1
|
Huang HY, Lin LZY, Lu XY, Jiang Y. F-α-DDB-derivative, a novel synthetic of bifendate, plus epirubicin improves antitumor efficacy against triple negative breast cancer without additional cardiotoxicity. Discov Oncol 2025; 16:690. [PMID: 40338489 PMCID: PMC12061825 DOI: 10.1007/s12672-025-02545-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Triple negative breast cancer (TNBC) refers to a molecular subtype of breast cancers (BC) with high rate of distant metastases and poor prognosis. Epirubicin (EPI) is widely used for the therapy of TNBC but it's limited in clinical use due to cardiotoxicity and chemotherapy resistance. We previously identified F-α-DDB-derivative, a novel synthetic of bifendate, as a potential agent to improve the therapeutic efficacy of TNBC in combination with EPI since F-α-DDB-derivative inhibited MDA-MB-468, but not as much as EPI. In this study, we investigated the antitumor activity of F-α-DDB-derivative in combination with EPI against TNBC in vitro and in vivo and whether co-treatment would induce additional cardiotoxicity. In human TNBC MDA-MB-468 cells, application of F-α-DDB-derivative (11.5, 23.0, 46.0 μg/ml) in combination with EPI (1.5, 3.0, 6.0 μg/ml) exhibited great inhibition on cell viability and cell proliferation. Additionally, F-α-DDB-derivative (5.75, 11.5, 23.0, 46.0 μg/ml) interacted with EPI (0.75, 1.5, 3.0, 6.0 μg/ml) synergistically to induce apoptosis in MDA-MB-468 cells. This suggests that F-α-DDB-derivative may be more sensitive to apoptotic pathways. Furthermore, we revealed that co-administration of EPI and F-α-DDB-derivative (ip, once every other day for 14 days) significantly increased the therapeutic efficacy of EPI (2.0 mg/kg) or F-α-DDB-derivative (20.0 mg/kg) in mice harboring MDA-MB-468 cell xenografts without additional cardiotoxicity compared to that in EPI monotherapy group. These results implicate that co-treatment of EPI and F-α-DDB-derivative may be a potential therapeutic approach for the treatment of TNBC.
Collapse
Affiliation(s)
- Hai-Yi Huang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lisa Zong Yong Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin-Yi Lu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of General Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Fujian, China.
| |
Collapse
|
2
|
Wang R, Lv X, Xu W, Li X, Tang X, Huang H, Yang M, Ma S, Wang N, Niu Y. Effects of the periodic fasting-mimicking diet on health, lifespan, and multiple diseases: a narrative review and clinical implications. Nutr Rev 2025; 83:e412-e426. [PMID: 38287649 DOI: 10.1093/nutrit/nuae003] [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] [Indexed: 01/31/2024] Open
Abstract
Dietary restriction and fasting have been recognized for their beneficial effects on health and lifespan and their potential application in managing chronic metabolic diseases. However, long-term adherence to strict dietary restrictions and prolonged fasting poses challenges for most individuals and may lead to unhealthy rebound eating habits, negatively affecting overall health. As a result, a periodic fasting-mimicking diet (PFMD), involving cycles of fasting for 2 or more days while ensuring basic nutritional needs are met within a restricted caloric intake, has gained widespread acceptance. Current research indicates that a PFMD can promote stem cell regeneration, suppress inflammation, extend the health span of rodents, and improve metabolic health, among other effects. In various disease populations such as patients with diabetes, cancer, multiple sclerosis, and Alzheimer's disease, a PFMD has shown efficacy in alleviating disease symptoms and improving relevant markers. After conducting an extensive analysis of available research on the PFMD, it is evident that its advantages and potential applications are comparable to other fasting methods. Consequently, it is proposed in this review that a PFMD has the potential to fully replace water-only or very-low-energy fasting regimens and holds promise for application across multiple diseases.
Collapse
Affiliation(s)
- Ruohua Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Xinyi Lv
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Wenyu Xu
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Xiaoqing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Xuanfeng Tang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - He Huang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Mengxia Yang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Shuran Ma
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Nan Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| | - Yucun Niu
- Department of Nutrition and Food Hygiene, College of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, China
| |
Collapse
|
3
|
Lu YY, Fang YY, Wang SS, Guo J, Song JL, Zhu L, Lin ZK, Wang R, Zhang SY, Qiu WS, Qi WW. Cepharanthine sensitizes gastric cancer cells to chemotherapy by targeting TRIB3-FOXO3-FOXM1 axis to inhibit autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156161. [PMID: 39454374 DOI: 10.1016/j.phymed.2024.156161] [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: 05/15/2024] [Revised: 08/28/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Gastric cancer is among the common solid tumors. Chemotherapy resistance is the most common issue in gastric cancer treatment. Inhibiting intracellular autophagy may be a feasible method for overcoming chemotherapy resistance. Cepharanthine (CEP), a natural small molecule extracted from the stephania cephalantha Hayata plant, has been demonstrated to significantly inhibit cancer growth and can regulate autophagy. Although CEP can significantly inhibit cancer growth, it remains unclear whether CEP can regulate autophagy in gastric cancer. This study aimed to investigate whether CEP can enhance the sensitivity of gastric cancer to chemotherapy and elucidate its molecular mechanism. METHODS Three gastric cancer cell lines (AGS, SGC7901, and MFC) and one normal gastric mucosal epithelial cell line (GES-1) were used for in vitro experiments. The characterization of autophagy in gastric cancer cells included the detection of autophagy markers and autophagy flux through immunofluorescence staining and Western blotting, as well as the assessment of lysosomal function using fluorescence staining (LysoTracker Red DND-99, Acridine Orange staining) and Western blotting. The cytotoxicity of CEP, autophagy inhibitors (chloroquine [CQ] and 3-methyladenine [3MA]), and chemotherapy drugs (doxorubicin [DOX] and cisplatin [CIS]) was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, cell colony formation, and fluorescence staining techniques (H2DCFDA, Dihydroethidium, and JC-1 staining). The interaction between CEP and autophagy inhibitors was tested in a 615 mice model, and changes in the gut microbiota were determined through accurate 16S absolute quantification sequencing. The signaling pathway and autophagy regulatory target TRIB3-FOXO3-FOXM1 were confirmed through molecular docking, RNA sequencing, bioinformatic analysis, transfection techniques, and Western blotting. RESULTS CEP blocked autophagic flux in gastric cancer cells without affecting lysosomal function. As a novel autophagy inhibitor, CEP could combine with conventional autophagy inhibitors (CQ and 3MA) to block intracellular autophagy, thereby inhibiting gastric cancer growth. During this process, changes in the gut microbiota were observed, including low-level changes in Odoribacterium, Erysipelatoclostridium, and ParaPrevotella and high-level changes in Ileibacterium, Enterorhabdus, and Bifidobacterium. Additionally, CEP synergistically inhibited the growth of gastric cancer when combined with chemotherapy drugs. Mechanistically, the TRIB3-FOXO3-FOXM1 signaling axis was found to be involved in the inhibition of gastric cancer by CEP combined with autophagy inhibitors and chemotherapy drugs, thereby mediating cell apoptosis. CONCLUSION This study links the TRIB3-FOXO3-FOXM1 axis with chemotherapy efficacy. Our findings demonstrated that CEP inhibits autophagy by modulating the FOXO3-FOXM1 axis. When combined with chemotherapy drugs (DOX and CIS), CEP, as an autophagy inhibitor, can limit TRIB3 protein expression, thereby regulating the FOXO3-FOXM1 axis and enhancing its ability to prevent gastric cancer growth. These findings may contribute to improving the prognosis of patients with gastric cancer. Furthermore, these results enrich the fundamental understanding of how autophagy inhibition can enhance clinical cancer treatment efficacy and provide insights into the potential mechanisms by which CEP functions as an anti-tumor drug, thereby exploring its value for clinical application.
Collapse
Affiliation(s)
- Yang-Yang Lu
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China
| | - Yuan-Yuan Fang
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China
| | - Sha-Sha Wang
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China
| | - Jing Guo
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China
| | - Jia-Lin Song
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China
| | - Liang Zhu
- Qingdao Municipal Hospital, Department of Orthopedic Surgery, No.1 Jiaozhou Road, Shibei District, Qingdao City, Shandong Province, Qingdao 266000, China
| | - Zhong-Kun Lin
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China; Department of Oncology, Shandong Provincial Third Hospital, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan City, Shandong Province, Jinan 250031, China
| | - Rui Wang
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China
| | - Si-Yi Zhang
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China
| | - Wen-Sheng Qiu
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China.
| | - Wei-Wei Qi
- The Affiliated Hospital of Qingdao University, Department of Oncology, No.16 Jiangsu Road, Shinan District, Qingdao 266000, China.
| |
Collapse
|
4
|
Dong X, Zhu W, Wang N. Cepharanthine inhibits the proliferation of glioblastoma cells by blocking the autophagy-lysosomal pathway. Toxicol Appl Pharmacol 2024; 493:117141. [PMID: 39500397 DOI: 10.1016/j.taap.2024.117141] [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: 09/09/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/09/2024]
Abstract
Cepharanthine (CEP) is a Stephania cepharantha-derived bioactive alkaloid that can inhibit the progression of numerous tumors. However, the effects and specific mechanisms of CEP performance in glioblastoma (GBM) remain unclear. Thus, this study focused on exploring the effects of CEP on GBM and clarifying the underlying mechanisms. U251 and U87 cells were selected to estimate the anti-GBM effects of CEP, and the possible targets of CEP were analyzed using RNA sequencing (RNA-seq). Validation experiments based on RNA-seq data were performed to clarify the key pathway by which CEP mediates GBM cells response. Results showed that CEP administration successfully inhibited the proliferation and induced the cell cycle arrest and apoptosis of the GBM cells. RNA-seq analysis after CEP administration identified 386 differentially expressed genes, which were highly enriched in the autophagy-lysosomal pathway. Subsequent findings demonstrated that CEP exhibited the potential to curb GBM progression by causing lysosomal and autophagic dysfunction. Taken together, our results indicate that CEP is a potential drug candidate for GBM intervention.
Collapse
Affiliation(s)
- Xiangjun Dong
- Department of Pediatrics, Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing Research Center for Prevention & Control of Maternal and Child Diseases and Public Health, Chongqing 401147, China
| | - Weiyi Zhu
- National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Nianrong Wang
- Department of Pediatrics, Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing Research Center for Prevention & Control of Maternal and Child Diseases and Public Health, Chongqing 401147, China.
| |
Collapse
|
5
|
Hu J, Chen NN, Li LG, Yu TT, Qin Y, Peng XC, Li HT, Li XY, Ma TQ, Lu YH, Han N, Xu Z, Hui YJ, Li TF. Cepharanthine-mediated endoplasmic reticulum stress inhibits Notch1 via binding GRP78 for suppressing hepatocellular carcinoma metastasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156162. [PMID: 39427519 DOI: 10.1016/j.phymed.2024.156162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND The metastasis of hepatocellular carcinoma (HCC) leads to a poor prognosis, wherein the activation of Notch1 is an essential contributor. Cepharanthine (Cep) has been identified for its effective antiviral function and versatile intracellular targets. Our previous study has only reported the anti-cancer efficacy of Cep in lung cancer, without an in-depth exploration. Herein, the present study aims to investigate the anti-metastasis effect in HCC, the target involved, and the molecular mechanism of Cep. METHODS Stable over-expression of Notch1-N1ICD yielded C5WN1 cells compared with C5WBF344 cells. The C5WN1 cells and C5WN1 cell-bearing mice were applied as the HCC model. The bioinformatics analysis, RNA sequencing, molecular docking, cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS), microscale thermophoresis (MST), and transient knockdown techniques were carried out to identify the underlying target. The apoptosis assay, immunofluorescent staining, qRT-PCR, Western blots, Elisa, flow cytometry, migration and scratching experiments, Transmission electron microscopy (TEM), laser scanning confocal microscopy (LSCM), micro-computed tomography (micro-CT), and histopathological experiments were conducted to assay the anti-HCC efficacy, functions, and mechanism. RESULTS Notch1 had an increased expression in HCC and contributed to metastasis thereupon. Surprisingly, Cep (2 μg/ml in vitro, 5 mg kg-1in vivo) presented potent Notch1 signaling pathway inhibitory effect and anti-metastasis efficacy in C5WN1 cells and in situ mice models as evidenced by reduced Notch1/MMP-2/MMP-9 expression, TGF-β release, decreased cell migration, diminished pulmonary metastases, and prolonged survival. RNA sequencing showed that the differential gene of Cep-treated HCC cells was positioned in the endoplasmic reticulum (ER). Molecular docking, CETSA, DARTS, and MST further identified that the possible target of Cep was GRP78, which was distributed in the ER. As expected, Cep (2 μg/ml) up-regulated the critical molecules of ER stress such as GRP78, induced β-amyloid accumulation, and promoted calcium burst in HCC. In contrast, suppression of GRP78 attenuated Cep-induced ER stress. Furthermore, inhibition of ER stress abated Cep-induced Notch1 inactivation and HCC cells' migration. CONCLUSIONS Taken together, the present study finds that Cep possesses excellent anti-metastasis of HCC, wherein the GRP78 could be directly bound and activated by Cep, leading to ER stress and Notch1 blockage. This study reveals for the first time the effect, critical target, and mechanism of the Cep-mediated anti-cancer effect, providing novel insights into the molecular target therapy by phytomedicine.
Collapse
Affiliation(s)
- Jun Hu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Nan-Nan Chen
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Liu-Gen Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Ting-Ting Yu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Yufei Qin
- Digestive Medicine Center, the Seventh Affiliated Hospital of Sun Yat-sen University, Zhenyuan road No 628, Shenzhen 442000, Guangdong, China
| | - Xing-Chun Peng
- Department of Pathology, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen 518118, Guangzhou Province, China
| | - Hai-Tao Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Xian-Yu Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Tian-Qi Ma
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Yao-Hua Lu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Ning Han
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Yuan-Jian Hui
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin south road No 32, Shiyan 442000, Hubei, China.
| | - Tong-Fei Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin road No 30, Shiyan 442000, Hubei, China.
| |
Collapse
|
6
|
Zhao F, Ding Z, Chen M, Ji M, Li F. Cepharanthine as an effective small cell lung cancer inhibitor: integrated insights from network pharmacology, RNA sequencing, and experimental validation. Front Pharmacol 2024; 15:1517386. [PMID: 39669201 PMCID: PMC11634586 DOI: 10.3389/fphar.2024.1517386] [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: 10/26/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Background Small cell lung cancer (SCLC) is an aggressive malignancy with limited treatment options and poor prognosis, underscoring the need for new therapeutic agents. Methods A library of 640 natural products was screened for anti-proliferative activity in SCLC cells. The effects of Cepharanthine (CE) on SCLC cells were assessed in vitro and in vivo. Network pharmacology and RNA sequencing (RNA-seq) were used to elucidate the molecular mechanisms. Pathway enrichment analysis was performed using Gene Set Enrichment Analysis (GSEA) with Hallmark and Reactome gene sets. Protein-protein interaction (PPI) networks, along with the Cytoscape cytoHubba plugin, were used to identify key hub genes. RT-PCR and Western blotting were employed to validate mRNA and protein expression. Molecular docking studies assessed the binding affinity of CE to potential targets. Bioinformatics analyses, including expression profiling, prognostic evaluation, and loss-of-function studies, were used to explore the role of specific genes in SCLC. Results CE was identified as a promising SCLC inhibitor. In vitro, CE significantly inhibited SCLC cell proliferation, colony formation, migration, and invasion, while promoting apoptosis. In vivo, CE treatment notably reduced tumor volume in xenograft models. Network pharmacology identified 60 potential target genes, with enrichment analysis indicating their involvement in cholesterol metabolism regulation. RNA-seq and experimental validation further confirmed that CE inhibits cholesterol synthesis in SCLC cells by downregulating key enzymes, including HMGCR, HMGCS1, IDI1, FDFT1, and SQLE. Molecular docking studies confirmed the binding of CE to these enzymes. Additionally, these enzymes were found to be highly expressed in SCLC cells, with elevated levels of HMGCS1, HMGCR, and IDI1 correlating with poor prognosis. Functional assays revealed that silencing these genes significantly suppressed SCLC cell proliferation. Conclusion This study identifies CE as a potential therapeutic agent for SCLC, acting through the suppression of cholesterol synthesis, and uncovers novel therapeutic targets for the treatment of this aggressive cancer.
Collapse
Affiliation(s)
- Fengyun Zhao
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Zhaowei Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mengting Chen
- South China Normal University, Guangzhou, Guangdong, China
| | - Mingfang Ji
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
7
|
Chen C, Xiang A, Lin X, Guo J, Liu J, Hu S, Rui T, Ye Q. Mitophagy: insights into its signaling molecules, biological functions, and therapeutic potential in breast cancer. Cell Death Discov 2024; 10:457. [PMID: 39472438 PMCID: PMC11522701 DOI: 10.1038/s41420-024-02226-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
Mitophagy, a form of selective autophagy that removes damaged or dysfunctional mitochondria, plays a crucial role in maintaining mitochondrial and cellular homeostasis. Recent findings suggest that defective mitophagy is closely associated with various diseases, including breast cancer. Moreover, a better understanding of the multifaceted roles of mitophagy in breast cancer progression is crucial for the treatment of this disease. Here, we will summarize the molecular mechanisms of mitophagy process. In addition, we highlight the expression patterns and roles of mitophagy-related signaling molecules in breast cancer progression and the potential implications of mitophagy for the development of breast cancer, aiming to provide better therapeutic strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Cong Chen
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Aizhai Xiang
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Xia Lin
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jian Liu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Shufang Hu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Tao Rui
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Qianwei Ye
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
8
|
Li LG, Zhang D, Huang Q, Yan M, Chen NN, Yang Y, Xiao RC, Liu H, Han N, Qureshi AM, Hu J, Leng F, Hui YJ. Mitochondrial disruption resulting from Cepharanthine-mediated TOM inhibition triggers ferroptosis in colorectal cancer cells. J Cancer Res Clin Oncol 2024; 150:460. [PMID: 39402386 PMCID: PMC11478973 DOI: 10.1007/s00432-024-05974-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Chemotherapy for colorectal cancer (CRC) urgently needs low-toxicity and highly effective phytomedicine. Cepharanthine (Cep) shown to have multiple anti-tumor effects, including colorectal cancer, whose pivotal mechanisms are not fully understood. Herein, the present work aims to reveal the impact of Cep on the mitochondrial and anti-injury functions of CRC cells. METHODS The TOM70/20 expression was screened by bioinformatic databases. SW480 cells were utilized as the colorectal cancer cell model. The expression of TOM70/20 and the downstream molecules were measured by western blots (WB). The ferroptosis was analyzed using Transmission electron microscopy (TEM), C11-BODIPY, PGSK, and DCFH-DA probes, wherein the detection was performed by flow cytometry and laser confocal microscopy. The anti-cancer efficacy was conducted by CCK-8 and Annexin-V/PI assay. The rescue experiments were carried out using Fer-1 and TOM70 plasmid transfection. RESULTS Bioinformatic data identified TOM20 and TOM70 were highly expressed in colorectal cancer, which could be down-regulated by Cep. Further findings disclosed that Cep treatment destroyed the mitochondria and inactivated the NRF2 signaling pathway, an essential pathway for resistance to ferroptosis, thereby promoting reactive oxygen species (ROS) generation in CRC cells. As a result, prominent ferroptosis could be observed in CRC cells in response to Cep, which thereby led to the reduced cell viability of cancer cells. On the contrary, recovery of TOM70 dampened the Cep-elicited mitochondria damage, ferroptosis, and anti-cancer efficacy. CONCLUSION In summary, Cep-mediated TOM inhibition inactivates the NRF2 signaling pathway, thereby triggering ferroptosis and achieving an anti-colorectal cancer effect. The current study provides an innovative chemotherapeutic approach for colorectal cancer with phytomedicine.
Collapse
Affiliation(s)
- Liu-Gen Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Di Zhang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Qi Huang
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Min Yan
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Nan-Nan Chen
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Yan Yang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Rong-Cheng Xiao
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Hui Liu
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Ning Han
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Abdul Moiz Qureshi
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Jun Hu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Fan Leng
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Yuan-Jian Hui
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China.
| |
Collapse
|
9
|
Bai XF, Hu J, Wang MF, Li LG, Han N, Wang H, Chen NN, Gao YJ, You H, Wang X, Xu X, Yu TT, Li TF, Ren T. Cepharanthine triggers ferroptosis through inhibition of NRF2 for robust ER stress against lung cancer. Eur J Pharmacol 2024; 979:176839. [PMID: 39033838 DOI: 10.1016/j.ejphar.2024.176839] [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: 05/12/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Severe endoplasmic reticulum (ER) stress elicits apoptosis to suppress lung cancer. Our previous research identified that Cepharanthine (CEP), a kind of phytomedicine, possessed powerful anti-cancer efficacy, for which the underlying mechanism was still uncovered. Herein, we investigated how CEP induced ER stress and worked against lung cancer. METHODS The differential expression genes (DEGs) and enrichment were detected by RNA-sequence. The affinity of CEP and NRF2 was analyzed by cellular thermal shift assay (CETSA) and molecular docking. The function assay of lung cancer cells was measured by western blots, flow cytometry, immunofluorescence staining, and ferroptosis inhibitors. RESULTS CEP treatment enriched DEGs in ferroptosis and ER stress. Further analysis demonstrated the target was NRF2. In vitro and in vivo experiments showed that CEP induced obvious ferroptosis, as characterized by the elevated iron ions, ROS, COX-2 expression, down-regulation of GPX4, and atrophic mitochondria. Moreover, enhanced Grp78, CHOP expression, β-amyloid mass, and disappearing parallel stacked structures of ER were observed in CEP group, suggesting ER stress was aroused. CEP exhibited excellent anti-lung cancer efficacy, as evidenced by the increased apoptosis, reduced proliferation, diminished cell stemness, and prominent inhibition of tumor grafts in animal models. Furthermore, the addition of ferroptosis inhibitors weakened CEP-induced ER stress and apoptosis. CONCLUSION In summary, our findings proved CEP drives ferroptosis through inhibition of NRF2 for induction of robust ER stress, thereby leading to apoptosis and attenuated stemness of lung cancer cells. The current work presents a novel mechanism for the anti-tumor efficacy of the natural compound CEP.
Collapse
Affiliation(s)
- Xiao-Feng Bai
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Jun Hu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Mei-Fang Wang
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Liu-Gen Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Ning Han
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Hansheng Wang
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Nan-Nan Chen
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Yu-Jie Gao
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Hui You
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Xiao Wang
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Xiang Xu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China
| | - Ting-Ting Yu
- Department of Pathology, Renmin Hospital of Shiyan, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Tong-Fei Li
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China; Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China.
| | - Tao Ren
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Renmin Road No. 30, Shiyan, Hubei, 442000, China.
| |
Collapse
|
10
|
Sousa-Squiavinato ACM, Morgado-Díaz JA. A glimpse into cofilin-1 role in cancer therapy: A potential target to improve clinical outcomes? Biochim Biophys Acta Rev Cancer 2024; 1879:189087. [PMID: 38395237 DOI: 10.1016/j.bbcan.2024.189087] [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: 10/13/2023] [Revised: 12/22/2023] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
Cofilin-1 (CFL1) modulates dynamic actin networks by severing and enhancing depolymerization. The upregulation of cofilin-1 expression in several cancer types is associated with tumor progression and metastasis. However, recent discoveries indicated relevant cofilin-1 functions under oxidative stress conditions, interplaying with mitochondrial dynamics, and apoptosis networks. In this scenario, these emerging roles might impact the response to clinical therapy and could be used to enhance treatment efficacy. Here, we highlight new perspectives of cofilin-1 in the therapy resistance context and discussed how cofilin-1 is involved in these events, exploring aspects of its contribution to therapeutic resistance. We also provide an analysis of CFL1 expression in several tumors predicting survival. Therefore, understanding how exactly coflin-1 plays, particularly in therapy resistance, may pave the way to the development of treatment strategies and improvement of patient survival.
Collapse
Affiliation(s)
| | - Jose Andrés Morgado-Díaz
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Lu YY, Zhu CY, Ding YX, Wang B, Zhao SF, Lv J, Chen SM, Wang SS, Wang Y, Wang R, Qiu WS, Qi WW. Cepharanthine, a regulator of keap1-Nrf2, inhibits gastric cancer growth through oxidative stress and energy metabolism pathway. Cell Death Discov 2023; 9:450. [PMID: 38086844 PMCID: PMC10716385 DOI: 10.1038/s41420-023-01752-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 11/09/2024] Open
Abstract
Cepharanthine (CEP), a bioactive compound derived from Stephania Cephalantha Hayata, is cytotoxic to various malignancies. However, the underlying mechanism of gastric cancer is unknown. CEP inhibited the cellular activity of gastric cancer AGS, HGC27 and MFC cell lines in this study. CEP-induced apoptosis reduced Bcl-2 expression and increased cleaved caspase 3, cleaved caspase 9, Bax, and Bad expression. CEP caused a G2 cell cycle arrest and reduced cyclin D1 and cyclin-dependent kinases 2 (CDK2) expression. Meanwhile, it increased oxidative stress, decreased mitochondrial membrane potential, and enhanced reactive oxygen species (ROS) accumulation in gastric cancer cell lines. Mechanistically, CEP inhibited Kelch-like ECH-associated protein (Keap1) expression while activating NF-E2 related factor 2 (Nrf2) nuclear translocations, increasing transcription of Nrf2 target genes quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HMOX1), and glutamate-cysteine ligase modifier subunit (GCLM). Furthermore, a combined analysis of targeted energy metabolism and RNA sequencing revealed that CEP could alter the levels of metabolic substances such as D (+) - Glucose, D-Fructose 6-phosphate, citric acid, succinic acid, and pyruvic acid, thereby altering energy metabolism in AGS cells. In addition, CEP significantly inhibited tumor growth in MFC BALB/c nude mice in vivo, consistent with the in vitro findings. Overall, CEP can induce oxidative stress by regulating Nrf2/Keap1 and alter energy metabolism, resulting in anti-gastric cancer effects. Our findings suggest a potential application of CEP in gastric cancer treatment.
Collapse
Affiliation(s)
- Yang-Yang Lu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chun-Yang Zhu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi-Xin Ding
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Wang
- Biomedical Centre, Qingdao University, Qingdao, China
| | - Shu-Fen Zhao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shu-Ming Chen
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Sha-Sha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Rui Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Sheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Wei-Wei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
12
|
Shi L, Wang S, Zhang S, Wang J, Chen Y, Li Y, Liu Z, Zhao S, Wei B, Zhang L. Research progress on pharmacological effects and mechanisms of cepharanthine and its derivatives. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2843-2860. [PMID: 37338575 DOI: 10.1007/s00210-023-02537-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/16/2023] [Indexed: 06/21/2023]
Abstract
Cepharanthine (CEP) is a bisbenzylisoquinoline alkaloid compound found in plants of the Stephania genus, which has biological functions such as regulating autophagy, inhibiting inflammation, oxidative stress, and apoptosis. It is often used for the treatment of inflammatory diseases, viral infections, cancer, and immune disorders and has great clinical translational value. However, there is no detailed research on its specific mechanism and dosage and administration methods, especially clinical research is limited. In recent years, CEP has shown significant effects in the prevention and treatment of COVID-19, suggesting its potential medicinal value waiting to be discovered. In this article, we comprehensively introduce the molecular structure of CEP and its derivatives, describe in detail the pharmacological mechanisms of CEP in various diseases, and discuss how to chemically modify and design CEP to improve its bioavailability. In summary, this work will provide a reference for further research and clinical application of CEP.
Collapse
Affiliation(s)
- Liangliang Shi
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Shuaizhe Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Shangzu Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jiawei Wang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yaping Chen
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yangyang Li
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhiwei Liu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Sichen Zhao
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Benjun Wei
- Gansu University of Traditional Chinese Medicine, Lanzhou, China.
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Lanzhou, China.
| | - Liying Zhang
- Gansu University of Traditional Chinese Medicine, Lanzhou, China.
- Key Laboratory of Traditional Chinese Medicine Exploration and Innovation Transformation in Gansu Province, Lanzhou, China.
| |
Collapse
|
13
|
Wang Y, Wang T, Wang H, Liu W, Li X, Wang X, Zhang Y. A mechanistic updated overview on Cepharanthine as potential anticancer agent. Biomed Pharmacother 2023; 165:115107. [PMID: 37423171 DOI: 10.1016/j.biopha.2023.115107] [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: 05/23/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/11/2023] Open
Abstract
The antitumor effects of traditional drugs have received increasing attention and active antitumor components extracted from traditional drugs have shown good efficacy with minimal adverse events. Cepharanthine(CEP for short) is an active component derived from the Stephania plants of Menispermaceae, which can regulate multiple signaling pathways alone or in combination with other therapeutic drugs to inhibit tumor cell proliferation, induce apoptosis, regulate autophagy, and inhibit angiogenesis, thereby inhibiting tumor progression. Therefore, we retrieved studies concerning CEP's antitumor effects in recent years and summarized the antitumor mechanism and targets, in order to gain new insights and establish a theoretical basis for further development and application of CEP.
Collapse
Affiliation(s)
- YingZheng Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Tong Wang
- School of Nursing, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China
| | - HuaXin Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - WeiDong Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xiao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - XiaoYan Wang
- College of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - YaNan Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China.
| |
Collapse
|
14
|
Liu K, Hong B, Wang S, Lou F, You Y, Hu R, Shafqat A, Fan H, Tong Y. Pharmacological Activity of Cepharanthine. Molecules 2023; 28:5019. [PMID: 37446681 DOI: 10.3390/molecules28135019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Cepharanthine, a natural bisbenzylisoquinoline (BBIQ) alkaloid isolated from the plant Stephania Cephalantha Hayata, is the only bisbenzylisoquinoline alkaloid approved for human use and has been used in the clinic for more than 70 years. Cepharanthine has a variety of medicinal properties, including signaling pathway inhibitory activities, immunomodulatory activities, and antiviral activities. Recently, cepharanthine has been confirmed to greatly inhibit SARS-CoV-2 infection. Therefore, we aimed to describe the pharmacological properties and mechanisms of cepharanthine, mainly including antitumor, anti-inflammatory, anti-pathogen activities, inhibition of bone resorption, treatment of alopecia, treatment of snake bite, and other activities. At the same time, we analyzed and summarized the potential antiviral mechanism of cepharanthine and concluded that one of the most important anti-viral mechanisms of cepharanthine may be the stability of plasma membrane fluidity. Additionally, we explained its safety and bioavailability, which provides evidence for cepharanthine as a potential drug for the treatment of a variety of diseases. Finally, we further discuss the potential new clinical applications of cepharanthine and provide direction for its future development.
Collapse
Affiliation(s)
- Ke Liu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Bixia Hong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Shuqi Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Fuxing Lou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yecheng You
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ruolan Hu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Amna Shafqat
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
15
|
Wang K, Wang X, Zhang M, Ying Z, Zhu Z, Tam KY, Li C, Zhou G, Gao F, Zeng M, Sze SCW, Wang X, Sha O. Trichosanthin Promotes Anti-Tumor Immunity through Mediating Chemokines and Granzyme B Secretion in Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24021416. [PMID: 36674931 PMCID: PMC9864620 DOI: 10.3390/ijms24021416] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/12/2023] Open
Abstract
Trichosanthin (TCS) is a type I ribosome-inactivating protein extracted from the tuberous root of the plant Trichosanthes. TCS shows promising potential in clinical drug abortion, anti-tumor and immunological regulation. However, the molecular mechanisms of its anti-tumor and immune regulation properties are still not well discovered. In the present study, we investigated the anti-tumor activity of TCS in hepatocellular carcinoma (HCC), both in vitro and in vivo. Both HCC cell lines and xenograft tumor tissues showed considerable growth inhibition after they were treated with TCS. TCS provoked caspase-mediated apoptosis in HCC cells and xenograft tumor tissues. The recruitment of CD8+ T cells to HCC tissues and the expression of chemokines, CCL2 and CCL22, were promoted upon TCS treatment. In addition, TCS induced an upregulation of Granzyme B (GrzB), TNF-α and IFN-γ in HCC tissues, which are the major cytotoxic mediators produced by T cells. Furthermore, TCS also resulted in an increase of mannose-6-phosphate receptor (M6PR), the major receptor of GrzB, in HCC tissues. In summary, these results suggest that TCS perhaps increases T-cell immunity via promoting the secretion of chemokines and accelerating the entry of GrzB to HCC cells, which highlights the potential role of TCS in anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Kaifang Wang
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518000, China
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hongkong 999077, China
| | - Xiaona Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518000, China
| | - Minghuan Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518000, China
| | - Zhenguang Ying
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518000, China
| | - Zeyao Zhu
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518000, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Chunman Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515000, China
| | - Guowei Zhou
- Department of Anatomy and Histology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518000, China
| | - Feng Gao
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518000, China
| | - Meiqi Zeng
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518000, China
| | - Stephen Cho Wing Sze
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hongkong 999077, China
| | - Xia Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518000, China
- Correspondence: (X.W.); (O.S.)
| | - Ou Sha
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518000, China
- Correspondence: (X.W.); (O.S.)
| |
Collapse
|
16
|
Chi XJ, Song YB, Liu DH, Wei LQ, An X, Feng ZZ, Lan XH, Lan D, Huang C. Significance of platelet adhesion-related genes in colon cancer based on non-negative matrix factorization-based clustering algorithm. Digit Health 2023; 9:20552076231203902. [PMID: 37766908 PMCID: PMC10521306 DOI: 10.1177/20552076231203902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Background Although surgical methods are the most effective treatments for colon adenocarcinoma (COAD), the cure rates remain low, and recurrence rates remain high. Furthermore, platelet adhesion-related genes are gaining attention as potential regulators of tumorigenesis. Therefore, identifying the mechanisms responsible for the regulation of these genes in patients with COAD has become important. The present study aims to investigate the underlying mechanisms of platelet adhesion-related genes in COAD patients. Methods The present study was an experimental study. Initially, the effects of platelet number and related genomic alteration on survival were explored using real-world data and the cBioPortal database, respectively. Then, the differentially expressed platelet adhesion-related genes of COAD were analyzed using the TCGA database, and patients were further classified by employing the non-negative matrix factorization (NMF) analysis method. Afterward, some of the clinical and expression characteristics were analyzed between clusters. Finally, least absolute shrinkage and selection operator regression analysis was used to establish the prognostic nomogram. All data analyses were performed using the R package. Results High platelet counts are associated with worse survival in real-world patients, and alternations to platelet adhesion-related genes have resulted in poorer prognoses, based on online data. Based on platelet adhesion-related genes, patients with COAD were classified into two clusters by NMF-based clustering analysis. Cluster2 had a better overall survival, when compared to Cluster1. The gene copy number and enrichment analysis results revealed that two pathways were differentially enriched. In addition, the differentially expressed genes between these two clusters were enriched for POU6F1 in the transcription factor signaling pathway, and for MATN3 in the ceRNA network. Finally, a prognostic nomogram, which included the ALOX12 and ACTG1 genes, was established based on the platelet adhesion-related genes, with a concordance (C) index of 0.879 (0.848-0.910). Conclusion The mRNA expression-based NMF was used to reveal the potential role of platelet adhesion-related genes in COAD. The series of experiments revealed the feasibility of targeting platelet adhesion-associated gene therapy.
Collapse
Affiliation(s)
- Xiao-jv Chi
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, China
| | - Yi-bei Song
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, China
| | - Deng-he Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, China
| | - Li-qiang Wei
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, China
| | - Xin An
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zi-zhen Feng
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiao-hua Lan
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dong Lan
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chao Huang
- School of Information and Management, Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Tian S, Lu Y, Gao H, Chen Z, Niu M, Wang C, Liu B. Epirubicin may enhance the inhibition of hepatocellular carcinoma induced by iodine‐125 seeds through downregulating WNT pathway. Asia Pac J Clin Oncol 2022; 19:355-364. [DOI: 10.1111/ajco.13873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Shilin Tian
- Department of Interventional MedicineShandong Cancer Hospital and InstituteShandong First Medical UniversityShandong Academy of Medical Sciences JinanChina
| | - Yue Lu
- Cheeloo College of MedicineShandong University JinanChina
| | - Haifeng Gao
- Department of OncologyDongying People's Hospital DongyingChina
| | - Zitong Chen
- Cheeloo College of MedicineShandong University JinanChina
| | - Min Niu
- Department of Gastrointestinal SurgeryTraditional Chinese Medicine Hospital of Jiyang County JinanChina
| | - Changjun Wang
- Department of RadiologyPeople's Hospital of Jiyang County JinanChina
| | - Bin Liu
- Department of Interventional MedicineThe Second Hospital of Shandong University JinanChina
- Interventional Oncology Institute of Shandong University JinanChina
| |
Collapse
|
18
|
Sanati M, Binabaj MM, Ahmadi SS, Aminyavari S, Javid H, Mollazadeh H, Bibak B, Mohtashami E, Jamialahmadi T, Afshari AR, Sahebkar A. Recent advances in glioblastoma multiforme therapy: A focus on autophagy regulation. Biomed Pharmacother 2022; 155:113740. [PMID: 36166963 DOI: 10.1016/j.biopha.2022.113740] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 11/02/2022] Open
Abstract
Despite conventional treatment options including chemoradiation, patients with the most aggressive primary brain tumor, glioblastoma multiforme (GBM), experience an average survival time of less than 15 months. Regarding the malignant nature of GBM, extensive research and discovery of novel treatments are urgently required to improve the patients' prognosis. Autophagy, a crucial physiological pathway for the degradation and recycling of cell components, is one of the exciting targets of GBM studies. Interventions aimed at autophagy activation or inhibition have been explored as potential GBM therapeutics. This review, which delves into therapeutic techniques to block or activate autophagy in preclinical and clinical research, aims to expand our understanding of available therapies battling GBM.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Maryam Moradi Binabaj
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Seyed Sajad Ahmadi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Elmira Mohtashami
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Xing J, Qi L, Liu X, Shi G, Sun X, Yang Y. Roles of mitochondrial fusion and fission in breast cancer progression: a systematic review. World J Surg Oncol 2022; 20:331. [PMID: 36192752 PMCID: PMC9528125 DOI: 10.1186/s12957-022-02799-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/24/2022] [Indexed: 12/02/2022] Open
Abstract
Background Mitochondria play critical roles in cellular physiological activity as cellular organelles. Under extracellular stimulation, mitochondria undergo constant fusion and fission to meet different cellular demands. Mitochondrial dynamics, which are involved in mitochondrial fusion and fission, are regulated by specialized proteins and lipids, and their dysregulation causes human diseases, such as cancer. The advanced literature about the crucial role of mitochondrial dynamics in breast cancer is performed. Methods All related studies were systematically searched through online databases (PubMed, Web of Science, and EMBASE) using keywords (e.g., breast cancer, mitochondrial, fission, and fusion), and these studies were then screened through the preset inclusion and exclusion criteria. Results Eligible studies (n = 19) were evaluated and discussed in the systematic review. These advanced studies established the roles of mitochondrial fission and fusion of breast cancer in the metabolism, proliferation, survival, and metastasis. Importantly, the manipulating of mitochondrial dynamic is significant for the progresses of breast cancer. Conclusion Understanding the mechanisms underlying mitochondrial fission and fusion during tumorigenesis is important for improving breast cancer treatments.
Collapse
Affiliation(s)
- Jixiang Xing
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Luyao Qi
- The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Xiaofei Liu
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Guangxi Shi
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaohui Sun
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yi Yang
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
20
|
Liu X, Tao M. SSX2IP as a novel prognosis biomarker plays an important role in the development of breast cancer. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00273-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
21
|
Liao M, Qin R, Huang W, Zhu HP, Peng F, Han B, Liu B. Targeting regulated cell death (RCD) with small-molecule compounds in triple-negative breast cancer: a revisited perspective from molecular mechanisms to targeted therapies. J Hematol Oncol 2022; 15:44. [PMID: 35414025 PMCID: PMC9006445 DOI: 10.1186/s13045-022-01260-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/28/2022] [Indexed: 02/08/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of human breast cancer with one of the worst prognoses, with no targeted therapeutic strategies currently available. Regulated cell death (RCD), also known as programmed cell death (PCD), has been widely reported to have numerous links to the progression and therapy of many types of human cancer. Of note, RCD can be divided into numerous different subroutines, including autophagy-dependent cell death, apoptosis, mitotic catastrophe, necroptosis, ferroptosis, pyroptosis and anoikis. More recently, targeting the subroutines of RCD with small-molecule compounds has been emerging as a promising therapeutic strategy, which has rapidly progressed in the treatment of TNBC. Therefore, in this review, we focus on summarizing the molecular mechanisms of the above-mentioned seven major RCD subroutines related to TNBC and the latest progress of small-molecule compounds targeting different RCD subroutines. Moreover, we further discuss the combined strategies of one drug (e.g., narciclasine) or more drugs (e.g., torin-1 combined with chloroquine) to achieve the therapeutic potential on TNBC by regulating RCD subroutines. More importantly, we demonstrate several small-molecule compounds (e.g., ONC201 and NCT03733119) by targeting the subroutines of RCD in TNBC clinical trials. Taken together, these findings will provide a clue on illuminating more actionable low-hanging-fruit druggable targets and candidate small-molecule drugs for potential RCD-related TNBC therapies.
Collapse
Affiliation(s)
- Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hong-Ping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,Antibiotics Research and Re-Evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Fu Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
22
|
Yuan L, Cai Y, Zhang L, Liu S, Li P, Li X. Promoting Apoptosis, a Promising Way to Treat Breast Cancer With Natural Products: A Comprehensive Review. Front Pharmacol 2022; 12:801662. [PMID: 35153757 PMCID: PMC8836889 DOI: 10.3389/fphar.2021.801662] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is one of the top-ranked malignant carcinomas associated with morbidity and mortality in women worldwide. Chemotherapy is one of the main approaches to breast cancer treatment. Breast cancer initially responds to traditional first- and second-line drugs (aromatase inhibitor, tamoxifen, and carboplatin), but eventually acquires resistance, and certain patients relapse within 5 years. Chemotherapeutic drugs also have obvious toxic effects. In recent years, natural products have been widely used in breast cancer research because of their low side effects, low toxicity, and good efficacy based on their multitarget therapy. Apoptosis, a programmed cell death, occurs as a normal and controlled process that promotes cell growth and death. Inducing apoptosis is an important strategy to control excessive breast cancer cell proliferation. Accumulating evidence has revealed that natural products become increasingly important in breast cancer treatment by suppressing cell apoptosis. In this study, we reviewed current studies on natural product–induced breast cancer cell apoptosis and summarized the proapoptosis mechanisms including mitochondrial, FasL/Fas, PI3K/AKT, reactive oxygen species, and mitogen-activated protein kinase–mediated pathway. We hope that our review can provide direction in the search for candidate drugs derived from natural products to treat breast cancer by promoting cell apoptosis.
Collapse
Affiliation(s)
- Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Liang Zhang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Sijia Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Pan Li
- Department of Pharmacy, Fengdu County Hospital of Traditional Chinese Medicine, Chongqing, China
- *Correspondence: Xiaoli Li, ; Pan Li,
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
- *Correspondence: Xiaoli Li, ; Pan Li,
| |
Collapse
|
23
|
Ogbu SC, Rojas S, Weaver J, Musich PR, Zhang J, Yao ZQ, Jiang Y. DSTYK Enhances Chemoresistance in Triple-Negative Breast Cancer Cells. Cells 2021; 11:97. [PMID: 35011659 PMCID: PMC8750327 DOI: 10.3390/cells11010097] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 12/14/2022] Open
Abstract
Breast cancer, as the most prevalent cancer in women, is responsible for more than 15% of new cancer cases and about 6.9% of all cancer-related death in the US. A major cause of therapeutic failure in breast cancer is the development of resistance to chemotherapy, especially for triple-negative breast cancer (TNBC). Therefore, how to overcome chemoresistance is the major challenge to improve the life expectancy of breast cancer patients. Our studies demonstrate that TNBC cells surviving the chronic treatment of chemotherapeutic drugs show significantly higher expression of the dual serine/threonine and tyrosine protein kinase (DSTYK) than non-treated parental cells. In our in vitro cellular models, DSTYK knockout via the CRISPR/Cas9-mediated technique results in apoptotic cell death of chemoresistant cells upon drug treatment. Moreover, DSTYK knockout promotes chemotherapeutic drug-induced tumor cell death in an orthotopic mouse model. These findings suggest that DSTYK exerts an important and previously unknown role in promoting chemoresistance. Our studies provide fundamental insight into the role of DSTYK in chemoresistance in TNBC cells and lay the foundation for the development of new strategies targeting DSTYK for improving TNBC therapy.
Collapse
Affiliation(s)
- Stella C. Ogbu
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (S.C.O.); (S.R.); (J.W.); (P.R.M.)
| | - Samuel Rojas
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (S.C.O.); (S.R.); (J.W.); (P.R.M.)
| | - John Weaver
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (S.C.O.); (S.R.); (J.W.); (P.R.M.)
| | - Phillip R. Musich
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (S.C.O.); (S.R.); (J.W.); (P.R.M.)
| | - Jinyu Zhang
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA; (J.Z.); (Z.Q.Y.)
| | - Zhi Q. Yao
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA; (J.Z.); (Z.Q.Y.)
| | - Yong Jiang
- Department of Biomedical Sciences, J. H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (S.C.O.); (S.R.); (J.W.); (P.R.M.)
| |
Collapse
|