1
|
Cui D, Zhang C, Zhang L, Zheng J, Wang J, He L, Jin H, Kang Q, Zhang Y, Li N, Sun Z, Zheng W, Wei J, Zhang S, Feng Y, Tan W, Zhong Z. Natural anti-cancer products: insights from herbal medicine. Chin Med 2025; 20:82. [PMID: 40490812 DOI: 10.1186/s13020-025-01124-y] [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: 12/30/2024] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
Herbal medicine exhibits a broad spectrum of potent anti-cancer properties, including the enhancement of tumor immune responses, reversal of multidrug resistance, regulation of autophagy and ferroptosis, as well as anti-proliferative, pro-apoptotic, and anti-metastatic effects. This review systematically explores recent advances (primarily documented since 2019) in research on key anti-cancer compounds derived from herbal medicine, such as apigenin, artemisinin, berberine, curcumin, emodin, epigallocatechin gallate (EGCG), ginsenosides, icariin, resveratrol, silibinin, triptolide, and ursolic acid (UA). These studies were sourced from scientific databases, including PubMed, Web of Science, Medline, Scopus, and Clinical Trials. The review focuses on the significant role that these natural products play in modern oncology, exploring their efficacy, mechanisms of action, and the challenges and prospects of integrating them into conventional cancer therapies. Furthermore, it highlights cutting-edge approaches in cancer research, such as the utilization of gut microbiota, omics technologies, synthetic derivatives, and advanced drug delivery systems (DDS). This review underscores the potential of these natural products to advance the development of novel anti-cancer treatments and support contemporary medicine. Additionally, recent multi-omics findings reveal how these compounds reshape transcriptional and metabolic networks, further broadening their therapeutic scope. Many natural products exhibit synergy with first-line chemotherapies or targeted therapies, thereby enhancing treatment efficacy and reducing side effects. Advanced nano-formulations and antibody-drug conjugates have also substantially improved their bioavailability, making them promising candidates for future translational research.
Collapse
Affiliation(s)
- Dianxin Cui
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 6/F, 3 Sassoon Road, Pokfulam, Hong Kong S.A.R., 999077, China
| | - Lili Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Jingbin Zheng
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Jie Wang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Luying He
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Haochun Jin
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Na Li
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zhenlong Sun
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wenying Zheng
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Jinchao Wei
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Siyuan Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 6/F, 3 Sassoon Road, Pokfulam, Hong Kong S.A.R., 999077, China.
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China.
| |
Collapse
|
2
|
Yu H, Xue T, Mao X. Chinese herbal extracts mediated programmed cell death in cancer and inflammation therapy. J Leukoc Biol 2025; 117:qiaf051. [PMID: 40313183 DOI: 10.1093/jleuko/qiaf051] [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: 08/23/2024] [Revised: 12/13/2024] [Accepted: 04/30/2025] [Indexed: 05/03/2025] Open
Abstract
Programmed cell death is a common phenomenon in the development of organisms. It is an active and orderly mode of cell death determined by genes. Programmed cell death is usually classified into 3 different types according to the cell morphological changes, stimulus, and biochemical pathways involved, namely, apoptosis, programmed necrosis, and autophagy. Chinese herbal extracts, mainly obtained from traditional Chinese medicine and their primary plants through the physicochemical extraction and separation process, are concentrated with 1 or more effective ingredients from the herbal materials. Recently, studies focused on the influence of traditional Chinese medicine on programmed cell death are increasing, involving the protection of the nervous system and cardio-cerebrovascular system, the prevention of gastrointestinal and immune function damage, the treatment against tumors, and so on. This review mainly focuses on the effects of Chinese herbal extracts on various types of programmed cell death. In addition, the therapeutic approaches and prospects of CHEs are also discussed. Although there are promising clinical applications of Chinese herbal extracts, some challenges are still waiting to be overcome by further research for the wider use of Chinese herbal extracts in clinical practice.
Collapse
Affiliation(s)
- Haihong Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
| | - Tingmao Xue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078
- Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Macao SAR, 999078, China
| |
Collapse
|
3
|
Wang Q, Sun Z, Wang M, Feng C, Chen M, Li H, Guo J, Zhang B, Ma K, Liu M, Pang J, Feng Y. Integrated analysis of single-cell and bulk RNA sequencing identifies APOC1 as a biomarker and therapeutic target for G0/G1 cell cycle arrest in cholangiocarcinoma. Genomics 2025; 117:111028. [PMID: 40064358 DOI: 10.1016/j.ygeno.2025.111028] [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: 08/12/2024] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 03/21/2025]
Abstract
Cholangiocarcinoma is characterized by its high malignancy, frequent recurrence and insensitivity to conventional radiotherapy and chemotherapy. This resistance may be associated with the presence of cells in the G0/G1 arrest phase within the cancer. Cancer cells in the G0/G1 phase are resistant to therapies targeting actively dividing cells, allowing them to evade conventional adjuvant treatments and survive. When conditions become favorable, these quiescent cells can re-enter the cell cycle, proliferate and potentially contribute to cancer recurrence. However, the biomarkers for identifying cells in the G0/G1 arrest phase within cholangiocarcinoma and the molecular mechanisms inducing G0/G1 arrest remain unclear. In our study, we first identified APOC1 as a characteristic gene for G0/G1 phase arrest in cholangiocarcinoma through bulk RNA sequencing (bulkRNA-seq). We then used single-cell RNA sequencing(scRNA-seq) for cell cycle inference and localized the expression peaks of APOC1 to verify its active cell cycle phase. Our experiments demonstrated that APOC1 can induce G0/G1 phase arrest in cholangiocarcinoma cells by inhibiting the Wnt/β-catenin signaling pathway, thereby suppressing cell proliferation, migration and invasion. This suggests that APOC1 may serve as a key regulatory factor and an important biomarker for cells in the G0/G1 phase of cholangiocarcinoma.
Collapse
Affiliation(s)
- Qinlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Zhaowei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China.
| | - Maobing Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Chuan Feng
- Medical Department of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Menshou Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Haoran Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Jingyun Guo
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Bingyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Kai Ma
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Miao Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Jinzhong Pang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China
| | - Yujie Feng
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, China.
| |
Collapse
|
4
|
Liu Y, Yang J, Yu F, Li L, Zhao N, Lu C, Lu A, He X. Research advances in traditional Chinese medicine formulae and active components targeting lipid metabolism for hepatocellular carcinoma therapy. Front Pharmacol 2025; 16:1528671. [PMID: 40351413 PMCID: PMC12062747 DOI: 10.3389/fphar.2025.1528671] [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: 11/15/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) has a relatively poor prognosis and a high degree of malignancy. However, the therapeutic drugs are limited. In recent years, abnormal lipid metabolism and its important role in HCC has been reported, and emerging studies found that some formulae and active components of traditional Chinese medicine (TCM) can regulate abnormal lipid metabolism in HCC, showing their good application prospects. Therefore, this article summarizes the changes and the roles of lipid metabolites in HCC progression, and discusses the role of formulae and active components of TCM for the treatment of HCC based on their regulation on abnormal lipid metabolism. A deeper understanding of their relationship may help the precise use of these formulae and active components in HCC.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Yang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fenghua Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Shanghai GuangHua Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Diao Z, Liang Y, Liu Y, Zhang D, Qiu L, Sun J, Lu Q, Liu Y, Cui D, Yin T. Curcumin-enhanced NIR-II-responsive gold nanobipyramids for targeted HSP 90 inhibition. Mater Today Bio 2025; 31:101541. [PMID: 39990737 PMCID: PMC11847553 DOI: 10.1016/j.mtbio.2025.101541] [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: 11/22/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/25/2025] Open
Abstract
Blockade of heat shock protein 90 (HSP90) expression in multimodal synergistic therapy has a great prospect for cancer treatment. Nanomaterials combined with bioinformatic analysis provides accurate guidance for the design of anti-HSP90 nanomedicines. Herein, a NIR-II-responsive nanoplatform was developed under bioinformatics guided to effectly inhibit HSP90 for enhanced synergistic mild-photothermal chemotherapy without any notable tissue damage. The nanoplatforms were assembled from NIR-II-responsive gold nanobipyramids (GNBs) combined with curcumin (Cur) via hydrophobic-hydrophobic interactions and hydrogen bonds. On the basis of drug discovery and network pharmacology, we found that Cur has impressive anti-HSP90 capability and analyzed its therapeutic mechanism against NSCLC. Under the irradiation of NIR-II light, the obtained GNBs-Cur blocked the expression of HPS90 and inhibited related antiapoptotic pathways, thus enhancing the mild PTT of GNBs under 1064 nm laser irradiation. Meanwhile, Cur served as chemotherapeutic agents to induce apoptosis in tumor cells. In vivo photoacoustic imaging-guided, GNBs-Cur achieved effective tumor elimination through mild-photothermal chemotherapy without systemic toxicity. Overall, this work provides a new therapeutic modality paradigm for potential NSCLC treatment on the basis of synergistic therapies.
Collapse
Affiliation(s)
- Zhenying Diao
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- Dongguan Research Center for Biomedical Nano Engineering Technology Research, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Youcheng Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, PR China
| | - Yong Liu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- Dongguan Research Center for Biomedical Nano Engineering Technology Research, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Dou Zhang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- Dongguan Research Center for Biomedical Nano Engineering Technology Research, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Long Qiu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- Dongguan Research Center for Biomedical Nano Engineering Technology Research, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Jianbo Sun
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Qiaoyou Lu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- Dongguan Research Center for Biomedical Nano Engineering Technology Research, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| | - Yanlei Liu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| | - Daxiang Cui
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, PR China
| | - Ting Yin
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
- Dongguan Research Center for Biomedical Nano Engineering Technology Research, Guangdong Medical University, Dongguan, 523808, Guangdong, PR China
| |
Collapse
|
6
|
Cao JF, Zhang X, Xia Q, Hang K, Men J, Tian J, Liao D, Xia Z, Li K. Insights into curcumin's anticancer activity in pancreatic ductal adenocarcinoma: Experimental and computational evidence targeting HRAS, CCND1, EGFR and AKT1. Bioorg Chem 2025; 157:108264. [PMID: 39954354 DOI: 10.1016/j.bioorg.2025.108264] [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: 11/26/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE Curcumin, as a natural polyphenolic compound, possesses antitumor, antioxidant properties and anti-inflammatory. Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor, and there is a lack of molecular mechanisms and therapeutic options regarding relevant therapeutic agents. Therefore, we investigated the mechanism of curcumin inhibiting pancreatic cancer growth by modulating proliferation of cells and cellular metabolism. METHODS Bioinformatics analysis was involved in analyzing the intersecting targets of curcumin and pancreatic ductal adenocarcinoma. The effect of curcumin on proliferation of PANC-1 cells was tested by CCK-8, and total RNA from PANC-1 was also analysed by transcriptome sequencing. Molecular docking was involved in verifying binding stability of curcumin to protein targets. Molecular dynamics simulated and evaluated binding free energy, hydrogen bonds and root mean square fluctuation of the complex. RESULTS PPI, GO and KEGG were involved in screening and analysing key interacting protein targets. 40 μg/mL curcumin significantly inhibited the growth and proliferation of PANC-1. Transcriptome sequencing results showed gene expression of Cyclin D1 (CCND1), AKT serine/threonine kinase 1 (AKT1), HRas proto-oncogene (HRAS), epidermal growth factor receptor (EGFR) was significantly down-regulated by curcumin treatment. Result of molecular dynamics and molecular docking inhibited the free binding energies of CCND1/Curcumin, HRAS/Curcumin, AKT1/Curcumin and EGFR/Curcumin were -21.13 ± 3.41 kcal/mol, -21.84 ± 4.38 kcal/mol, -20.61 ± 1.82 kcal/mol and -27.37 ± 1.94 kcal/mol, respectively. CONCLUSION We found curcumin may not only regulate cell cycle progression in PDAC and apoptosis by down-regulating HRAS, thereby inhibiting CCND1 and its downstream signaling pathways, but also inhibit energy metabolism reprogramming, Ras-RAF-MEK-ERK and other downstream signalling pathways by down-regulating EGFR and AKT1, thereby affecting tumor cell metastasis, survival and proliferation.
Collapse
Affiliation(s)
- Jun-Feng Cao
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China
| | - Xiao Zhang
- Chengdu Medical College, Chengdu 610500 Sichuan, China
| | - Qingjie Xia
- Institute of Neurological Diseases, Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Kuan Hang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Jie Men
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China
| | - Jin Tian
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China
| | - Dunshui Liao
- Institute of Neurological Diseases, Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Zengliang Xia
- Institute of Neurological Diseases, Translation Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China
| | - Kezhou Li
- College of Medicine, Southwest Jiaotong University, Chengdu 610031 Sichuan, China; Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041 Sichuan, China.
| |
Collapse
|
7
|
Luo QY, Yang J, Di T, Xia ZF, Zhang L, Pan WT, Shi S, Yang LQ, Sun J, Qiu MZ, Yang DJ. The novel BCL-2/BCL-XL inhibitor APG-1252-mediated cleavage of GSDME enhances the antitumor efficacy of HER2-targeted therapy in HER2-positive gastric cancer. Acta Pharmacol Sin 2025; 46:1082-1096. [PMID: 39592733 PMCID: PMC11950313 DOI: 10.1038/s41401-024-01414-5] [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: 06/21/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
HER2-positive gastric cancer has a poor prognosis, with a high incidence of drug resistance and a lack of effective treatments for drug-resistant patients. The exploration of the mechanism of resistance to HER2-targeted therapy in HER2-positive gastric cancer and the identification of effective strategies to reverse it are urgently needed. In this study, we found that HER2-targeted agents upregulated the expression of GSDME and that the overexpression of GSDME attenuated the sensitivity of HER2-targeted agents. Furthermore, we observed that the BCL-2/BCL-XL inhibitor APG-1252 plus lapatinib promoted GSDME-mediated pyroptosis and exhibited remarkable antitumor activity both in vitro and in vivo. Mechanistically, APG-1252 combined with lapatinib synergistically induced GSDME-mediated pyroptosis in HER2-positive gastric cancer by activating caspase-dependent pathways and blocking the phospho-AKT/GSK-3β/MCL-1 signaling pathway. Our data indicated that the combination of lapatinib and APG-1252 had a synergistic antitumor effect on HER2-positive gastric cancer through the induction of caspase-3/GSDME-mediated apoptosis and pyroptosis.
Collapse
Affiliation(s)
- Qiu-Yun Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Clinical Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jing Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Tian Di
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zeng-Fei Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wen-Tao Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, 215000, China
| | - Shan Shi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Qiong Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jian Sun
- Department of Clinical Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Miao-Zhen Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Da-Jun Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, 215000, China.
| |
Collapse
|
8
|
Zheng S, Xue T, Xue C, Li S, Zao X, Li X, Cao X, Du H, Qi W, Seetoh WS, Wang W, Zhang P, Ye Y. Regulatory mechanisms of signaling pathways in liver cancer treatment with traditional Chinese medicine. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119386. [PMID: 39848414 DOI: 10.1016/j.jep.2025.119386] [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: 06/14/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese Medicine (TCM), as a longstanding therapeutic approach, offers unique advantages and potential in the treatment of liver cancer. Recent studies have highlighted its role in preventing liver cancer progression by modulating key signaling pathways. TCM's multi-component, multi-target, and multi-pathway mechanisms of action have garnered significant attention in the medical community for their ability to address complex diseases like liver cancer. AIM OF THE STUDY This review examines the current status and challenges in the application of TCM to regulate specific signaling pathways, including PI3K/Akt, NF-κB, TGF-β, Wnt/β-Catenin, and Notch, in liver cancer treatment. The goal is to further elucidate the critical roles of these pathways in liver cancer progression and provide new insights into the modern scientific interpretation of TCM. MATERIALS AND METHODS Literature was retrieved from PubMed and Web of Science databases using keywords such as "traditional Chinese medicine," "Chinese medicine," and "signaling pathway." The articles reviewed span from 2004 to 2024. RESULTS TCM demonstrates significant therapeutic and preventive effects in liver cancer by modulating signaling pathways involved in tumorigenesis. These pathways influence processes such as cell growth, invasion, proliferation, and inflammatory responses, contributing to the anti-cancer effects of TCM. CONCLUSION By modulating key signaling pathways such as PI3K/Akt, NF-κB, TGF-β, Wnt/β-Catenin, and Notch, TCM plays an important role in both the treatment and prevention of liver cancer, offering a promising therapeutic approach grounded in traditional practices and modern scientific understanding.
Collapse
Affiliation(s)
- Shihao Zheng
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Beijing University of Chinese Medicine, 100102, China.
| | - Tianyu Xue
- Hebei Provincial Hospital of Traditional Chinese Medicine, 050000, China
| | - Chengyuan Xue
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Beijing University of Chinese Medicine, 100102, China
| | - Size Li
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Beijing University of Chinese Medicine, 100102, China
| | - Xiaobin Zao
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China
| | - Xiaoke Li
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 100029, China
| | - Xu Cao
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 100029, China
| | - Hongbo Du
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 100029, China
| | - Wenying Qi
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Beijing University of Chinese Medicine, 100102, China
| | - Wei Song Seetoh
- Beijing University of Chinese Medicine, 100102, China; School of Biological Sciences, Nanyang Technological University, 637551, China
| | - Wei Wang
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Beijing University of Chinese Medicine, 100102, China
| | - Peng Zhang
- Department of Spleen and Gastroenterology, Dongfang Hospital, Beijing University of Chinese Medicine, 100078, China.
| | - Yongan Ye
- Department of Spleen and Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, 100007, China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 100029, China.
| |
Collapse
|
9
|
Li X, Huang Y, He Y, Ye A. Wogonoside alleviates the proliferation and promotes the apoptosis in liver cancer cells by regulating PI3K/Akt signaling pathway. Discov Oncol 2025; 16:244. [PMID: 40011302 DOI: 10.1007/s12672-025-01995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
INTRODUCTION Primary liver cancer is associated with high morbidity and mortality rate. In about 50% of cases, primary liver cancer is related to the phosphoinositide-3-kinase (PI3K)/Akt signaling pathway. Wogonoside is an active component extracted from Scutellaria baicalensis. Its antitumor effects in liver cancer are scarcely known. AIM This study explores the correlation between wogonoside and the PI3K/Akt signaling pathway in liver cancer in vitro. METHODS THLE-2 cells and HepG2 cells were treated with different concentrations of wogonoside to establish low-, medium- and high- dose groups, and the concentration of each dose group was determined by CCK-8 assay. Subsequent experiments were evaluated the viability, proliferation, invasion, wound healing, apoptosis rate of HepG2 cells, as well as the expression levels of relevant targets. In silico network pharmacology was performed to investigate the relationship between wogonoside and the PI3K/Akt signaling pathway, providing insights into the connection between wogonoside and liver cancer. RESULTS Compared with the control group, the viability, proliferation, invasion, migration and wound healing ability of wogonoside-treated HepG2 cells were significantly declined in a dose- and time-dependent manner. Wogonoside significantly reduced the relative expression level of Bcl-2/Bax relative protein. Wogonoside also decreased the relative protein expression of phospho-PI3K/PI3K and phospho-AKT/AKT and the mRNA levels of PI3K and AKT. In addition, potential key genes, biological processes, and pathways associated with the therapeutic effects of wogonoside on liver cancer were explored. CONCLUSION Wogonoside can alleviate the proliferation and promote the apoptosis of HepG2 cells, which may be related to the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xinfang Li
- Department of Clinical Lab, Zhuji Affiliated Hospital of Wenzhou Medical University, Shaoxing, 311800, Zhejiang Province, China
- Zhuji People's Hospital of Zhejiang Province, Shaoxing University, Shaoxing, 311899, Zhejiang Province, China
| | - Yitong Huang
- Department of Internal Medicine, Zhuji Maternal and Child Health Hospital, Shaoxing, 311899, Zhejiang, China
| | - Yibo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310003, Zhejiang Province, China
| | - Angzhi Ye
- Department of Clinical Lab, Zhuji Central Hospital, Zhugong Road 98, Shaoxing, 311800, Zhejiang Province, China.
- Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China.
| |
Collapse
|
10
|
Chen C, Wang Q, Liu J. Curcumin inhibits growth and triggers apoptosis in human castration-resistant prostate cancer cells via IGF-1/PI3K/Akt pathway. J Int Med Res 2025; 53:3000605231220807. [PMID: 39921429 PMCID: PMC11806470 DOI: 10.1177/03000605231220807] [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: 07/25/2023] [Accepted: 11/23/2023] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVE This study aimed to investigate the possible mechanism by which curcumin inhibits human prostate cancer (PCa) and castration-resistant prostate cancer (CRPC). METHODS CRPC cells were treated with curcumin and their viability was assessed by MTT assay and apoptosis was detected by annexinV/propidium iodide double-staining and terminal deoxynucleotidyl transferase dUTP nick-end labeling assays. Expression levels of insulin-like growth factor 1 receptor (IGF-1R) were determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting. Phosphoinositide 3-kinase (PI3K), Akt, and forkhead box protein O1 (FOXO1) expression and phosphorylation were assessed by western blotting. RESULTS The highly expressed PCa-related molecule IGF-1R was down-regulated in CRPC cells after curcumin treatment, as determined by RT-qPCR and western blotting. In addition, curcumin inhibited the tumor-related PI3K/Akt signaling pathway in CRPC cells. Moreover curcumin down-regulated the IGF-1/PI3K/Akt signaling pathway in tumors derived from CRPC cells. CONCLUSIONS These results demonstrated that curcumin inhibits growth and triggers apoptosis of human CRPC cells via the IGF-1/PI3K/Akt pathway, thus providing potential new therapeutic strategies for PCa and CRPC.
Collapse
Affiliation(s)
- Chao Chen
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Qiwu Wang
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Jiwen Liu
- Department of Urology, The General Hospital of Western Theater Command PLA, Chengdu, China
| |
Collapse
|
11
|
Karatayli E, Sadiq SC, Schattenberg JM, Grabbe S, Biersack B, Kaps L. Curcumin and Its Derivatives in Hepatology: Therapeutic Potential and Advances in Nanoparticle Formulations. Cancers (Basel) 2025; 17:484. [PMID: 39941855 PMCID: PMC11816286 DOI: 10.3390/cancers17030484] [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: 12/20/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Curcumin, a plant-derived polyphenol, shows promise in hepatology for treating both malignant and non-malignant liver diseases and a subset of extrahepatic cancers. Curcumin has hepatoprotective, anti-inflammatory, antifibrotic, and antiproliferative properties, as is evident in preclinical and clinical studies. This highlights its potential as an adjunct to established cancer therapies, especially in the context of hepatocellular carcinoma and secondary liver malignancies. Curcumin also demonstrates potential in metabolic dysfunction-associated steatotic liver disease (MASLD), owing to its antifibrotic and lipid-lowering effects. However, its clinical use is limited, relating to its poor bioavailability and rapid metabolism. Nanotechnology, including liposomal and polymeric carriers, alongside synthetic curcumin derivatives, offers strategies to enhance the bioavailability and pharmacokinetic properties. We propose to revisit the use of curcumin in nanoparticle preparations in chronic liver disease and summarize current evidence in this review article.
Collapse
Affiliation(s)
- Ersin Karatayli
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
| | - Shifana C. Sadiq
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
| | - Jörn M. Schattenberg
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany;
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, 95440 Bayreuth, Germany
| | - Leonard Kaps
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany; (E.K.); (S.C.S.); (J.M.S.)
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, 55128 Mainz, Germany;
| |
Collapse
|
12
|
Su J, Liu X, Zhao X, Ma H, Jiang Y, Wang X, Wang P, Zhao M, Hu X. Curcumin Inhibits the Growth of Hepatocellular Carcinoma via the MARCH1-mediated Modulation of JAK2/STAT3 Signaling. Recent Pat Anticancer Drug Discov 2025; 20:145-157. [PMID: 38243928 DOI: 10.2174/0115748928261490231124055059] [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: 05/17/2023] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Curcumin has been reported to have anti-hepatocellular carcinoma (HCC) effects, but the underlying mechanism is not well known. OBJECTIVES To investigate whether membrane-associated RING-CH 1 (MARCH1) is involved in the curcumin-induced growth suppression in HCC and its underlying molecular mechanism. A few recent patents for curcumin for cancer are also reviewed in this article. METHODS The effect of curcumin on growth inhibition of HCC cells was analyzed through in vitro and in vivo experiments, and the expression levels of MARCH1, Bcl-2, VEGF, cyclin B1, cyclin D1, and JAK2/STAT3 signaling molecules were measured in HCC cells and the xenograft tumors in nude mice. Cell transfection with MARCH1 siRNAs or expression plasmid was used to explore the role of MARCH1 in the curcumin-induced growth inhibition of HCC cells. RESULTS Curcumin inhibited cell proliferation, promoted apoptosis, and arrested the cell cycle at the G2/M phase in HCC cells with the decrease of Bcl-2, VEGF, cyclin B1, and cyclin D1 expression as well as JAK2 and STAT3 phosphorylation, resulting in the growth suppression of HCC cells. MARCH1 is highly expressed in HCC cells, and its expression was downregulated after curcumin treatment in a dose-dependent manner. The knockdown of MARCH1 by siRNA decreased the phosphorylation levels of JAK2 and STAT3 and inhibited the growth of HCC cells. In contrast, opposite results were observed when HCC cells overexpressed MARCH1. A xenograft tumor model in nude mice also showed that curcumin downregulated MARCH1 expression and decelerated the growth of transplanted HCC with the downregulation of JAK2/STAT3 signaling and functional molecules. The ADC value of MRI analysis showed that curcumin slowed down the progression of HCC. CONCLUSION Our results demonstrated that curcumin may inhibit the activation of JAK2/STAT3 signaling pathway by downregulating MARCH1 expression, resulting in the growth suppression of HCC. MARCH1 may be a novel target of curcumin in HCC treatment.
Collapse
Affiliation(s)
- Jiaqi Su
- Department of Imaging, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Xianbing Liu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Xiaoyue Zhao
- Department of Clinical Psychology, Yantai Affiliated Hospital of Binzhou Medial University, Yantai, 264100, Shandong, China
| | - Hongjie Ma
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Yuzhu Jiang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Xu Wang
- Department of Imaging, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Peiyuan Wang
- Department of Imaging, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Mingdong Zhao
- Department of Imaging, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, China
| |
Collapse
|
13
|
Dong J, Wang Z, Fei F, Jiang Y, Jiang Y, Guo L, Liu K, Cui L, Meng X, Li J, Wang H. Selenium Enhances the Growth of Bovine Endometrial Stromal Cells by PI3K/AKT/GSK-3β and Wnt/β-Catenin Pathways. Vet Sci 2024; 11:674. [PMID: 39729014 DOI: 10.3390/vetsci11120674] [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: 10/24/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
The bovine uterus is susceptible to bacterial infections after calving, particularly from Escherichia coli (E. coli), which often results in endometritis. Additionally, postpartum stress in cows can elevate cortisol levels in the body, inhibiting endometrial regeneration and reducing immune function, thereby further increasing the risk of infection. Selenium (Se) is a common feed additive in dairy farming, known for its anti-inflammatory and antioxidant effects. The aim of this study was to investigate the regulatory role of Se in the growth of bovine endometrial stromal cells (BESCs) under the conditions of LPS-induced inflammatory damage at high cortisol levels. BESCs were treated with 1, 2, 4 μM Se in combination with co-treatment of LPS and cortisol. The results indicated that LPS inhibited the cell viability and reduced the mRNA expression of CTGF, TGF-β1, and TGF-β3. Additionally, LPS increased apoptosis, hindered the cell cycle progression by blocking it in the G0/G1 phase, and suppressed the PI3K/AKT/GSK-3β and Wnt/β-catenin signaling pathways. Furthermore, increased concentrations of cortisol can exacerbate the impacts of LPS on cell proliferation and apoptosis. Conversely, the supplementation of Se promoted cell viability, increased the mRNA expression of TGF-β1 and TGF-β3, and enhanced cell cycle progression, while simultaneously repressing cell apoptosis as well as activating the PI3K/AKT/GSK-3β and Wnt/β-catenin signaling pathways. The above findings demonstrated that Se can promote cell proliferation, reduce cell apoptosis, and aid in the growth of BESCs damaged by LPS under high levels of cortisol. The potential mechanisms may be associated with the regulation of the PI3K/AKT/GSK-3β and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Zi Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Fan Fei
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Yeqi Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Yongshuai Jiang
- Guangling College, Yangzhou University, Yangzhou 225009, China
- School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Long Guo
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Kangjun Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Luying Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Xia Meng
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
14
|
Hanna DH, Al-Atmani AK, AlRashidi AA, Shafee EE. Camellia sinensis methanolic leaves extract: Phytochemical analysis and anticancer activity against human liver cancer cells. PLoS One 2024; 19:e0309795. [PMID: 39541389 PMCID: PMC11563400 DOI: 10.1371/journal.pone.0309795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/19/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The study's primary goal is to ascertain whether there is a relationship between the processed green tea methanolic extract's (GTME) phytochemical components and its potential effectiveness against human liver cancer cells. The GTME's phytochemical composition was identified using gas chromatography-mass spectrometry, and the extract's capacity to lower cellular proliferation and cause apoptosis in HepG2 cancerous liver cell lines was checked. RESULTS The findings of the gas chromatography-mass chromatogram showed that GTME included bioactive antioxidants and anticancer substances. Additionally, utilizing the MTT, comet assay, and acridine assay, GTME revealed a selective cytotoxic impact with a significant IC50 value (27.3 µg/ml) on HepG2 cells without any harmful effects on WI-38 healthy cells. Also, compared to untreated cells, the extract-treated HepG2 cells had an upsurge in the proportion of cells that have undergone apoptosis and displayed a comet nucleus, which is a sign of DNA damage. In addition, HepG2 cells treated with GTME revealed a stop in the G1 phase and sub-G1 apoptotic cells (37.32%) in a flow cytometry analysis. Furthermore, reactive oxygen species were shown to be responsible for HepG2 apoptosis, and the tested extract significantly reduced their levels in the treated cells. Lastly, compared to untreated cells in treated HepG2 cells, GTME significantly changed protein expression levels linked with cell cycle arrest in the G1 phase and apoptosis. CONCLUSION These findings provided information about the processes through which the GTME inhibited the growth of HepG2. Therefore, it has potential as an effective natural therapy for the treatment of human liver cancer. However, to validate these findings, animal models must be used for in vivo studies.
Collapse
Affiliation(s)
- Demiana H. Hanna
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| | - Ahlam K. Al-Atmani
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| | | | - E. El. Shafee
- Faculty of Science, Department of Chemistry, Cairo University, Giza, Egypt
| |
Collapse
|
15
|
Chen J, Cheng H, Bai C, Wang D, Fu J, Hao J, Wang Y, Xuewu Z. Sorbaria sorbifolia flavonoid derivative induces mitochondrial apoptosis in human hepatoma cells through Bclaf1. Front Pharmacol 2024; 15:1459520. [PMID: 39444606 PMCID: PMC11496133 DOI: 10.3389/fphar.2024.1459520] [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: 07/04/2024] [Accepted: 08/30/2024] [Indexed: 10/25/2024] Open
Abstract
4',5,7-Trihydroxy-8-methoxyflavone is an anticancer monomer component isolated from the traditional Chinese medicine Sorbaria sorbifolia. 4',5-Dihydroxy-7-piperazinemethoxy-8-methoxy flavonoids (DMF) with good solubility and anti-tumor effects was obtained by chemical modification in the early stage. This study explored the mechanism by which DMF regulates the mitochondrial apoptosis of human hepatoma cells through Bcl-2-associated transcription factor 1 (Bclaf1). DMF inhibited the proliferation of human hepatoma cells in a concentration- and time-dependent manner and induced cell mitochondrial apoptosis. The molecular docking and cell assay results demonstrated that DMF inhibits Bclaf1 expression by binding to its active site. Lentivirus transfection was used to construct cells with stable knockout and overexpression of Bclaf1, and a Hep3B xenograft model was constructed in nude mice. The mechanism by which DMF induced the mitochondrial apoptosis of human hepatoma cells through Bclaf1 was further verified in vitro and in vivo. These findings indicated that DMF induced human hepatoma cell mitochondrial apoptosis through Bclaf1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhang Xuewu
- College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
16
|
Zhou Y, Wang Z, Ren S, Li W. Mechanism of action of protopanaxadiol ginsenosides on hepatocellular carcinoma and network pharmacological analysis. CHINESE HERBAL MEDICINES 2024; 16:548-557. [PMID: 39606268 PMCID: PMC11589304 DOI: 10.1016/j.chmed.2024.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/24/2024] [Accepted: 06/18/2024] [Indexed: 11/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies globally, posing a major challenge to global health care. Protopanaxadiol ginsenosides (PDs) have been believed to significantly improve liver diseases. PDs, such as Rg3, have been developed as a new class of anti-cancer drugs. Ginsenosides Rb1, Rd, Rg3, and Rh2 exhibit effective anti-inflammatory and anti-tumor activities. Studies have confirmed that PDs could be used to treat HCC. However, the mechanism of action of PDs on HCC remains unclear. In the study, we reviewed the anti-HCC effects and mechanisms of PDs including Rb1, Rd, Rg3, Rg5, Rh2, Rk1, and Compound K (CK). Then, we searched for relevant targets of PDs and HCC from databases and enriched them for analysis. Subsequently, molecular docking was simulated to reveal molecular mechanisms. We found that PDs may treat HCC through multiple signaling pathways and related targets. PDs could inhibit the proliferation, invasion, and metastasis of HCC while promoting apoptosis and inducing differentiation. In conclusion, this review and network pharmacological analysis might offer a direction for in-depth research on related mechanisms. These insights will aid in the direction of further pharmacological studies and the development of safe and effective clinical drugs.
Collapse
Affiliation(s)
- Yue Zhou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Shen Ren
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
17
|
Jiang Y, Hui D, Pan Z, Yu Y, Liu L, Yu X, Wu C, Sun M. Curcumin promotes ferroptosis in hepatocellular carcinoma via upregulation of ACSL4. J Cancer Res Clin Oncol 2024; 150:429. [PMID: 39311951 PMCID: PMC11420324 DOI: 10.1007/s00432-024-05878-0] [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: 04/26/2024] [Accepted: 07/01/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Ferroptosis, a novel iron-ion-dependent metabolic cell death mode with lipid peroxides as the main driving substrate, plays an irreplaceable role in the development and preventive treatment of hepatocellular carcinoma. Curcumin has potent pharmacological anti-tumor effects. AIM OF THE STUDY We aimed to evaluate the ex vivo and in vivo cancer inhibitory activity of curcumin and its specific mechanism of action. MATERIALS AND METHODS We used the hepatocellular carcinoma cell lines HepG2 and SMMC7721 to assess the direct inhibition of hepatocellular carcinoma proliferation by curcumin in vitro and a tumor xenograft model to evaluate the in vivo cancer inhibitory effect of curcumin. RESULTS In this study, we found that ferroptosis's inhibitors specifically reversed the curcumin-induced cell death pattern in HCC. After curcumin intervention, there was a substantial increase in MDA levels and iron ion levels, and a decrease in intracellular GSH levels. Meanwhile, the expression of GPX4 and SLC7A11 was significantly reduced at the protein levels, while ACSL4 and PTGS2 expression was significantly increased. CONCLUSIONS This study showed that curcumin significantly decreased the proliferation of HCC cells and significantly increased the sensitivity of ferroptosis. These results suggest that ACSL4 is a viable target for curcumin-induced ferroptosis in treating HCC.
Collapse
Affiliation(s)
- Yulang Jiang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Dengcheng Hui
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Ziyang Pan
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Yongxin Yu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Lu Liu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Xiaofan Yu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Chao Wu
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China
| | - Mingyu Sun
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.528 Zhangheng Road, Zhangjiang Hi-Tech Park, Shanghai, 201203, China.
| |
Collapse
|
18
|
Jin Q, Jiao W, Lian Y, Chitrakar B, Sang Y, Wang X. Study on antihepatocellular carcinoma effect of 6-shogaol and curcumin through network-based pharmacological and cellular assay. Front Pharmacol 2024; 15:1367417. [PMID: 39224778 PMCID: PMC11368042 DOI: 10.3389/fphar.2024.1367417] [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: 01/08/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Background Hepatocellular carcinoma currently has the third highest mortality rate in the world. Patients with hepatocellular carcinoma are on the rise and at a younger age, but research into the pharmacological effects of cancer is mostly single-component, and natural plant products can have additive or synergistic effects that can better amplify the effects of intervention in cancer. Aim To evaluate the synergistic therapeutic effects of 6-shogaol and curcumin against hepatocellular carcinoma line HepG2 cells. Methods In this study, a network pharmacology approach was used to predict and validate the mol ecular targets and pathways of the hepatocellular carcinoma (HCC) of 6-shogaol and curcumin in combination and to investigate their mechanism of action. The results were also validated by cellular assays. HepG2 cells were treated with 6-shogaol and curcumin as well as the combination of the two. The combination index of 6-shogaol and curcumin in HepG2 cells was calculated using Compusyn software according to the Chou-Talalay equation. The synergistic anti-cancer effect was next investigated by MTT assay, apoptosis assay and cell cycle assay. The combined anti-hepatocellular carcinoma effect of the Ras-mediated PI3K/AKT and MAPK signalling pathways was analysed using protein blotting assays. Results A network pharmacology-based screening identified 72 core targets of 6-curcumin and curcumin in hepatocellular carcinoma, and predicted that the main signalling pathway is the Ras signalling pathway. The anti-cancer effects of 6-shogaol and curcumin were validated in cell-based assays and the optimal synergistic concentrations of 5 μmoL/L for 6-shogaol and 30 μmoL/L for curcumin were determined. 6-shogaol and curcumin synergistically blocked the cell cycle in the G2/M phase and promoted apoptosis. Immunoblot analysis confirmed for the first time the combined action of both in down-regulating the Ras-mediated PI3K/AKT and MAPK signaling pathways. In addition, 6-shogaol and curcumin acting together downregulated Cyclin-B, CDK-1, Bcl-2, and upregulated BAX. Conclusion 6-shogaol and curcumin act synergistically to alter the morphology of hepatocellular carcinoma cells, block the cell cycle in the G2/M phase, inhibit proliferation and division, and effectively promote late apoptosis. The combined action of these two components provides a theoretical basis for the further development of novel anti-liver cancer products.
Collapse
Affiliation(s)
- Qiuxia Jin
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Wenya Jiao
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Yunhe Lian
- Chenguang Biotechnology Group Co., Ltd., Handan, China
| | - Bimal Chitrakar
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Yaxin Sang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| | - Xianghong Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
19
|
Guo J, Jiang X, Lian J, Li H, Zhang F, Xie J, Deng J, Hou X, Du Z, Hao E. Evaluation of the effect of GSK-3β on liver cancer based on the PI3K/AKT pathway. Front Cell Dev Biol 2024; 12:1431423. [PMID: 39156976 PMCID: PMC11327086 DOI: 10.3389/fcell.2024.1431423] [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: 05/11/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
The PI3K/AKT/GSK-3β signaling pathway plays a pivotal role in numerous physiological and pathological processes, including cell proliferation, apoptosis, differentiation, and metabolic regulation. Aberrant activation of the PI3K/AKT pathway is intricately linked to development of tumor. GSK-3β, belonging to the serine/threonine protein kinase family, is crucial in the pathogenesis of liver cancer. As a key rate-limiting enzyme in the glucose metabolism pathway, GSK-3β significantly impacts the growth, proliferation, metastasis, and apoptosis of liver cancer cells. It is also implicated in chemotherapy resistance. Elevated expression of GSK-3β diminishes the sensitivity of liver cancer cells to chemotherapeutic agents, thereby playing a substantial role in the development of drug resistance. Consequently, targeting of GSK-3β, particularly within the PI3K/AKT signaling pathway, is regarded as a promising therapeutic strategy for liver cancer. The precise identification and subsequent modulation of this pathway represent a substantial potential for innovative clinical interventions in the management of liver cancer.
Collapse
Affiliation(s)
- Jiageng Guo
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Xinya Jiang
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jing Lian
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Huaying Li
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Fan Zhang
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Jinling Xie
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaotao Hou
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
20
|
Yashmi F, Fakhri S, Shiri Varnamkhasti B, Amin MN, Khirehgesh MR, Mohammadi-Noori E, Hosseini M, Khan H. Defining the mechanisms behind the hepatoprotective properties of curcumin. Arch Toxicol 2024; 98:2331-2351. [PMID: 38837048 DOI: 10.1007/s00204-024-03758-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/09/2024] [Indexed: 06/06/2024]
Abstract
As a critical cause of human dysfunctionality, hepatic failure leads to approximately two million deaths per year and is on the rise. Considering multiple inflammatory, oxidative, and apoptotic mechanisms behind hepatotoxicity, it urges the need for finding novel multi-targeting agents. Curcumin is a phenolic compound with anti-inflammatory, antioxidant, and anti-apoptotic roles. Curcumin possesses auspicious health benefits and protects against several diseases with exceptional safety and tolerability. This review focused on the hepatoprotective mechanisms of curcumin. The need to develop novel delivery systems of curcumin (e.g., nanoparticles, self-micro emulsifying, lipid-based colloids, solid lipid nanoparticles, cyclodextrin inclusion, phospholipid complexes, and nanoemulsions) is also considered.
Collapse
Affiliation(s)
- Farinam Yashmi
- Department of Pharmacy, Acibadem University, Istanbul, Turkey
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Behrang Shiri Varnamkhasti
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammed Namiq Amin
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Khirehgesh
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Mohammadi-Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahsa Hosseini
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
| |
Collapse
|
21
|
Rahman MA, Rakib-Uz-Zaman SM, Chakraborti S, Bhajan SK, Gupta RD, Jalouli M, Parvez MAK, Shaikh MH, Hoque Apu E, Harrath AH, Moon S, Kim B. Advancements in Utilizing Natural Compounds for Modulating Autophagy in Liver Cancer: Molecular Mechanisms and Therapeutic Targets. Cells 2024; 13:1186. [PMID: 39056768 PMCID: PMC11274515 DOI: 10.3390/cells13141186] [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: 04/16/2024] [Revised: 06/30/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Autophagy, an intrinsic catabolic mechanism that eliminates misfolded proteins, dysfunctional organelles, and lipid droplets, plays a vital function in energy balance and cytoplasmic quality control, in addition to maintaining cellular homeostasis. Liver cancer such as hepatocellular carcinoma (HCC) is one of the most common causes of cancer deaths globally and shows resistance to several anticancer drugs. Despite the rising incidence and poor prognosis of malignant HCC, the underlying molecular mechanisms driving this aggressive cancer remain unclear. Several natural compounds, such as phytochemicals of dietary and non-dietary origin, affect hepatocarcinogenesis signaling pathways in vitro and in vivo, which may help prevent and treat HCC cells. Current HCC cells treatments include chemotherapy, radiation, and surgery. However, these standard therapies have substantial side effects, and combination therapy enhances side effects for an acceptable therapeutic benefit. Therefore, there is a need to develop treatment strategies for HCC cells that are more efficacious and have fewer adverse effects. Multiple genetic and epigenetic factors are responsible for the HCC cells to become resistant to standard treatment. Autophagy contributes to maintain cellular homeostasis, which activates autophagy for biosynthesis and mitochondrial regulation and recycling. Therefore, modifying autophagic signaling would present a promising opportunity to identify novel therapies to treat HCC cells resistant to current standard treatments. This comprehensive review illustrates how natural compounds demonstrate their anti-hepatocellular carcinoma function through autophagy.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - S M Rakib-Uz-Zaman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Somdeepa Chakraborti
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
| | - Sujay Kumar Bhajan
- Department of Biotechnology & Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science & Technology University, Gopalganj 8100, Bangladesh;
| | - Rajat Das Gupta
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA;
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | | | - Mushfiq H. Shaikh
- Department of Otolaryngology-Head & Neck Surgery, Western University, London, ON N6A 4V2, Canada;
| | - Ehsanul Hoque Apu
- Department of Biomedical Sciences, College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA;
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
- Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1–5 Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea;
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1–5 Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea;
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
22
|
Guo Z, Meng Y, Zhou S, Li J, Li X, Feng R, Zou Y, Liao W, Wu W, Xu M, Zeng X, Zhao W, Zhong H. Atomic force microscopy correlates mechanical and electrical properties of HepG2 cells with curcumin concentration. J Pharm Biomed Anal 2024; 243:116107. [PMID: 38489959 DOI: 10.1016/j.jpba.2024.116107] [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: 12/28/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Hepatocellular carcinoma (HCC) is a highly prevalent cancer with a significant impact on human health. Curcumin, a natural compound, induces cytoskeletal changes in liver cancer cells and modifies the distribution of lipids, proteins, and polysaccharides on plasma membranes, affecting their mechanical and electrical properties. In this study, we used nanomechanical indentation techniques and Kelvin probe force microscopy (KPFM) based on atomic force microscopy (AFM) to investigate the changes in surface nanomechanical and electrical properties of nuclear and cytoplasmic regions of HepG2 cells in response to increasing curcumin concentrations. CCK-8 assays and flow cytometry results demonstrated time- and concentration-dependent inhibition of HepG2 cell proliferation by curcumin. Increasing curcumin concentration led to an initial increase and then decrease in the mechanical properties of nuclear and cytoplasmic regions of HepG2 cells, represented by the Young's modulus (E), as observed through nanoindentation. KPFM measurements indicated decreasing trends in both cell surface potential and height. Fluorescence microscopy results indicated a positive correlation between curcumin concentration and phosphatidylserine translocation from the inner to the outer membrane, which influenced the electrical properties of HepG2 cells. This study provides valuable insights into curcumin's mechanisms against cancer cells and aids nanoscale evaluation of therapeutic efficacy and drug screening.
Collapse
Affiliation(s)
- Zeling Guo
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Yu Meng
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Shang Zhou
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Jiangting Li
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Xinyu Li
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Rongrong Feng
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Yulan Zou
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Wenchao Liao
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Weiting Wu
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Mingjing Xu
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China
| | - Xiangfu Zeng
- The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, People's Republic of China.
| | - Weidong Zhao
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China.
| | - Haijian Zhong
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, People's Republic of China.
| |
Collapse
|
23
|
Ebrahimi M, Dabbagh A, Madadi F. Propofol-induced hippocampal Neurotoxicity: A mitochondrial perspective. Brain Res 2024; 1831:148841. [PMID: 38428475 DOI: 10.1016/j.brainres.2024.148841] [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: 01/19/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Propofol is a frequently used anesthetic. It can induce neurodegeneration and inhibit neurogenesis in the hippocampus. This effect may be temporary. It can, however, become permanent in vulnerable populations, such as the elderly, who are more susceptible to Alzheimer's disease, and neonates and children, whose brains are still developing and require neurogenesis. Current clinical practice strategies have failed to provide an effective solution to this problem. In addition, the molecular mechanism of this toxicity is not fully understood. Recent advances in molecular research have revealed that apoptosis, in close association with mitochondria, is a crucial mechanism through which propofol contributes to hippocampal toxicity. Preventing the toxicity of propofol on the hippocampus has shown promise in in-vivo, in-vitro, and to a lesser extent human studies. This study seeks to provide a comprehensive literature review of the effects of propofol toxicity on the hippocampus via mitochondria and to suggest translational suggestions based on these molecular results.
Collapse
Affiliation(s)
- Moein Ebrahimi
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Dabbagh
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Firoozeh Madadi
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Anesthesiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Iksen, Witayateeraporn W, Hardianti B, Pongrakhananon V. Comprehensive review of Bcl-2 family proteins in cancer apoptosis: Therapeutic strategies and promising updates of natural bioactive compounds and small molecules. Phytother Res 2024; 38:2249-2275. [PMID: 38415799 DOI: 10.1002/ptr.8157] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/04/2024] [Accepted: 01/29/2024] [Indexed: 02/29/2024]
Abstract
Cancer has a considerably higher fatality rate than other diseases globally and is one of the most lethal and profoundly disruptive ailments. The increasing incidence of cancer among humans is one of the greatest challenges in the field of healthcare. A significant factor in the initiation and progression of tumorigenesis is the dysregulation of physiological processes governing cell death, which results in the survival of cancerous cells. B-cell lymphoma 2 (Bcl-2) family members play important roles in several cancer-related processes. Drug research and development have identified various promising natural compounds that demonstrate potent anticancer effects by specifically targeting Bcl-2 family proteins and their associated signaling pathways. This comprehensive review highlights the substantial roles of Bcl-2 family proteins in regulating apoptosis, including the intricate signaling pathways governing the activity of these proteins, the impact of reactive oxygen species, and the crucial involvement of proteasome degradation and the stress response. Furthermore, this review discusses advances in the exploration and potential therapeutic applications of natural compounds and small molecules targeting Bcl-2 family proteins and thus provides substantial scientific information and therapeutic strategies for cancer management.
Collapse
Affiliation(s)
- Iksen
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacy, Sekolah Tinggi Ilmu Kesehatan Senior Medan, Medan, Indonesia
| | - Wasita Witayateeraporn
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Besse Hardianti
- Laboratory of Pharmacology and Clinical Pharmacy, Faculty of Health Sciences, Almarisah Madani University, South Sulawesi, Indonesia
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
25
|
Cao JL, Li SM, Tang YJ, Hou WS, Wang AQ, Li TZ, Jin CH. Network pharmacology analysis and experimental verification of the antitumor effect and molecular mechanism of isocryptomerin on HepG2 cells. Drug Dev Res 2024; 85:e22165. [PMID: 38400652 DOI: 10.1002/ddr.22165] [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: 11/20/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
Isocryptomerin (ISO) is a flavonoid isolated from the natural medicine Selaginellae Herba, which has various pharmacological activities. This study investigated the antitumor effect and underlying molecular mechanism of ISO on hepatocellular carcinoma (HCC) HepG2 cells. The cell viability assay revealed that ISO has a considerable killing effect on HCC cell lines. The apoptosis assay showed that ISO induced mitochondria-dependent apoptosis through the Bad/cyto-c/cleaved (cle)-caspase-3/cleaved (cle)-PARP pathway. The network pharmacological analysis found 13 key target genes, and epidermal growth factor receptor (EGFR), AKT, mitogen-activated protein kinase (MAPK), and reactive oxygen species (ROS) signaling pathways were strongly associated with ISO against HCC. Further verification of the results showed that ISO induced apoptosis by increasing p-p38 and p-JNK expression and decreasing p-EGFR, p-SRC, p-ERK, and p-STAT3 expression. Furthermore, ISO induced G0/G1 phase arrest by downregulating p-AKT, Cyclin D, and CDK 4 expression and upregulating p21 and p27 expression in HepG2 cells. Moreover, ISO inhibited HepG2 cell migration by decreasing p-GSK-3β, β-catenin, and N-cadherin expression and increasing E-cadherin expression. Additionally, ISO promoted ROS accumulation in HepG2 cells, and ISO-induced apoptosis, arrest cell cycle, and inhibition of migration were reversed by an ROS scavenger, N-acetyl- l-cysteine. Overall, ISO induced cell apoptosis and cell cycle arrest and inhibited cell migration by ROS-mediated EGFR, AKT, and MAPK signaling pathways in HepG2 cells.
Collapse
Affiliation(s)
- Jing-Long Cao
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shu-Mei Li
- Hemodialysis Center, Daqing Oilfield General Hospital, Daqing, China
| | - Yan-Jun Tang
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wen-Shuang Hou
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - An-Qi Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Tian-Zhu Li
- Department of Molecular Biology, College of Basic Medical Science, Chifeng University, Chifeng, China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, China
- National Coarse Cereals Engineering Research Center, Daqing, China
| |
Collapse
|
26
|
Razali NSC, Lam KW, Rajab NF, Jamal ARA, Kamaludin NF, Chan KM. Curcumin piperidone derivatives induce caspase-dependent apoptosis and suppress miRNA-21 expression in LN-18 human glioblastoma cells. Genes Environ 2024; 46:4. [PMID: 38303058 PMCID: PMC10832295 DOI: 10.1186/s41021-023-00297-y] [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: 07/22/2023] [Accepted: 12/27/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Previously, we have reported on the two curcuminoid analogues with piperidone derivatives, namely FLDP-5 and FLDP-8 have more potent anti-proliferative and anti-migration effects than curcumin. In this study, we further investigated the mode of cell death and the mechanism involved in the cell death process induced by these analogues on human glioblastoma LN-18 cells. RESULTS The FLDP-5 and FLDP-8 curcuminoid analogues induced LN-18 cell death through apoptosis in a concentration-dependent manner following 24 h of treatment. These analogues induced apoptosis in LN-18 cells through significant loss of mitochondrial mass and mitochondrial membrane potential (MMP) as early as 1-hour of treatment. Interestingly, N-acetyl-l-cysteine (NAC) pretreatment did not abolish the apoptosis induced by these analogues, further confirming the cell death process is independent of ROS. However, the apoptosis induced by the analogues is caspases-dependent, whereby pan-caspase pretreatment inhibited the curcuminoid analogues-induced apoptosis. The apoptotic cell death progressed with the activation of both caspase-8 and caspase-9, which eventually led to the activation of caspase-3, as confirmed by immunoblotting. Moreover, the existing over-expression of miRNA-21 in LN-18 cells was suppressed following treatment with both analogues, which suggested the down-regulation of the miRNA-21 facilitates the cell death process. CONCLUSION The FLDP-5 and FLDP-8 curcuminoid analogues downregulate the miRNA-21 expression and induce extrinsic and intrinsic apoptotic pathways in LN-18 cells.
Collapse
Affiliation(s)
- Nur Syahirah Che Razali
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| | - Kok Wai Lam
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| | - Nor Fadilah Rajab
- Center for Health Ageing and Wellness Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| | - A Rahman A Jamal
- UKM Medical Molecular Biology Institute, UKM Medical Centre, Cheras, 56000, Malaysia
| | - Nurul Farahana Kamaludin
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| | - Kok Meng Chan
- Center for Toxicology and Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia.
- Product Stewardship and Toxicology, Group Health, Safety and Environment (GHSE), Petroliam Nasional Berhad (PETRONAS), Kuala Lumpur, 50088, Malaysia.
| |
Collapse
|
27
|
Zhang S, Wang Y, Wang B, Zeng Y, Li J, Wang X, Hu C, Weng Z, Wang Z. Effect of curcumin on malignant hepatocytes and mitochondria studied using atomic force microscopy. Micron 2024; 177:103573. [PMID: 38043195 DOI: 10.1016/j.micron.2023.103573] [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: 10/07/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023]
Abstract
Mitochondria are emerging as potential targets for the cancer treatment. In this study, the effects of curcumin on the activity, migration, and mitochondrial membrane potential (MMP) of malignant hepatocytes (SMMC-7721 cells) were determined using cell viability, migration, and MMP assays. Changes in the morphology and biomechanics of SMMC-7721 cells and their mitochondria were studied using both optical microscopy and atomic force microscopy (AFM). The cell survival rate, migration and MMP depended on the concentration of curcumin. Optical microscopy studies showed that curcumin altered the cell morphology. AFM studies showed that the changes in the morphology and nanomechanics of SMMC-7721 cells and their mitochondria, were induced by curcumin. As the concentration of curcumin increased, the cell length, width, and adhesion decreased, but the height, roughness and Young's modulus increased. In contrast, the mitochondrial length, width, height and roughness increased, but the adhesion and Young's modulus decreased. There was a close relationship between mitochondria and cells in terms of function, morphology and biomechanics. This study shows the effects of curcumin on SMMC-7721 cells and their mitochondria from biology and biophysics perspectives. The findings aid in comprehensively understanding the interactions between mitochondria and malignant hepatocytes.
Collapse
Affiliation(s)
- Shengli Zhang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Ying Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Bowei Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Yi Zeng
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Jiani Li
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Xingyue Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Cuihua Hu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China
| | - Zhankun Weng
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, China.
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528400, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; JR3CN & IRAC, University of Bedfordshire, Luton LU1 3JU, UK.
| |
Collapse
|
28
|
Zhang B, Zhou B, Huang G, Huang J, Lin X, Li Z, Lian Y, Huang Q, Ye Y. Nitidine chloride inhibits G2/M phase by regulating the p53/14-3-3 Sigma/CDK1 axis for hepatocellular carcinoma treatment. Heliyon 2024; 10:e24012. [PMID: 38283241 PMCID: PMC10818205 DOI: 10.1016/j.heliyon.2024.e24012] [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: 10/22/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Background Liver cancer had become the sixth most common cancer. Nitidine chloride (NC) has demonstrated promising anti-HCC properties; however, further elucidation of its mechanism of action is necessary. Methods The anti-HCC targets of NC were identified through the utilization of multiple databases and ChIPs data analysis. The GO and KEGG analyses to determine the specific pathway affected by NC. The Huh 7 and Hep G2 cells were subjected to a 24-h treatment with NC, followed by evaluating the impact of NC on cell proliferation and cell cycle. The involvement of the p53/14-3-3 Sigma/CDK1 axis in HCC cells was confirmed by qPCR and WB analysis of the corresponding genes and proteins. Results The GO and KEGG analysis showed the targets were related to cell cycle and p53 signaling pathways. In vitro experiments showed that NC significantly inhibited the proliferation of HCC cells and induced G2/M phase arrest. In addition, qPCR and WB experiments showed that the expression of p53 in HCC cells increased after NC intervention, while the expression of 14-3-3 Sigma and CDK1 decreased. Conclusion NC can inhibit the proliferation of HCC cells and induce G2/M cell cycle arrest, potentially by regulating the p53/14-3-3 Sigma/CDK1 axis.
Collapse
Affiliation(s)
- Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Bo Zhou
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Guihong Huang
- Department of Pharmacy, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541199, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin, Guangxi, 541199, China
| | - Jing'an Huang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Xiaoxin Lin
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Zonghuai Li
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Yuanchu Lian
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Qiujie Huang
- Guangxi University of Chinese Medicine, Teaching Experiment and Training Center, Nanning, China
| | - Yong Ye
- School of Pharmacy, Guangxi Medical University, Guangxi, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, China
| |
Collapse
|
29
|
Wu Y, Zhang F, Xu P, Li P. Brucine Inhibits Proliferation of Pancreatic Ductal Adenocarcinoma through PI3K/AKT Pathway-induced Mitochondrial Apoptosis. Curr Cancer Drug Targets 2024; 24:749-759. [PMID: 38310464 DOI: 10.2174/0115680096274284231116104554] [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: 07/26/2023] [Revised: 09/23/2023] [Accepted: 10/03/2023] [Indexed: 02/05/2024]
Abstract
INTRODUCTION The purpose of this research was to settle the role of brucine in pancreatic ductal adenocarcinoma (PDAC) and the mechanisms involved. METHODS The findings of this study suggest that brucine exerts inhibitory effects on cell growth, clonogenicity, and invasive potential of Panc02 and Mia Paca-2 cells. These effects may be linked to an increase in apoptotic-prone cell population. RESULTS Gene sequencing data suggests that these effects are mediated through the induction of apoptosis. Experimental evidence further supports the notion that brucine reduces mitochondrial membrane potential and upregulates Bax expression while downregulating Bcl-2 expression. These effects are believed to be a result of brucine-mediated suppression of PI3K/Akt activity, which serves as a regulatory factor of mTOR, Bax, and Bcl-2. Suppression of PI3K activity enhances the tumor-suppressing effects of brucine. CONCLUSION Overall, these findings suggest that brucine has therapeutic potential as a remedy option for PDAC.
Collapse
Affiliation(s)
- You Wu
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, Anhui, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, 230022, Hefei, Anhui, China
| | - Fenglin Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, Anhui, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, 230022, Hefei, Anhui, China
| | - Panling Xu
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, Anhui, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, 230022, Hefei, Anhui, China
| | - Ping Li
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, Anhui, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, 230022, Hefei, Anhui, China
| |
Collapse
|
30
|
Srinivas AN, Suresh D, Chidambaram SB, Santhekadur PK, Kumar DP. Apoptosis antagonizing transcription factor-mediated liver damage and inflammation to cancer: Therapeutic intervention by curcumin in experimental metabolic dysfunction associated steatohepatitis-hepatocellular carcinoma. J Cell Physiol 2024; 239:135-151. [PMID: 37942831 DOI: 10.1002/jcp.31151] [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: 07/27/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
In tandem with the expanding obesity pandemic, the prevalence of metabolic dysfunction associated steatohepatitis (MASH, formerly known as NASH)- driven hepatocellular carcinoma (HCC) is predicted to rise globally, creating a significant need for therapeutic interventions. We previously identified the upregulation of apoptosis antagonizing transcription factor (AATF), which is implicated in facilitating the progression from MASH to HCC. The objective of this study was to examine whether the intervention of curcumin could alleviate AATF-mediated MASH, inhibit tumor growth, and elucidate the underlying mechanism. A preclinical murine model mimicking human MASH-HCC was employed, subjecting mice to either a chow diet normal water (CDNW) or western diet sugar water (WDSW) along with very low dose of carbon tetrachloride (CCl4 - 0.2 μL/g, weekly). Mice receiving curcumin (CUR) alongside WDSW/CCl4 exhibited significant improvements, including reduced liver enzymes, dyslipidemia, steatosis, inflammation, and hepatocellular ballooning. Curcumin treatment also suppressed hepatic expression of inflammatory, fibrogenic, and oncogenic markers. Of note, there was a significant reduction in the expression of AATF upon curcumin treatment in WDSW/CCl4 mice and human HCC cells. In contrast, curcumin upregulated Kruppel-like factor 4 (KLF4) in MASH liver and HCC cells, which is known to downregulate sp1 (specificity protein-1) expression. Thus, curcumin treatment effectively inhibited the progression of MASH to HCC by downregulating the expression of AATF via the KLF4-Sp1 signaling pathway. These preclinical findings establish a novel molecular connection between curcumin and AATF in reducing hepatocarcinogenesis, and provide a strong rationale for the development of curcumin as a viable treatment for MASH-HCC in humans.
Collapse
Affiliation(s)
- Akshatha N Srinivas
- Department of Biochemistry, CEMR lab, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Diwakar Suresh
- Department of Biochemistry, CEMR lab, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Saravana B Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Prasanna K Santhekadur
- Department of Biochemistry, CEMR lab, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Divya P Kumar
- Department of Biochemistry, CEMR lab, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| |
Collapse
|
31
|
Kotb RR, Afifi AM, El-Houseini ME, Ezz-Elarab M, Basalious EB, Omran MM, Abdellateif MS. The potential immuno-stimulating effect of curcumin, piperine, and taurine combination in hepatocellular carcinoma; a pilot study. Discov Oncol 2023; 14:169. [PMID: 37704828 PMCID: PMC10499730 DOI: 10.1007/s12672-023-00785-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND This is a phase II clinical trial to investigate the immunotherapeutic effect of Curcumin, Piperine, and Taurine (CPT) combination in hepatocellular carcinoma (HCC). METHODS Twenty-six HCC patients aged (50-80 years) were recruited for administration of a daily dose of 5 g of curcumin, 50 mg of piperine, and 500 mg of taurine divided into three doses for successive 3 months. The three components (CPT) were prepared in one capsule. Patients were assessed after each month (cycle) for the plasma levels of CD4, CD8, CD25, Interleukins-2 (IL-2), IL-6, IL-12, Interferon-gamma (IFN- γ), Lactate dehydrogenase (LDH), and Vascular endothelial growth factor (VEGF), FOXP3 mRNA, and miRNA 21. RESULTS There was a significant increase in the plasma levels of CD4 and CD8, while a significant decrease in the CD25 level after the second and third cycles compared to the baseline level [P < 0.001 for both]. Also, there was a significant increase in the plasma levels of IL-2, IL-12, and IFN-γ [ P = 0.001, P = 0.006, and P = 0.029; respectively], while there was a significant decrease in IL-6, VEGF-α, LDH, and Alpha-fetoprotein (AFP) after CPT administration compared to the baseline levels [P < 0.001, P < 0.001, P = 0.020, and P = 0.004; respectively]. The expression level of miRNA-21 was significantly decreased after CPT administration compared to the baseline level [5.5±0.88, 4.1±0.78, 3±0.75, and 2.5±0.76; respectively, P<0.001]. Though there was a noticeable decrease in the FOXP3 expression after each cycle, however, it didn't reach a significant level [5.3±0.8, 4.2±0.76, 3.2±0.67, and 2.5±0.79; respectively, P=0.184]. CONCLUSION CPT could exhibit a potential immune-stimulating effect in HCC patients. The current trial had been registered at the National Hepatology and Tropical Medicine Research Institute (NHTMRI), with a registration number of NHTMRI-IRB 2-21 on 5th January 2021.
Collapse
Affiliation(s)
- Raghda R Kotb
- Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Ahmed M Afifi
- Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Motawa E El-Houseini
- Medical Biochemistry and molecular biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Ezz-Elarab
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Emad B Basalious
- Pharmaceutics and Industrial Pharmacy department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Mervat M Omran
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Biological science division, University of Chicago, Chicago, IL, USA
| | - Mona S Abdellateif
- Medical Biochemistry and molecular biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
32
|
Wang W, Li M, Wang L, Chen L, Goh BC. Curcumin in cancer therapy: Exploring molecular mechanisms and overcoming clinical challenges. Cancer Lett 2023; 570:216332. [PMID: 37541540 DOI: 10.1016/j.canlet.2023.216332] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Cancer poses a significant global health burden, necessitating the widespread use of chemotherapy and radiotherapy as conventional frontline interventions. Although targeted therapy and immunotherapy have shown remarkable advancements, the challenges of resistance development and severe side effects persist in cancer treatment. Consequently, researchers have actively sought more effective alternatives with improved safety profiles. In recent years, curcumin, a natural polyphenolic phytoalexin, has garnered considerable attention due to its broad spectrum of biological effects. This concise review provides valuable insights into the role of curcumin in cancer therapy, with a focus on elucidating its molecular mechanisms in inducing programmed cell death of tumor cells and suppressing tumor cell metastasis potential. Additionally, we discuss the challenges associated with the clinical application of curcumin and explore current endeavors aimed at overcoming these limitations. By shedding light on the promising potential of curcumin, this review contributes to the advancement of cancer treatment strategies.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Mingqin Li
- Department of Medical Cardiology, Zhongxiang TCM Hospital of Hubei, Zhongxiang, 431900, China
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; National University Cancer Institute, National University of Singapore, 119074, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| | - Lu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; National University Cancer Institute, National University of Singapore, 119074, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore; Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore, Singapore
| |
Collapse
|
33
|
Shao H, Chen J, Li A, Ma L, Tang Y, Chen H, Chen Y, Liu J. Salvigenin Suppresses Hepatocellular Carcinoma Glycolysis and Chemoresistance Through Inactivating the PI3K/AKT/GSK-3β Pathway. Appl Biochem Biotechnol 2023; 195:5217-5237. [PMID: 37129745 PMCID: PMC10354167 DOI: 10.1007/s12010-023-04511-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/03/2023]
Abstract
Salvigenin is a Trimethoxylated Flavone enriched in Scutellariae Barbatae Herba and Scutellariae Radix and is demonstrated to have anti-tumor properties in colon cancer. Notwithstanding, the function and mechanism of Salvigenin in hepatocellular carcinoma (HCC) are less well studied. Different doses of Salvigenin were taken to treat HCC cells. Cell viability, colony formation ability, cell migration, invasion, apoptosis, glucose uptake, and lactate production levels were detected. As shown by the data, Salvigenin concentration dependently dampened HCC cell proliferation, migration, and invasion, weakened glycolysis by abating glucose uptake and lactate generation, and suppressed the profiles of glycolytic enzymes. Moreover, Salvigenin strengthened HCC cells' sensitivity to 5-fluorouracil (5-FU) and attenuated HCC 5-FU-resistant cells' resistance to 5-FU. Through network pharmacological analysis, we found Salvigenin potentially regulates PI3K/AKT pathway. As shown by the data, Salvigenin repressed the phosphorylated levels of PI3K, AKT, and GSK-3β. The PI3K activator 740Y-P induced PI3K/AKT/GSK-3β pathway activation and promotive effects in HCC cells. However, Salvigenin substantially weakened 740Y-P-mediated effects. In-vivo assay revealed that Salvigenin hampered the growth and promoted apoptosis of HCC cells in nude mice. Collectively, Salvigenin impedes the aerobic glycolysis and 5-FU chemoresistance of HCC cells by dampening the PI3K/AKT/GSK-3β pathway.
Collapse
Affiliation(s)
- Hui Shao
- Department of Infection, Zhejiang Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai City, 317000, Zhejiang Province, China
| | - Jingyan Chen
- Department of Infection, Zhejiang Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai City, 317000, Zhejiang Province, China
| | - Ali Li
- Department of Infection, Zhejiang Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai City, 317000, Zhejiang Province, China
| | - Lili Ma
- Department of Infection, Zhejiang Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai City, 317000, Zhejiang Province, China
| | - Yongzhi Tang
- Department of Infection, Zhejiang Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai City, 317000, Zhejiang Province, China
| | - Huazhong Chen
- Department of Infection, Zhejiang Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai City, 317000, Zhejiang Province, China
| | - Yongping Chen
- Department of Infectious and Liver Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Junyan Liu
- Department of Infection, Zhejiang Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai City, 317000, Zhejiang Province, China.
| |
Collapse
|
34
|
Zhang J, Liu Y, Wang X, Wang Z, Xing E, Li J, Wang D. Curcumin inhibits proliferation of hepatocellular carcinoma cells by blocking PTPN1 and PTPN11 expression. Oncol Lett 2023; 26:307. [PMID: 37332329 PMCID: PMC10272960 DOI: 10.3892/ol.2023.13893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/04/2023] [Indexed: 06/20/2023] Open
Abstract
The antitumor mechanism of curcumin is unclear, especially in hepatocellular carcinoma (HCC) cells. To clarify the mechanism of action of curcumin in the effective treatment of HCC, the targets of curcumin were screened and validated. Candidate genes of curcumin for HCC were screened using the traditional Chinese medicine systems pharmacology (TCMSP) database and validated using The Cancer Genome Atlas (TCGA) database. The correlation of mRNA expression levels between key candidate genes was identified in the TCGA liver hepatocellular carcinoma (LIHC) dataset. The effects on prognosis were analyzed to identify the target gene of curcumin, which inhibits HCC cell proliferation. Based on the subcutaneous xenograft model of human HCC in nude mice, the expression levels of target proteins were observed using immunohistochemistry. The analysis results of the present study identified the target genes of curcumin, which were obtained by screening the TCSMP database. The protein tyrosine phosphatase non-receptor type 1 (PTPN1) was obtained from TCGA database analysis of the targeted genes. The expression levels of PTPN1 and its homologous sequence genes in TCGA LIHC project was analyzed to identify the potential target gene of curcumin, for use in HCC treatment. Next, xenograft experiments were performed to investigate the therapeutic effects of curcumin in an animal model. Curcumin was demonstrated to inhibit the growth of HCC xenograft tumors in mice. Immunohistochemistry results demonstrated that the protein expression levels of PTPN1 and PTPN11 in the curcumin group were significantly lower compared with those in the control group. In conclusion, these results demonstrated that curcumin inhibits the proliferation of HCC cells by inhibiting the expression of PTPN1 and PTPN11.
Collapse
Affiliation(s)
- Jingru Zhang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yang Liu
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaojie Wang
- Quality Department, Shandong Runzhong Pharmaceutical Co., Ltd., Yantai, Shandong 264003, P.R. China
| | - Zhiyi Wang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Enjia Xing
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Jingmin Li
- Department of Human Anatomy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Dong Wang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
35
|
Wang X, Tian Y, Lin H, Cao X, Zhang Z. Curcumin induces apoptosis in human hepatocellular carcinoma cells by decreasing the expression of STAT3/VEGF/HIF-1α signaling. Open Life Sci 2023; 18:20220618. [PMID: 37333486 PMCID: PMC10276545 DOI: 10.1515/biol-2022-0618] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/01/2023] [Accepted: 04/26/2023] [Indexed: 06/20/2023] Open
Abstract
Curcumin is the most abundant derivative of turmeric rhizome. Although studies have proved that curcumin could inhibit the growth of tumors, its specific molecular mechanism has not yet been fully elucidated. This study aims to systematically elaborate the mechanisms of curcumin against hepatocellular carcinoma. The anti-tumor effect of curcumin was determined by the cell viability test. Flow cytometry was applied to examine the cell cycle and the apoptosis of cancer cells, and the cancer cell migration was detected by wound healing experiments. The expressions of signal transducers and activators of transcription 3 (STAT3), vascular endothelial growth factor (VEGF), and hypoxia-inducible factor-1α (HIF-1α) in cancer cells were examined by immunostaining and analyzed by the Image J analysis system. After treatment with curcumin, the apoptosis ratio of HepG2 cells increased significantly (P < 0.05). The proliferation of cancer cells was arrested at the S-phase cell cycle, and the migration of cancer cells was inhibited by the increasing concentration of curcumin, together with the decreasing expressions of STAT3, VEGF, and HIF-1α signaling pathways. The results indicate that curcumin could effectively inhibit the growth and migration of hepatocarcinoma cells by inducing cancer cell apoptosis, blocking the cancer cell cycle in the S phase, and reducing the expression of STAT3, VEGF, and HIF-1α signaling pathways.
Collapse
Affiliation(s)
- Xiaoping Wang
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, No. 6 Wenhui East Road, Weicheng District, Xianyang, 712082 Shaanxi, China
- Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, 712082 Shaanxi, China
| | - Yu Tian
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, No. 6 Wenhui East Road, Weicheng District, Xianyang, 712082 Shaanxi, China
- Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, 712082 Shaanxi, China
| | - Huanping Lin
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, No. 6 Wenhui East Road, Weicheng District, Xianyang, 712082 Shaanxi, China
- Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, 712082 Shaanxi, China
| | - Xiaolan Cao
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, No. 6 Wenhui East Road, Weicheng District, Xianyang, 712082 Shaanxi, China
- Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, 712082 Shaanxi, China
| | - Zhendong Zhang
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, No. 6 Wenhui East Road, Weicheng District, Xianyang, 712082 Shaanxi, China
- Joint Laboratory for Research on Active Components and Pharmacological Mechanism of Tibetan, Materia Medica of Tibetan Medical Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, 712082 Shaanxi, China
| |
Collapse
|
36
|
Zhang CY, Liu S, Yang M. Antioxidant and anti-inflammatory agents in chronic liver diseases: Molecular mechanisms and therapy. World J Hepatol 2023; 15:180-200. [PMID: 36926234 PMCID: PMC10011909 DOI: 10.4254/wjh.v15.i2.180] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/30/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Chronic liver disease (CLD) is a continuous process that causes a reduction of liver function lasting more than six months. CLD includes alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), chronic viral infection, and autoimmune hepatitis, which can lead to liver fibrosis, cirrhosis, and cancer. Liver inflammation and oxidative stress are commonly associated with the development and progression of CLD. Molecular signaling pathways such as AMP-activated protein kinase (AMPK), C-Jun N-terminal kinase, and peroxisome proliferator-activated receptors (PPARs) are implicated in the pathogenesis of CLD. Therefore, antioxidant and anti-inflammatory agents from natural products are new potent therapies for ALD, NAFLD, and hepatocellular carcinoma (HCC). In this review, we summarize some powerful products that can be potential applied in all the stages of CLD, from ALD/NAFLD to HCC. The selected agents such as β-sitosterol, curcumin, genistein, and silymarin can regulate the activation of several important molecules, including AMPK, Farnesoid X receptor, nuclear factor erythroid 2-related factor-2, PPARs, phosphatidylinositol-3-kinase, and lysyl oxidase-like proteins. In addition, clinical trials are undergoing to evaluate their efficacy and safety.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
37
|
Wang Y, Li J, Xia L. Plant-derived natural products and combination therapy in liver cancer. Front Oncol 2023; 13:1116532. [PMID: 36865794 PMCID: PMC9971944 DOI: 10.3389/fonc.2023.1116532] [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: 12/05/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Liver cancer is one of the malignant cancers globally and seriously endangers human health because of its high morbidity and mortality. Plant-derived natural products have been evaluated as potential anticancer drugs due to low side effects and high anti-tumor efficacy. However, plant-derived natural products also have defects of poor solubility and cumbersome extraction process. In recent years, a growing numbers of plant derived natural products have been used in combination therapy of liver cancer with conventional chemotherapeutic agents, which has improved clinical efficacy through multiple mechanisms, including inhibition of tumor growth, induction of apoptosis, suppression of angiogenesis, enhancement of immunity, reversal of multiple drug resistance and reduction of side effects. The therapeutic effects and mechanisms of plant-derived natural products and combination therapy on liver cancer are reviewed to provide references for developing anti-liver-cancer strategies with high efficacy and low side effects.
Collapse
Affiliation(s)
- Yuqin Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jinyao Li
- *Correspondence: Jinyao Li, ; Lijie Xia,
| | - Lijie Xia
- *Correspondence: Jinyao Li, ; Lijie Xia,
| |
Collapse
|
38
|
Miyazaki K, Morine Y, Xu C, Nakasu C, Wada Y, Teraoku H, Yamada S, Saito Y, Ikemoto T, Shimada M, Goel A. Curcumin-Mediated Resistance to Lenvatinib via EGFR Signaling Pathway in Hepatocellular Carcinoma. Cells 2023; 12:612. [PMID: 36831279 PMCID: PMC9954241 DOI: 10.3390/cells12040612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Lenvatinib is a multi-kinase inhibitor approved as a first-line treatment for patients with unresectable advanced hepatocellular carcinoma (HCC). However, its response rate is unsatisfactory, primarily due to the acquisition of resistance, which limits its clinical significance for treating patients with HCC. Recent evidence suggests that epidermal growth factor receptor (EGFR) activation can trigger Lenvatinib-resistance; and is considered an important therapeutic target in HCC. Curcumin, one of the most studied naturally occurring botanicals with robust anti-cancer activity, is also reported to be a potent tyrosine kinase inhibitor. In this study, we hypothesized that the anti-EGFR potential of Curcumin might help overcome Lenvatinib resistance in HCC. We established two Lenvatinib-resistant cells and discovered that a combination of Curcumin and Lenvatinib exhibited a synergistic anti-tumor efficacy in the resistant HCC cell lines. In line with previous reports, Lenvatinib-resistant cell lines revealed significant activation of the EGFR, and genomewide transcriptomic profiling analysis identified that the PI3K-AKT pathway was associated with Lenvatinib resistance. The combination treatment with Curcumin and Lenvatinib dramatically suppressed gene and protein expression of the EGFR-PI3K-AKT pathway, suggesting Curcumin overcomes Lenvatinib resistance via inhibition of EGFR. We further validated these findings in tumor spheroids derived from resistant cell lines. In conclusion, we, for the first time, report that Curcumin reverses Lenvatinib resistance in HCC, and that their combination has clinical application potential for adjunctive treatment in HCC.
Collapse
Affiliation(s)
- Katsuki Miyazaki
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Yuji Morine
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Chiharu Nakasu
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Yuma Wada
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Hiroki Teraoku
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Shinichiro Yamada
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Yu Saito
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Tetsuya Ikemoto
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Mitsuo Shimada
- Department of Surgery, Tokushima University, Tokushima 779-1510, Japan
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
39
|
The Role of Ferroptosis and Cuproptosis in Curcumin against Hepatocellular Carcinoma. Molecules 2023; 28:molecules28041623. [PMID: 36838613 PMCID: PMC9964324 DOI: 10.3390/molecules28041623] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/31/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Among cancer-related deaths, hepatocellular carcinoma (HCC) ranks fourth, and traditional Chinese medicine (TCM) treatment is an important complementary alternative therapy for HCC. Curcumin is a natural ingredient extracted from Curcuma longa with anti-HCC activity, while the therapeutic mechanisms of curcumin remain unclear, especially on ferroptosis and cuproptosis. METHODS Differentially expressed genes (DEGs) of curcumin treatment in PLC, KMCH, and Huh7 cells were identified, respectively. The common genes among them were then obtained to perform functional enrichment analysis and prognostic analysis. Moreover, weighted gene co-expression network analysis (WGCNA) was carried out for the construction of the co-expression network. The ferroptosis potential index (FPI) and the cuproptosis potential index (CPI) were subsequently used to quantitatively analyze the levels of ferroptosis and cuproptosis. Finally, single-cell transcriptome analysis of liver cancer was conducted. RESULTS We first identified 702, 515, and 721 DEGs from curcumin-treated PLC, KMCH, and Huh7 cells, respectively. Among them, HMOX1, CYP1A1, HMGCS2, LCN2, and MTTP may play an essential role in metal ion homeostasis. By WGCNA, grey60 co-expression module was associated with curcumin treatment and involved in the regulation of ion homeostasis. Furthermore, FPI and CPI assessment showed that curcumin had cell-specific effects on ferroptosis and cuproptosis in different HCC cells. In addition, there are also significant differences in ferroptosis and cuproptosis levels among 16 HCC cell subtypes according to single-cell transcriptome data analysis. CONCLUSIONS We developed CPI and combined it with FPI to quantitatively analyze curcumin-treated HCC cells. It was found that ferroptosis and cuproptosis, two known metal ion-mediated forms of programmed cell death, may have a vital effect in treating HCC with curcumin, and there are significant differences in various liver cancer cell types and curcumin treatment which should be considered in the clinical application of curcumin.
Collapse
|
40
|
Li Z, Hu S, Pu LY, Li Z, Zhu G, Cao Y, Li L, Ma Y, Liu Z, Li X, Liu G, Chen K, Wu Z. Design, synthesis and biological evaluation of a novel colchicine-magnolol hybrid for inhibiting the growth of Lewis lung carcinoma in Vitro and in Vivo. Front Chem 2022; 10:1094019. [PMID: 36583151 PMCID: PMC9792613 DOI: 10.3389/fchem.2022.1094019] [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: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Colchicine is a bioactive alkaloid originally from Colchicum autumnale and possesses excellent antiproliferative activity. However, colchicine-associated severe toxicity, gastrointestinal side effects in particular, limits its further therapeutic use. In the current study, we thus designed and synthesized a novel hybrid (CMH) by splicing colchicine and magnolol, a multifunctional polyphenol showing favorable gastrointestinal protection. The antitumor activity of CMH in Lewis lung carcinoma (LLC) was then evaluated in vitro and in vivo. Biologically, CMH inhibited the growth of LLC cells with an IC50 of 0.26 μM, 100 times more potently than cisplatin (26.05 μM) did. Meanwhile, the cytotoxicity of CMH was 10-fold lower than that of colchicine in normal human lung cells (BEAS-2B). In C57BL/6 mice xenograft model, CMH (0.5 mg/kg) worked as efficacious as colchicine (0.5 mg/kg) to inhibit tumor growth and 2 times more potently than cisplatin (1 mg/kg). In terms of mortality, 7 out of 10 mice died in colchicine group (0.75 mg/kg), while no death was observed in groups receiving CMH or cisplatin at 0.75 mg/kg. Mechanistic studies using Western blot revealed that CMH dose-dependently suppressed the protein expression of phosphorylated ERK. Molecular docking analysis further indicated that CMH was well fitted in the colchicine binding site of tubulin and formed several hydrogen bonds with tubulin protein. These results enable our novel hybrid CMH as a potential antineoplastic agent with lower toxicity, and provide perquisites for further investigation to confirm the therapeutic potentiality of this novel hybrid.
Collapse
Affiliation(s)
- Zhiyue Li
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China,Shenzhen Institute of Geriatrics, Shenzhen, China
| | - Shengquan Hu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China,Shenzhen Institute of Geriatrics, Shenzhen, China
| | - Liu-Yang Pu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China,Shenzhen Institute of Geriatrics, Shenzhen, China,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Ziwen Li
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Guanbao Zhu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China,Guangxi University of Chinese Medicine, Nanning, China
| | - Yongkai Cao
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Limin Li
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yucui Ma
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhanyan Liu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xinping Li
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Guangjie Liu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Keji Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Zhengzhi Wu, ; Keji Chen,
| | - Zhengzhi Wu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China,Shenzhen Institute of Geriatrics, Shenzhen, China,*Correspondence: Zhengzhi Wu, ; Keji Chen,
| |
Collapse
|
41
|
Wu H, Qian D, Bai X, Sun S. Targeted Pyroptosis Is a Potential Therapeutic Strategy for Cancer. JOURNAL OF ONCOLOGY 2022; 2022:2515525. [PMID: 36467499 PMCID: PMC9715319 DOI: 10.1155/2022/2515525] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/21/2022] [Accepted: 11/15/2022] [Indexed: 12/01/2023]
Abstract
As a type of regulated cell death (RCD) mode, pyroptosis plays an important role in several kinds of cancers. Pyroptosis is induced by different stimuli, whose pathways are divided into the canonical pathway and the noncanonical pathway depending on the formation of the inflammasomes. The canonical pathway is triggered by the assembly of inflammasomes, and the activation of caspase-1 and then the cleavage of effector protein gasdermin D (GSDMD) are promoted. While in the noncanonical pathway, the caspase-4/5/11 (caspase 4/5 in humans and caspase 11 in mice) directly cleave GSDMD without the assembly of inflammasomes. Pyroptosis is involved in various cancers, such as lung cancer, gastric cancer, hepatic carcinoma, breast cancer, and colorectal carcinoma. Pyroptosis in gastric cancer, hepatic carcinoma, breast cancer, and colorectal carcinoma is related to the canonical pathway, while both the canonical and noncanonical pathway participate in lung cancer. Moreover, simvastatin, metformin, and curcumin have effect on these cancers and simultaneously promote the pyroptosis of cancer cells. Accordingly, pyroptosis may be an important therapeutic target for cancer.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
- Clinical Medicine, Three Class, 2020 Grade, Kunming Medical University, Kunming, China
| | - Dianlun Qian
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiangfeng Bai
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
42
|
Zhang J, Huang Y, Xu J, Zhao R, Xiong C, Habu J, Wang Y, Luo X. Global publication trends and research hotspots of curcumin application in tumor: A 20-year bibliometric approach. Front Oncol 2022; 12:1033683. [PMID: 36300100 PMCID: PMC9589263 DOI: 10.3389/fonc.2022.1033683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/23/2022] [Indexed: 01/03/2023] Open
Abstract
Malignant tumor is a disease caused by the imbalance of cell growth and proliferation mechanism, which seriously threatens human health and life safety. However, side effects and drug resistance are the key factors that limit the efficacy of anti-tumor drugs. Therefore, it is urgent and necessary to explore and unearth natural, safe and effective chemosensitizers in tumor researches. Curcumin is the main active ingredient in Curcuma, which has anti-inflammatory, anti-inflammatory and anti-oxidation effects, and has inhibitory effects on a variety of cancers. Bibliometric analysis is a scientific and quantitative method to assess the published articles, which can help researchers to find the development trends and the research hotspots of a specific research field, providing the development of future research for researchers. This study searched the Web Science Core Collection (woscc) for publications related to curcumin and tumors from January 1, 2001 to December 31, 2021. The specific characteristics of 1707 publications were analyzed by using Microsoft Excel software, CiteSpace, Vosviewer and online analysis platform of literature metrology. China had the largest number of published articles, with 579 publications. Aggarwal BB’s articles total citations and average citations were the most. PLoS One had the largest number of publications, with 32 publications. The current research focuses on “nanoparticles”, “delivery”, “micells” and “doxorubicin”. At present, nano based drug delivery system to improve the bioavailability of curcumin and thus to treat tumors will be the focus of future research.
Collapse
|