stimulus-responsive biomacromolecule nanosystem combined with a low dosage of a natural compound.
Collapse
Affiliation(s)
- Yuehuang Wu
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Min Zhou
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Ruimiao Lin
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Lixue Yu
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Xiaokun Zhang
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Jingjing Xie
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| |
Collapse
|
50
|
Zhang S, Liu L, Yue B, Wu X, Ji H, Wang J, Jiang Z, Liu C, Wu X. Seven lower toxicity celastrol derivatives by biotransformation of Pestalotiopsis sp. LGT-1. PHYTOCHEMISTRY 2023:113750. [PMID: 37279870 DOI: 10.1016/j.phytochem.2023.113750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/08/2023]
Abstract
Biotransformation of toxic components by plant endophytes has become an effective method to reduce the toxicity of target compounds and discover lead compounds. In this context, an endophytic fungus, Pestalotiopsis sp. LGT-1, from Tripterygium wilfordii Hook F. (TwHF), was used to reduce the toxicity of celastrol which is also produced by TwHF and is considered an attractive molecule with a variety of biological activities. Seven celastrol derivatives (1-7) were isolated from the coculture fermentation broth of LGT-1 and celastrol. Their structures were elucidated by spectroscopic data analysis including 1D and 2D NMR, as well as HRESIMS. Their absolute configurations were determined by analysis of NOESY, ECD data and NMR calculations. In cell proliferation experiments, the toxicity of seven compounds was 10.11- to 1.24-fold lower in normal cells than the prototype compound celastrol. These derivatives serve as potential candidates for future pharmaceutical applications.
Collapse
Affiliation(s)
- Senyu Zhang
- College of Pharmacy, Ningxia Medical University; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Lumei Liu
- College of Pharmacy, Ningxia Medical University; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Bangwen Yue
- College of Pharmacy, Ningxia Medical University; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Xinyuan Wu
- College of Pharmacy, Ningxia Medical University; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Hongyan Ji
- Department of Pharmaceutics, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jianhuan Wang
- College of Pharmacy, Ningxia Medical University; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Zhibo Jiang
- School of Chemistry and Chemical Engineering, Key Laboratory for Chemical Engineering and Technology, State Ethnic Affairs Commission, North Minzu University, Yinchuan, China.
| | - Cheng Liu
- College of Pharmacy, Ningxia Medical University; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China.
| | - Xiuli Wu
- College of Pharmacy, Ningxia Medical University; Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|