1
|
Bai Q, Wang C, Ding N, Wang Z, Liu R, Li L, Piao H, Song Y, Yan G. Eupalinolide B targets DEK and PANoptosis through E3 ubiquitin ligases RNF149 and RNF170 to negatively regulate asthma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156657. [PMID: 40120540 DOI: 10.1016/j.phymed.2025.156657] [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: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE We investigated the mechanism by which eupalinolide B (EB) regulates DEK protein ubiquitination and degradation, and its impact on DEK-mediated receptor-interacting protein kinase 1 (RIPK)-PANoptosis pathway in allergic asthma. STUDY DESIGN AND METHODS In vitro studies were conducted on human bronchial epithelial cells (BEAS-2B) treated with EB and human-recombinant DEK. Mass spectrometry analysis, RNA sequencing, molecular docking, and functional assays were used to assess the interactions and effects of EB, DEK, and ring finger protein 149 and 170 (RNF149 and RNF170). In vivo experiments involved a house dust mite-induced asthma model in mice and evaluation of airway inflammation, DEK expression, and PANoptosis markers. RESULTS In vitro, EB could bind to DEK. RNF149 and RNF170 were identified as regulatory factors of DEK, polyubiquitinating the K349 site in the DEK coding DNA sequence region 270-350 through K48 linkages and leading to its degradation. RNA sequencing showed that DEK overexpression upregulated the expression of genes such as RIPK1, FADD, and Caspase 8. Treatment with DEK siRNA or EB reduced the activation of the RIPK1-PANoptosis pathway in BEAS-2B-DEK cells. In vivo, EB significantly reduced the levels of DEK in house dust mite-induced mice and alleviated pulmonary inflammatory cell infiltration, goblet cell hyperplasia, collagen fiber deposition, and eosinophil proportion in BALF. Knocking out the DEK gene reduced RIPK1-induced PANoptosis, and inhibited airway inflammation and cell apoptosis. CONCLUSION EB promotes the degradation of DEK by RNF149 and RNF170, inhibits the RIPK1-PANoptosis pathway, and may effectively suppress asthma. EB may become a potential drug for treating airway inflammation in asthma.
Collapse
Affiliation(s)
- Qiaoyun Bai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Ningpo Ding
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Zhiguang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, PR China
| | - Ruobai Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Hongmei Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, PR China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, PR China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, PR China.
| |
Collapse
|
2
|
Wang TT, Zhou MY, Gong XN, Huang Y, Li FL, Gu SL, Zhang MY, Li LL, Xu ZS, Li R, Cai L. Eupalinolide B alleviates corticosterone-induced PC12 cell injury and improves depression-like behaviors in CUMS rats by regulating the GSK-3β/β-catenin pathway. Biochem Pharmacol 2025; 235:116831. [PMID: 40021022 DOI: 10.1016/j.bcp.2025.116831] [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: 10/23/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Eupalinolide B (EB), a primary bioactive compound isolated from Eupatorium lindleyanum DC., has exhibited various pharmacological properties, such as antitumor, anti-inflammatory, and notably, neuroprotective effects in neurodegenerative diseases. However, the in-depth studies on the antidepressant potential of EB and its underlying mechanisms are still lacking. Herein, we investigated the therapeutic effects of EB on corticosterone (CORT)-induced neurotoxicity in PC12 cells and its antidepressant-like effects in rats subjected to chronic unpredictable mild stress (CUMS). In particular, we focused on the molecular mechanisms related to modulating the GSK-3β/β-catenin pathway. Our findings revealed that EB promoted cell proliferation while decreasing apoptosis and oxidative stress in CORT-induced PC12 cells. In vivo, EB alleviated the depressive-like behaviors in CUMS rats, as assayed by the sucrose preference test, open field test, and forced swim test. Additionally, EB attenuated the hippocampal pathological damage and increased Ki67- and doublecortin-positive cell numbers in hippocampal dentate gyrus, thus restoring hippocampal neurogenesis in CUMS rats. The binding of EB to GSK-3β was confirmed using molecular docking and cellular thermal shift assays. Overexpression of GSK-3β diminished the therapeutic effects of EB on CORT-induced PC12 cells, further indicating that GSK-3β is the target of EB. Mechanistically, EB hindered GSK-3β activity and thus activated β-catenin signaling in both CORT-induced PC12 cells and CUMS rat hippocampus, as demonstrated by increased p-GSK-3β (Ser9), reduced p-β-catenin, and elevated β-catenin expression. Collectively, this study offers new insights into the antidepressant mechanisms of EB, highlighting its potential as a candidate for depression treatment.
Collapse
Affiliation(s)
- Tian-Tian Wang
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022 Anhui Province, PR China; Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Xue-Na Gong
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Fei-Long Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Man-Yu Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032 Anhui Province, PR China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230026 Anhui Province, PR China.
| | - Li Cai
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022 Anhui Province, PR China; Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032 Anhui Province, PR China.
| |
Collapse
|
3
|
Ouyang X, Wang J, Qiu X, Hu D, Cui J. Current developments of pharmacotherapy targeting heme oxygenase 1 in cancer (Review). Int J Oncol 2025; 66:26. [PMID: 39981901 DOI: 10.3892/ijo.2025.5732] [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: 11/25/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
Malignant tumors are non-communicable diseases that impact human health and quality of life. Identifying and targeting the underlying genetic drivers is a challenge. Heme oxygenase-1 (HO-1), a stress-inducible enzyme also known as heat shock protein 32, plays a crucial role in maintaining cellular homeostasis. It mitigates oxidative stress-induced damage and exhibits anti-apoptotic properties. HO-1 is expressed in a wide range of malignancies and is associated with tumor growth. However, the precise role of HO-1 in tumor development remains controversial. Drugs, both naturally occurring and chemically synthesized, can inhibit tumor growth by modulating HO-1 expression in cancer cells. The present review aimed to discuss biological functions of HO-1 pharmacological therapies targeting HO-1.
Collapse
Affiliation(s)
- Xiaohu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jingbo Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaoyuan Qiu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Cui
- Health Management Center, Hubei Provincial Hospital of Integrated Chinese & Western Medicine, Wuhan, Hubei 430015, P.R. China
| |
Collapse
|
4
|
Gu SL, Liu XS, Xu ZS, Li LL, Wu XJ, Li FL, Huang Y, Ran X, Li R. Eupalinolide B alleviates rheumatoid arthritis through the promotion of apoptosis and autophagy via regulating the AMPK/mTOR/ULK-1 signaling axis. Int Immunopharmacol 2025; 148:114179. [PMID: 39874849 DOI: 10.1016/j.intimp.2025.114179] [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: 12/12/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025]
Abstract
The excessive proliferation of fibroblast-like synoviocytes (FLS) leads to synovial hyperplasia, a key pathological hallmark of rheumatoid arthritis (RA). Eupalinolide B (EB), a sesquiterpene lactone of Eupatorium lindleyanum DC., has anti-inflammatory effects and anti-proliferative activity in tumor cells. However, its potential use in RA treatment is unclear. This study explored EB's anti-rheumatoid activities by promoting apoptosis and autophagy in RA-FLS and the synovium of adjuvant-induced arthritis (AIA) rats, focusing on its regulation of the AMPK/mTOR/ULK-1 axis. Our findings revealed that EB inhibited proliferation, induced apoptosis, and promoted autophagy in RA-FLS. Autophagy inhibition using 3-methyladenine (3-MA) diminished EB's anti-proliferative effects, suggesting that EB promotes RA-FLS autophagy as a death mechanism. Z-VAD-FMK, a pan-caspase inhibitor, decreased EB-induced autophagy, while 3-MA co-treatment reduced caspase-3 activity, demonstrating that EB-induced apoptosis and autophagy promoted each other to support its anti-proliferative effects. In vivo, EB exhibited clear anti-arthritic effects in AIA rats, as shown by reduced paw swelling, arthritis index, serum levels of TNF-α, IL-1β, and MCP-1, and joint damage, along with decreased Ki67 expression, increased apoptosis, and enhanced autophagy in AIA rat synovium. Mechanistically, EB regulated the AMPK/mTOR/ULK-1 axis in RA-FLS and AIA rat synovium, as evidenced by higher expression of p-AMPK and p-ULK-1 and lower levels of p-mTOR. Notably, co-treatment of the AMPK inhibitor compound C negated EB's beneficial effects in RA-FLS and AIA rats. Collectively, EB demonstrated exact anti-RA effects by inducing apoptosis and autophagy via the regulation of the AMPK/mTOR/ULK-1 axis, highlighting its potential for RA therapy.
Collapse
Affiliation(s)
- Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Xue-Song Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Xin-Jie Wu
- The First Clinical Medical College, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Fei-Long Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Xiang Ran
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230026, Anhui Province, China.
| |
Collapse
|
5
|
Wu C, Shen X, Lou P, Song D. Dioscin pretreatment ameliorates ferroptosis in cardiomyocytes after myocardial infarction via inhibiting endoplasmic reticulum stress. Mol Med 2025; 31:32. [PMID: 39881220 PMCID: PMC11780909 DOI: 10.1186/s10020-025-01102-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Myocardial infarction (MI) remains a leading cause of mortality globally, often resulting in irreversible damage to cardiomyocytes. Ferroptosis, a recently identified form of regulated cell death driven by iron-dependent lipid peroxidation, has emerged as a significant contributor to post-MI cardiac injury. The endoplasmic reticulum (ER) stress response has been implicated in exacerbating ferroptosis. METHODS Here, we investigated the potential of Dioscin, a natural compound known for its diverse pharmacological properties, in mitigating ferroptosis in cardiomyocytes following MI by targeting ER stress. RESULTS In animal models subjected to MI, administration of Dioscin notably improved cardiac function, reduced infarct size by approximately 24%, and prevented adverse remodeling, highlighting its therapeutic potential. Through in vitro and in vivo models of MI, we demonstrated that Dioscin treatment significantly attenuates ferroptosis in cardiomyocytes, as evidenced by a decrease in lipid peroxidation by about 19% and preserved mitochondrial integrity. Moreover, Dioscin exerted its protective effects by inhibiting ER stress markers, such as the phosphorylation levels of PERK and eIF2α proteins, and the expression levels of BIP and ATF4 proteins, thus disrupting the ER stress-mediated signaling cascade associated with ferroptosis. CONCLUSION Overall, our findings suggested that Dioscin holds promise as a therapeutic agent against post-MI cardiac injury by mitigating ferroptosis via the suppression of ER stress. Further investigations into the precise molecular mechanisms and clinical translation of Dioscin's cardioprotective effects are warranted, offering a potential avenue for novel therapeutic interventions in MI-related cardiac complications.
Collapse
Affiliation(s)
- Chang Wu
- The First People's Hospital of Lin'an District, No. 360, Yikang Street, Jinnan Subdistrict, Lin'an District, Hangzhou, Zhejiang, 311300, China
- Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xueping Shen
- Center of Prenatal Diagnosis Huzhou Maternity & Child Health Care Hospital, Huzhou, Zhejiang, 313000, China
| | - Pan Lou
- The Second People's Hospital of Anji, Huzhou, Zhejiang, 313307, China
| | - Dongyan Song
- The First People's Hospital of Lin'an District, No. 360, Yikang Street, Jinnan Subdistrict, Lin'an District, Hangzhou, Zhejiang, 311300, China.
- Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
6
|
Kuang W, Zhuge R, Song P, Yi L, Zhang S, Zhang Y, Wong YK, Chen R, Zhang J, Wang Y, Liu D, Gong Z, Wang P, Ouyang X, Wang J. Eupalinolide B inhibits periodontitis development by targeting ubiquitin conjugating enzyme UBE2D3. MedComm (Beijing) 2025; 6:e70034. [PMID: 39811801 PMCID: PMC11731104 DOI: 10.1002/mco2.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 01/16/2025] Open
Abstract
Periodontitis is a chronic periodontal inflammatory disease caused by periodontal pathogens commonly seen in adults. Eupalinolide B (EB) is a sesquiterpenoid natural product extracted from Eupatorium lindleyanum and has been reported as a potential drug for cancers and immune disorders. Here, we explored the ameliorative effects and underlying molecular mechanism of EB on periodontitis for the first time. We demonstrated that EB ameliorates periodontal inflammation and alveolar bone resorption with a ligated periodontitis mouse model. In addition, the impact of EB on macrophages inflammation was examined in the Raw264.7 cell line. We identified ubiquitin-conjugating enzyme, UBE2D3, as the direct covalent binding protein targets of EB by using a chemoproteomic method based on activity-based protein profiling, biolayer interferometry method, and cellular thermal shift assay. Furthermore, the direct binding site of EB to UBE2D3 was identified using high-resolution mass spectrometry and confirmed by experiments. Taken together, EB ameliorates periodontitis by targeting UBE2D3 to suppress the ubiquitination degradation of IκBα, leading to inactivation of nuclear transcription factor-κB signaling pathway. And this was confirmed by siRNA-mediated gene knockdown in inflammatory macrophages. Our results suggested that EB may be a new kind of UBE2D3 inhibitor and may become a promising therapeutic agent for anti-periodontitis.
Collapse
Affiliation(s)
- Wenhua Kuang
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
| | - Ruishen Zhuge
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
- Department of Periodontology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijingChina
| | - Ping Song
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
- National Clinical Research Center for Chinese Medicine CardiologyXiyuan Hospital, China Academy of Chinese Medical SciencesBeijingChina
| | - Letai Yi
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
- Inner Mongolia Medical UniversityHohhotChina
| | - Shujie Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Ying Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Yin Kwan Wong
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
| | - Ruixing Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of PharmaceuticsGuizhou Medical UniversityGuiyangChina
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Yuanbo Wang
- Department of Periodontology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijingChina
| | - Dandan Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Zipeng Gong
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of PharmaceuticsGuizhou Medical UniversityGuiyangChina
| | - Peili Wang
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
- National Clinical Research Center for Chinese Medicine CardiologyXiyuan Hospital, China Academy of Chinese Medical SciencesBeijingChina
| | - Xiangying Ouyang
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
- Department of Periodontology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijingChina
| | - Jigang Wang
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for GeriatricsShenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and TechnologyShenzhenChina
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
- State Key Laboratory of Antiviral Drugs, School of PharmacyHenan UniversityKaifengChina
| |
Collapse
|
7
|
Xu C, Wen S, Du X, Zou X, Leung ELH, Zhou G, Wu Q, Shen B. Targeting regulated cell death (RCD) with naturally derived sesquiterpene lactones in cancer therapy. Pharmacol Res 2025; 211:107553. [PMID: 39706282 DOI: 10.1016/j.phrs.2024.107553] [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: 09/30/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Regulated cell death (RCD) is a type of cell death modulated by specific signal transduction pathways. Currently, known RCD types include apoptosis, autophagy, ferroptosis, necroptosis, cuproptosis, pyroptosis, and NETosis. Mutations in cancer cells may prevent the RCD pathway; therefore, targeting RCD in tumors has become a promising therapeutic approach. Sesquiterpene lactones represent a diverse and extensive class of plant-derived phytochemicals that serve as potential sources for developing various drugs. Recent studies have shown that sesquiterpene lactones have promising potential in cancer treatment. This review systematically summarizes recent progress in the study of sesquiterpene lactones as antitumor agents, highlighting their role in targeting various RCD pathways, including those involved in apoptosis, autophagy, ferroptosis, necroptosis, and cuproptosis. The primary purpose of the present review is to provide a clear picture of the regulation of RCD by sesquiterpene lactones against different targets in various cancers, which will facilitate the development of new strategies for cancer therapy.
Collapse
Affiliation(s)
- Cong Xu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China; State Key Laboratory of Quality Research in Chinese Medicines and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao
| | - Shaodi Wen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Xiaoyue Du
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Xinhua Zou
- Department of Vascular and Tumor Interventional Medicine, Affiliated Hospital of Jining Medical University, Jining 272000, China
| | | | - Guoren Zhou
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines and Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing 210000, China; DongTai People's Hospital, Dongtai, Jiangsu, China.
| |
Collapse
|
8
|
Huang L, Li G, Zhang Y, Zhuge R, Qin S, Qian J, Chen R, Kwan Wong Y, Tang H, Wang P, Xiao W, Wang J. Small-molecule targeting BCAT1-mediated BCAA metabolism inhibits the activation of SHOC2-RAS-ERK to induce apoptosis of Triple-negative breast cancer cells. J Adv Res 2024:S2090-1232(24)00476-4. [PMID: 39490614 DOI: 10.1016/j.jare.2024.10.021] [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: 08/18/2024] [Revised: 10/03/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer with the worst prognosis. Exploring novel carcinogenic factors and therapeutic drugs for TNBC remains a focus to improve prognosis. Branched-chain amino acid transaminase 1 (BCAT1), a crucial enzyme in branched-chain amino acid (BCAA) metabolism, has been linked to various tumor developments, but its carcinogenic function and mechanism in TNBC remain unclear. Eupalinolide B (EB) is a naturally-derived small-molecule with anti-tumor activity, but its role in TNBC remains unknown. OBJECTIVES By exploring the targets and pharmacological mechanisms of EB in inhibiting TNBC, this study aimed to discover novel therapeutic targets and potential inhibitors for TNBC, and elucidate novel pathogenic mechanisms of TNBC. METHODS The inhibitory effect of EB on TNBC was investigated using mouse models and cellular phenotypic experiments. Activity-based protein profiling (ABPP) technology, pull down-WB, CETSA-WB and MST were utilized to discover and validate the targets of EB. The oncogenic role of BCAT1 was determined through clinical data analysis and biochemical experiments. To elucidate the mechanism by which EB inhibited TNBC, many methods, including but not limited to HPLC and proteomic sequencing were used. RESULTS We found that EB significantly inhibited TNBC progression. We identified BCAT1 as the direct target of EB and confirmed that BCAT1 was critical for TNBC development. EB inhibited BCAT1-involved BCAA metabolism to reduce the synthesis of BCAAs (including Leu, Ile, and Val), thereby inhibiting SHOC2 (a Leu-rich repeat protein) expression and the downstream SHOC2-participating RAS-ERK signaling pathway, ultimately leading to apoptosis of TNBC cells. CONCLUSION Collectively, this study not only elucidates the oncogenic role of BCAT1 and its downstream SHOC2-RAS-ERK signaling axis in TNBC progression but also opens up avenues for potential therapies targeting BCAT1 or BCAA metabolism (using EB alone or in combination with its inhibitor candesartan) for TNBC treatment.
Collapse
Affiliation(s)
- Ling Huang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Guanjun Li
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Ying Zhang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China; Oncology Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 Bei Xian Ge, Xi Cheng District, Beijing 100053, China
| | - Ruishen Zhuge
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China; Department of Periodontology, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, 100081, China
| | - Shijie Qin
- Innovative Vaccine and Immunotherapy Research Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jinjun Qian
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Ave., Nanjing, Jiangsu 210023, China
| | - Ruixing Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yin Kwan Wong
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China
| | - Huan Tang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Peili Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Wei Xiao
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China; Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Jigang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
9
|
Xu Q, Ren L, Ren N, Yang Y, Pan J, Zheng Y, Wang G. Ferroptosis: a new promising target for hepatocellular carcinoma therapy. Mol Cell Biochem 2024; 479:2615-2636. [PMID: 38051404 DOI: 10.1007/s11010-023-04893-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the sixed most common malignant tumor in the world. The study for HCC is mired in the predicament confronted with the difficulty of early diagnosis and high drug resistance, the survival rate of patients with HCC being low. Ferroptosis, an iron-dependent cell death, has been discovered in recent years as a cell death means with tremendous potential to fight against cancer. The in-depth researches for iron metabolism, lipid peroxidation and dysregulation of antioxidant defense have brought about tangible progress in the firmament of ferroptosis with more and more results showing close connections between ferroptosis and HCC. The potential role of ferroptosis has been widely used in chemotherapy, immunotherapy, radiotherapy, and nanotherapy, with the development of various new drugs significantly improving the prognosis of patients. Based on the characteristics and mechanisms of ferroptosis, this article further focuses on the main signaling pathways and promising treatments of HCC, envisioning that existing problems in regard with ferroptosis and HCC could be grappled with in the foreseeable future.
Collapse
Affiliation(s)
- Qiaoping Xu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medical, Hangzhou, 310006, China
| | - Lanqi Ren
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Ning Ren
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Yibei Yang
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Junjie Pan
- Fourth Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Yu Zheng
- Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310051, China
| | - Gang Wang
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Westlake University School of Medical, Hangzhou, 310006, China.
| |
Collapse
|
10
|
Zhao L, Miao H, Quan M, Wang S, Zhang Y, Zhou H, Zhang X, Lin Z, Piao J. β-Lapachone induces ferroptosis of colorectal cancer cells via NCOA4-mediated ferritinophagy by activating JNK pathway. Chem Biol Interact 2024; 389:110866. [PMID: 38218311 DOI: 10.1016/j.cbi.2024.110866] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/26/2023] [Accepted: 01/07/2024] [Indexed: 01/15/2024]
Abstract
β-Lapachone is a natural product that can promote ROS generation and ultimately triggers tumor cells death by inducing DNA damage. Recent studies have indicated that the targeting of ferroptosis or iron metabolism is a feasible strategy for treating cancer. In this study, bulk RNA-seq analysis suggested that β-Lapachone might induce ferroptosis in CRC cells. We further tested this hypothesis using a xenograft model of human colorectal cancer as an animal model and in SW620 and DLD-1 of CRC cell lines. Western blot was used to determine the key proteins of ferroptosis (SLC7A11, GPX4), autophagy (LC3B, P62, ATG7), ferritinophagy (NCOA4, FTH1, TFRC), and JNK pathway (p-JNK, JNK, p-c-Jun, c-Jun). The levels of MDA, GSH/GSSG, lipid ROS, and intracellular ferrous iron were determined after β-Lapachone treatment, and inhibitors of various pathways, including NAC, Ferrostatin-1, DFO, 3-MA, and SP600125 were utilized to explore the molecular mechanism underlying β-Lapachone-mediated ferroptosis. As the result, we identified that β-Lapachone inhibited cell proliferation and induced apoptosis, autophagy, and ROS generation. In addition, β-Lapachone induced ferroptosis as demonstrated by intra-cellular iron overload, increased levels of lipid ROS and MDA. Mechanistically, JNK signaling pathway was involved in β-Lapachone-induced xCT/GPX4-mediated ferroptosis and NCOA4-mediated ferritinophagy in CRC cells. In vivo experiments in nude mice demonstrated that β-Lapachone significantly inhibited CRC growth and induced ferroptosis and NCOA4-mediated ferritinophagy. These findings not only identify a novel role for β-Lapachone in ferroptosis but also indicate that β-Lapachone may be a valuable candidate for the research and development of anti-cancer therapeutic agents.
Collapse
Affiliation(s)
- Lei Zhao
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Hui Miao
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Mingqi Quan
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Shuhao Wang
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Yu Zhang
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Houkun Zhou
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Xianglan Zhang
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Zhenhua Lin
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Junjie Piao
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China.
| |
Collapse
|
11
|
Consoli V, Fallica AN, Sorrenti V, Pittalà V, Vanella L. Novel Insights on Ferroptosis Modulation as Potential Strategy for Cancer Treatment: When Nature Kills. Antioxid Redox Signal 2024; 40:40-85. [PMID: 37132605 PMCID: PMC10824235 DOI: 10.1089/ars.2022.0179] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
Significance: The multifactorial nature of the mechanisms implicated in cancer development still represents a major issue for the success of established antitumor therapies. The discovery of ferroptosis, a novel form of programmed cell death distinct from apoptosis, along with the identification of the molecular pathways activated during its execution, has led to the uncovering of novel molecules characterized by ferroptosis-inducing properties. Recent advances: As of today, the ferroptosis-inducing properties of compounds derived from natural sources have been investigated and interesting findings have been reported both in vitro and in vivo. Critical Issues: Despite the efforts made so far, only a limited number of synthetic compounds have been identified as ferroptosis inducers, and their utilization is still limited to basic research. In this review, we analyzed the most important biochemical pathways involved in ferroptosis execution, with particular attention to the newest literature findings on canonical and non-canonical hallmarks, together with mechanisms of action of natural compounds identified as novel ferroptosis inducers. Compounds have been classified based on their chemical structure, and modulation of ferroptosis-related biochemical pathways has been reported. Future Directions: The outcomes herein collected represent a fascinating starting point from which to take hints for future drug discovery studies aimed at identifying ferroptosis-inducing natural compounds for anticancer therapies. Antioxid. Redox Signal. 40, 40-85.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| |
Collapse
|
12
|
Li Y, Liu X, Li L, Zhang T, Gao Y, Zeng K, Wang Q. Characterization of the metabolism of eupalinolide A and B by carboxylesterase and cytochrome P450 in human liver microsomes. Front Pharmacol 2023; 14:1093696. [PMID: 36762117 PMCID: PMC9905117 DOI: 10.3389/fphar.2023.1093696] [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: 11/09/2022] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Eupalinolide A (EA; Z-configuration) and eupalinolide B (EB; E-configuration) are bioactive cis-trans isomers isolated from Eupatorii Lindleyani Herba that exert anti-inflammatory and antitumor effects. Although one pharmacokinetic study found that the metabolic parameters of the isomers were different in rats, metabolic processes relevant to EA and EB remain largely unknown. Our preliminary findings revealed that EA and EB are rapidly hydrolyzed by carboxylesterase. Here, we investigated the metabolic stability and enzyme kinetics of carboxylesterase-mediated hydrolysis and cytochrome P450 (CYP)-mediated oxidation of EA and EB in human liver microsomes (HLMs). We also explored differences in the hydrolytic stability of EA and EB in human liver microsomes and rat liver microsomes (RLMs). Moreover, cytochrome P450 reaction phenotyping of the isomers was performed via in silico methods (i.e., using a quantitative structure-activity relationship model and molecular docking) and confirmed using human recombinant enzymes. The total normalized rate approach was considered to assess the relative contributions of five major cytochrome P450s to EA and EB metabolism. We found that EA and EB were eliminated rapidly, mainly by carboxylesterase-mediated hydrolysis, as compared with cytochrome P450-mediated oxidation. An inter-species difference was observed as well, with faster rates of EA and EB hydrolysis in rat liver microsomes. Furthermore, our findings confirmed EA and EB were metabolized by multiple cytochrome P450s, among which CYP3A4 played a particularly important role.
Collapse
Affiliation(s)
- Yingzi Li
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Xiaoyan Liu
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Ludi Li
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Tao Zhang
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Yadong Gao
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China,*Correspondence: Kewu Zeng, ; Qi Wang,
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing, China,Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, Beijing, China,Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, China,*Correspondence: Kewu Zeng, ; Qi Wang,
| |
Collapse
|
13
|
Eupalinolide O Induces Apoptosis in Human Triple-Negative Breast Cancer Cells via Modulating ROS Generation and Akt/p38 MAPK Signaling Pathway. JOURNAL OF ONCOLOGY 2022; 2022:8802453. [PMID: 36185619 PMCID: PMC9519309 DOI: 10.1155/2022/8802453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
Background Triple-negative breast cancer (TNBC) is a subtype of breast cancer with limited therapeutic options. Eupalinolide O (EO) was reported to inhibit tumor growth. This study is aimed at exploring the role of EO on TNBC both in vivo and in vitro. Methods. In in vitro experiments, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and clonogenic assay were conducted to measure the impact of EO on TNBC cell growth at different concentrations and time points. Flow cytometry was conducted to evaluate cell apoptosis. Mitochondrial membrane potential (MMP) loss, caspase-3 activity, and reactive oxygen species (ROS) generation were assessed. The expressions of apoptosis-related mRNAs and Akt/p38 MAPK signaling pathway-related proteins were measured. In in vivo experiments, by injecting TNBC cells into the nude mice to induce xenograft tumor, mice were treated with EO for 20 days. Then, in vivo bioluminescence imaging system was utilized to monitor the growth and distribution of TNBC cells. Tumor volume and weight were also recorded. Hematoxylin-eosin (HE) staining and ELISA assay were applied to observe tumor tissue morphology and ROS levels. Furthermore, western blotting was conducted to observe the expression of apoptosis-related proteins and Akt/p38 MAPK signaling pathway-associated proteins. Results EO inhibited the cell viability and proliferation of TNBC cells but not normal epithelial cells. Furthermore, EO induced apoptosis, decreased MMP, and elevated caspase-3 activity and ROS content in TNBC cells. Meanwhile, the expression of apoptosis-related mRNAs and Akt/p38 MAPK pathway-related proteins was regulated by EO treatment. Besides, in vivo experiments demonstrated EO not only suppressed tumor growth, Ki67 expression, ROS generation, and Akt phosphorylation but also upregulated caspase-3 expression and p-38 phosphorylation. Conclusion EO may induce cell apoptosis in TNBC via regulating ROS generation and Akt/p38 MAPK pathway, indicating EO may be a candidate drug for TNBC.
Collapse
|