1
|
Tan CH, Sivakumar H, Luo DG, Cen YX. Integrative Network Pharmacology and Molecular Docking Analyses on the Mechanisms of San-Zhong-Kui-Jian-Tang in Treating Oral Squamous Cell Carcinoma. Curr Med Sci 2025:10.1007/s11596-025-00067-7. [PMID: 40526272 DOI: 10.1007/s11596-025-00067-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 06/19/2025]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is an aggressive cancer with a high mortality rate. San-Zhong-Kui-Jian-Tang (SZKJT), a Chinese herbal formula, has long been used as an adjuvant therapy in cancer clinical practice. Although its therapeutic effects and molecular mechanisms in OSCC have been previously elucidated, the potential interactions and mechanisms between the active phytochemicals and their therapeutic targets are still lacking. METHODS The present study employed network pharmacology and topology approaches to establish a "herbal ingredients-active phytochemicals-target interaction" network to explore the potential therapeutic targets of SZKJT-active phytochemicals in the treatment of OSCC. The role of the target proteins in oncogenesis was assessed via GO and KEGG enrichment analyses, and their interactions with the active phytochemicals of SZKJT were calculated via molecular docking and dynamic simulations. The pharmacokinetic properties and toxicity of the active phytochemicals were also predicted. RESULTS: A total of 171 active phytochemicals of SZKJT fulfilled the bioavailability and drug-likeness screening criteria, with the flavonoids quercetin, kaempferol, and naringenin having the greatest potential. The 4 crucial targets of these active phytochemicals are PTGS2, TNF, BCL2, and CASP3, which encode cyclooxygenase-2, tumor necrosis factor (TNF), BCL-2 apoptosis regulator, and caspase-3, respectively. The interactions between phytochemicals and target proteins were predicted to be thermodynamically feasible and stable via molecular docking and dynamics simulations. Finally, the results revealed that the IL-6/JAK/STAT3 pathway and TNF signaling via NF-κB are the two prominent pathways targeted by SZKJT. CONCLUSION In summary, this study provides computational data for in-depth exploration of the mechanism by which SZKJT activates phytochemicals to treat OSCC.
Collapse
Affiliation(s)
- Chun Hoe Tan
- Department of Biotechnology, School of Nursing and Applied Sciences, Lincoln University College, 47301, Selangor, Malaysia.
| | - Haresh Sivakumar
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Da-Gui Luo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yu-Xin Cen
- The First Clinical Medical Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
2
|
ZHANG Y, LYU W, XU X, LI J, HU S, WANG Y. Clinical effect of Shugan Jieyu San for improving liver function and alleviating depression in patients with triple negative breast cancer. J TRADIT CHIN MED 2025; 45:633-638. [PMID: 40524302 PMCID: PMC12134316 DOI: 10.19852/j.cnki.jtcm.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/20/2024] [Indexed: 06/19/2025]
Abstract
OBJECTIVE To evaluate the therapeutic effect of a self-formulated Traditional Chinese Medicine (TCM), Shugan Jieyu San, on improvement of liver function and depression alleviation in the patients with triple negative breast cancer (TNBC). METHODS A total of 60 patients diagnosed with TNBC and depression were enrolled and randomly assigned to either the control group (receiving routine tumor treatment and duloxetine) or the treatment group (receiving the TCM in addition to routine treatment). RESULTS Both treatment and control groups showed a reduction in depressive symptoms and improved daily living abilities after treatment. However, the treatment group demonstrated significantly better outcomes compared to the control group. Furthermore, the levels of dopamine and serotonin in the serum increased in both groups after 8 weeks of treatment, while the treatment group exhibited superior results. CONCLUSIONS This TCM showed promising results in reducing depressive symptoms and improving daily abilities in patients with TNBC and comorbid depression, which was verified by the increase in serum levels of dopamine and serotonin, suggesting the potential involvement of these neurotransmitters in the therapeutic effects of this TCM.
Collapse
Affiliation(s)
- Yang ZHANG
- 1 Department of Traditional Chinese Medicine, Cangzh ou Central Hospital, Cangzhou 061001, China
| | - Wang LYU
- 1 Department of Traditional Chinese Medicine, Cangzh ou Central Hospital, Cangzhou 061001, China
| | - Xin XU
- 1 Department of Traditional Chinese Medicine, Cangzh ou Central Hospital, Cangzhou 061001, China
| | - Jie LI
- 2 Thyroid and Breast Surgery Department, Cangzhou Central Hospital, Cangzhou 061001, China
| | - Shengli HU
- 3 Information construction office, Cangzhou Central Hospital, Cangzhou 061001, China
| | - Ying WANG
- 1 Department of Traditional Chinese Medicine, Cangzh ou Central Hospital, Cangzhou 061001, China
| |
Collapse
|
3
|
Zhang B, Fu D, Wang X, Hu X. Network-based analysis and experimental validation of identified natural compounds from Yinchen Wuling San for acute myeloid leukemia. Front Pharmacol 2025; 16:1591164. [PMID: 40520175 PMCID: PMC12162526 DOI: 10.3389/fphar.2025.1591164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 05/09/2025] [Indexed: 06/18/2025] Open
Abstract
Objective Traditional Chinese medicine (TCM) has garnered attention for its potential in cancer therapy. Yinchen Wuling San (YWLS), a classical herbal formula, has been traditionally used for liver-related conditions, but its bioactive components and molecular mechanisms relevant to hematologic malignancies such as acute myeloid leukemia (AML) remain unclear. This study aims to identify the active compounds and potential molecular targets of Yinchen Wuling San in the context of AML through network pharmacology analysis, and to experimentally validate the effects of selected candidate compounds in AML models. Methods Active ingredients from six YWLS herbs were screened via the TCMSP database using oral bioavailability ≥30% and DL ≥0.18 thresholds. Targets were predicted using SwissTargetPrediction, and AML-related genes were obtained from DisGeNET and GeneCards. Key overlapping targets were analyzed via STRING PPI networks and GO/KEGG enrichment. Molecular docking was performed between three core compounds (genkwanin, isorhamnetin, quercetin) and hub proteins (e.g., SRC) using Sybyl-X. ADME profiles were predicted using SwissADME, and molecular dynamics simulations (GROMACS) assessed complex stability. These compounds were further evaluated in vitro (viability, apoptosis, cell cycle, RT-qPCR, flow cytometry) and in vivo using an AML xenograft mouse model. Results Of 621 YWLS targets, 113 overlapped with 1,247 AML-related genes. PPI analysis identified hub genes, including AKT1, SRC, and EGFR. Enrichment analysis highlighted PI3K-AKT, MAPK, and JAK-STAT pathways. Genkwanin, isorhamnetin, and quercetin were predicted to target SRC, with strong molecular docking affinities. ADME analysis suggested favorable pharmacokinetics, and molecular dynamics simulations confirmed structural stability. In vitro, these compounds exhibited dose-dependent cytotoxicity, induced apoptosis, modulated the cell cycle, and downregulated SRC expression. Notably, Genkwanin promoted CD8+ T cell proliferation and inhibited leukemia growth, improving survival in a leukemia xenograft model. Conclusion YWLS compounds, particularly Genkwanin, exhibit significant anti-leukemic activity via apoptosis induction, cell cycle modulation, and promote T cells proliferation. Genkwanin emerges as a promising therapeutic candidate for AML, warranting further clinical investigation.
Collapse
Affiliation(s)
- Biyu Zhang
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
- School of Medicine, Jiujiang University, Jiujiang, China
| | - Denggang Fu
- Medical University of South Carolina, Charleston, SC, United States
| | - Xin Wang
- School of Medicine, Jiujiang University, Jiujiang, China
| | - Xuelei Hu
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| |
Collapse
|
4
|
Shi W, Wang Z, Yu Z, Shen Y, Xin W, Chen W. Qingyihuaji formula reprograms metabolism to suppress pancreatic cancer growth and progression through LINC00346-OMA1-ATF4 signaling. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119893. [PMID: 40294662 DOI: 10.1016/j.jep.2025.119893] [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: 05/06/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qingyihuaji Formula (QYHJ) has been used to treat human pancreatic cancer for many years and are fully documented in the Pharmacopoeia of the People's Republic of China (2020 Edition), however, its pharmacological mechanisms remain largely unknown. AIM OF THE STUDY Here, we aimed to provide evidences for uncovering the underlying molecular mechanisms of QYHJ for pancreatic cancer management. MATERIALS AND METHODS Bioinformatic analysis, quantitative real-time PCR, western blotting, glucose consumption, immunofluorescence and glycolytic activity assay were performed to determine the underlying mechanisms. The effects of QYHJ treatment, overexpression or knockdown of LINC00346 and ATF4 on the cell proliferation, migration, cellular ROS, apoptosis and metabolism were investigated. A xenograft mouse model was further established to evaluate the mechanism in vivo. RESULTS We found that QYHJ inhibits LINC00346-OMA1-ATF4 signal transduction and aerobic glycolysis in pancreatic cancer cells. Overexpression of LINC00346 and ATF4 reversed the inhibition of glycolytic metabolism and growth-suppressive effects after QYHJ treatment in vitro and in vivo. Moreover, there was a significant negative correlation between expression levels of LINC00346-OMA1 with overall survival in patients with pancreatic cancer and a positive correlation between OMA1 and ATF4 levels in human tumors. CONCLUSION Our findings indicate QYHJ shows the ability to suppress pancreatic cancer growth and progression, which is in mediated through antagonization of LINC00346 and activation of OMA1-ATF4. Targeting LINC00346-OMA1-ATF4 signaling may be promising effective therapeutic strategies for pancreatic cancer intervention.
Collapse
Affiliation(s)
- Weidong Shi
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200433, China; Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Ziyu Wang
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200433, China; Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Zhengyong Yu
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200433, China; College of Notoginseng Medicine and Pharmacy, Wenshan University, Wenshan, China
| | - Yilan Shen
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200433, China; Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenfeng Xin
- College of Notoginseng Medicine and Pharmacy, Wenshan University, Wenshan, China.
| | - Wei Chen
- Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200433, China.
| |
Collapse
|
5
|
Li Y, Wang F. Research Progress on Traditional Chinese Medicines Reversing Multidrug Resistance and Mechanisms in Lung Cancer. Cancer Biother Radiopharm 2025. [PMID: 40402865 DOI: 10.1089/cbr.2025.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025] Open
Abstract
Lung cancer continues to be a primary contributor to cancer-related deaths globally, and multidrug resistance (MDR) poses a significant obstacle in its management. Traditional Chinese medicines (TCMs), recognized for their comprehensive therapeutic strategies and low incidence of adverse effects, have garnered attention due to their capacity to mitigate MDR in cancer cells. Nevertheless, deciphering the precise mechanisms through which TCMs reverse MDR in lung cancer presents a substantial scientific challenge. The objective of this review is to examine prevalent manifestations of MDR in lung cancer and underscore recent advancements in understanding how TCMs might surmount this form of resistance. The review begins by investigating the unique characteristics of TCMs and their pivotal function in reversing MDR in lung cancer. Subsequently, it explores various forms of MDR in lung cancer, such as aberrant expression of cell membrane transport proteins, dysregulation of intracellular enzyme systems, disrupted apoptosis, and heightened cellular repair mechanisms, emphasizing their detrimental impact on lung cancer treatment outcomes. Central to this review is a thorough analysis of the intricate mechanisms by which TCMs counteract MDR, along with an assessment of their efficacy in lung cancer therapy. Based on this analysis, the review offers insights into potential future research directions for utilizing TCMs to overcome MDR. This review seeks to provide a thorough examination of the role of TCMs in reversing MDR in lung cancer and to stimulate additional research into their clinical applications.
Collapse
Affiliation(s)
- Yuying Li
- School of Clinic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chengdu Shuangliu District Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Fei Wang
- School of Clinic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
Yang P, Wang X, Yang J, Yan B, Sheng H, Li Y, Yang Y, Wang J. AI-Driven Multiscale Study on the Mechanism of Polygonati Rhizoma in Regulating Immune Function in STAD. ACS OMEGA 2025; 10:19770-19796. [PMID: 40415801 PMCID: PMC12096195 DOI: 10.1021/acsomega.5c00981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/27/2025]
Abstract
Polygonati Rhizoma, a traditional Chinese medicine, has demonstrated immunomodulatory and anticancer properties, yet its precise mechanisms in stomach adenocarcinoma (STAD) remain underexplored. This study aims to uncover the multitarget mechanisms of Polygonati Rhizoma in regulating the tumor immune microenvironment in STAD using artificial intelligence (AI)-driven network pharmacology, bioinformatics, and single-cell RNA sequencing, offering new insights into its immunotherapeutic potential. This study harnessed the power of AI to unravel the molecular mechanisms underlying Polygonati Rhizoma's effects. AI-driven methodologies screened 38 putative constituents, retaining 8 based on ADME criteria. Machine Learning algorithms predicted potential targets, which were cross-referenced with 5,569 immune-related genes from GeneCards, revealing 52 immune-associated targets. Differential expression analysis of the STAD data set identified 18 overlapping DEGs with prognostic significance and immune cell infiltration correlations. Key targets (AKT1, TP53, PTGS2 and VEGFA) emerged as central nodes in the network, with AI-assisted molecular docking confirming strong binding affinities, particularly between diosgenin and these core proteins. Molecular dynamics simulations further validated these interactions. Single-cell RNA sequencing revealed distinct target-gene expression patterns across malignant, stromal, and immune cell subsets in digestive-system tumors. In vitro, Polygonati Rhizoma extract significantly inhibited HGC-27 cell viability and increased intracellular ROS levels. These findings underscore the critical role of AI in integrating multiscale analyses, unveiling a multitarget immunomodulatory and antitumor mechanism for Polygonati Rhizoma in STAD, and providing a foundation for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Peizheng Yang
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| | - Xiangyu Wang
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| | - Jianhua Yang
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
- School
of Medical Informatics Engineering, Anhui University of Chinese
Medicine, Hefei, Anhui230038, China
| | - Biaobiao Yan
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| | - Haiyang Sheng
- Global
Biometrics and Data Sciences, Bristol Myers
Squibb, Lawrenceville, New Jersey10154, United States
| | - Yan Li
- Key
Laboratory of Industrial Ecology and Environmental Engineering (MOE),
Department of Materials Sciences and Chemical Engineering, Dalian University of Technology, Dalian, Liaoning116023, China
| | - Yinfeng Yang
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
- School
of Medical Informatics Engineering, Anhui University of Chinese
Medicine, Hefei, Anhui230038, China
| | - Jinghui Wang
- School
of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui230038, China
- Anhui
Provincial Key Laboratory of Chinese Medicinal Formula, Anhui University of Chinese Medicine, Hefei230038, China
| |
Collapse
|
7
|
Teng G, Zhang M, Pan Y, Karampoor S, Mirzaei R. Modulating the tumor microenvironment: The role of traditional Chinese medicine in improving lung cancer treatment. Open Life Sci 2025; 20:20251100. [PMID: 40417000 PMCID: PMC12103189 DOI: 10.1515/biol-2025-1100] [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: 07/23/2024] [Revised: 02/02/2025] [Accepted: 03/17/2025] [Indexed: 05/27/2025] Open
Abstract
The holistic approach of traditional Chinese medicine (TCM) has been increasingly being focused on as a potential adjuvant to conventional lung cancer therapies in an attempt at modulating the tumor microenvironment (TME). Covering a diverse range of herbal medicine, acupuncture, and dietary therapy, TCM brings a unique perspective to influencing the TME. Importantly, the study has found the effects of specific TCM compounds, such as cantharidin, boehmenan, shikonin, and salidroside, on lung cancer in the TME. These compounds interact intricately with key apoptotic regulators, oxidative stress pathways, and inflammation-related mechanisms, suggesting their potential role in enhancing conventional therapies. TCM compounds could modulate a variety of cellular and molecular pathways, potentially inhibiting tumor proliferation, invasion, and metastasis. Besides, the practices of TCM alleviate the side effects of conventional treatments and enhance immune function, hence promoting the quality of life among lung cancer patients. In this regard, this review gives a contemporary account of the state of affairs on the part of TCM within the framework of the treatment of lung cancer with reference to its recent developments, and diverse roles.
Collapse
Affiliation(s)
- Geling Teng
- Department of Respiratory and Critical Care Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, 250013, China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yuling Pan
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
8
|
Hsiao YC, Chung CH, Lin CJ, Tsai LJ, Kuo SW, Lu CH, Chien WC. Role of traditional Chinese medicine on fracture, hospitalization, and total mortality risks in patients with hyperthyroidism and osteoporosis. Medicine (Baltimore) 2025; 104:e42484. [PMID: 40388732 PMCID: PMC12091615 DOI: 10.1097/md.0000000000042484] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 05/02/2025] [Indexed: 05/21/2025] Open
Abstract
Previous studies have confirmed that hyperthyroidism is one of the common causes of secondary osteoporosis and can aggravate the disease severity in patients with osteoporosis. This study is mainly based on the Taiwan National Health Insurance Database and through big data analysis shows that combining traditional Chinese medicine (TCM) treatment can help the health of patients with hyperthyroidism and osteoporosis. There were 4980 patients who received TCM treatment and 19,920 controls who did not receive TCM treatment selected from Taiwan National Health Insurance Database in a 4:1 ratio of gender, age, and index year. Cox proportional hazards analyzes were performed to compare fracture, inpatient, and all-cause mortality over an average follow-up period of 15 years. A total of 4745/5823/3487 enrolled subjects (19.06%/23.39%/14.00%) suffered fractures/hospitalization/all-cause death which TCM group was 452/987/511 (15.10%/19.82%/10.26%); control group was 3993/4836/2976 (20.05%/24.28%/14.94%). Cox proportional hazards regression analysis showed that subjects in the TCM group had lower fractures, hospital mortality, and all-cause mortality (adjusted HR = 0.563; 95% confidence intervals [CI] = 0.392-0.680, P < .001; adjusted HR = 0.614; 95% CI = 0.474-0.714, P < .001; adjusted HR = 0.691; 95% CI = 0.569-0.792, P < .001). Kaplan-Meier analysis showed that the cumulative risk of fracture, hospitalization and death were lower in TCM group with significant differences (all log-rank P < .001). The analysis of this study shows that patients with hyperthyroidism and osteoporosis even with comorbidity of atrial fibrillation, atrial flutter, or heart failure combined with TCM treatment are associated with a lower risk of fractures, hospitalization, or all-cause death, providing clinicians with a treatment option.
Collapse
Affiliation(s)
- Yuan-Chih Hsiao
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Chi-Hsiang Chung
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan, ROC
| | - Chien-Jung Lin
- Department of Chinese Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Li-Jen Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Shi-Wen Kuo
- Department of Internal Medicine, The Division of Endocrinology and Metabolism, Taipei Tzu Chi Hospital, Taiwan, ROC
| | - Chieh-Hua Lu
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Wu-Chien Chien
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
9
|
Lu T, Xu J, Cao Q, Zhang J, Chen J, Lin L, Shan B, Wei J. Culturally Adapted Shared Decision-Making Tool for Breast Cancer Clinical Trials in China: A Nurse-Led Approach. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2025:10.1007/s13187-025-02646-7. [PMID: 40369337 DOI: 10.1007/s13187-025-02646-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
This study aimed to develop and validate a culturally adapted, nurse-led shared decision-making question prompt list (SDM-QPL) to enhance decision-making support for Chinese breast cancer patients considering clinical trials. A mixed-methods approach was employed, involving a systematic literature review (52 studies), semi-structured interviews with 15 patients and 8 healthcare providers, a three-round Delphi consensus with 13 experts, and psychometric testing. The process yielded a 40-item QPL tailored to cultural needs, such as familial decision-making and Traditional Chinese Medicine (TCM) use. The SDM-QPL, organized into five modules, achieved strong expert consensus (Kendall's W = 0.82, P < 0.001) and retained 92% of nurse-sensitive items. Psychometric evaluation confirmed high validity (S-CVI/Ave = 0.89) and reliability (Cronbach's α = 0.91). Qualitative analysis identified key barriers: information overload (93.3%), risk misperceptions (86.7%), and nursing communication gaps (80.0%). This nurse-led SDM-QPL, grounded in Elwyn's Three-Talk Model, addresses China's unique cultural and structural challenges, fostering triadic dialogue among patients, families, and providers. The tool empowers oncology nurses to bridge communication gaps, improve trial participation equity, and support patient-centered care in collectivist settings.
Collapse
Affiliation(s)
- Ting Lu
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China.
| | - Junxia Xu
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Qingrong Cao
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Jing Zhang
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Jian Chen
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Lin Lin
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Benjie Shan
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Jie Wei
- Department of Comprehensive Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| |
Collapse
|
10
|
Chang CM, Jassi C, Kuo WW, Lin YJ, Chiu CH, Wu TT, Kuo CH, Ho TJ, Lin SZ, Huang CY. Kang Ru enhances paclitaxel's efficacy against breast cancer progression. Am J Cancer Res 2025; 15:2180-2192. [PMID: 40520866 PMCID: PMC12163460 DOI: 10.62347/xmgx2636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/10/2025] [Indexed: 06/18/2025] Open
Abstract
Breast cancer and its associated drug resistance present significant clinical challenges. From a translational medicine perspective, Traditional Chinese Herbal Medicine (TCHM) offers promising integrative approach to bridge the experimental findings with clinical application, enhancing cancer treatment outcomes and mitigate drug resistance. In this study, we evaluated a TCM herbal formulation, Kang Ru (KR), comprising Artemisia argyi, Ohwia caudata, and Scoparia dulcis Linn, each traditionally and experimentally recognized for their potential therapeutic properties. We aimed to evaluate the anti-cancer effects of KR on breast cancer and its potential to enhance the efficacy of the chemotherapeutic agent paclitaxel. Using breast cancer cell lines we evaluated the effects of KR, both alone and in combination with paclitaxel on cell growth, viability, migration, and invasion. We further evaluated the potential of KR in enhancing paclitaxel to reduce tumor progression using a breast cancer xenograft model in mice. Our findings demonstrated that KR significantly inhibited cell growth and migration in breast cancer cell lines. Moreover, the combination of KR with paclitaxel demonstrated synergistic effects, effectively reducing cell viability, inducing cell apoptosis and inhibiting cell migration. In an animal model, the combination of KR and paclitaxel significantly enhanced breast cancer suppression, corroborating the in vitro findings. In conclusion, KR is a novel Chinese herbal formulation, with significant anti-cancer potential, enhancing paclitaxel's efficacy in inhibiting breast cancer progression. These findings suggest that KR could be a promising adjunctive strategy to improve breast cancer treatment and warrant further translational research to facilitate its clinical development as a complementary therapeutic option.
Collapse
Affiliation(s)
- Chun-Ming Chang
- Department of General Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationHualien 970, Taiwan
- School of Medicine, Tzu Chi UniversityHualien, Taiwan
| | - Chikondi Jassi
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi HospitalHualien 970, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical UniversityTaichung 40676, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical UniversityTaichung 406, Taiwan
| | - Yu-Jung Lin
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi HospitalHualien 970, Taiwan
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationNew Taipei City, Taiwan
| | - Chih-Hao Chiu
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi HospitalHualien 970, Taiwan
| | - Tsai-Ting Wu
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi HospitalHualien 970, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of TaipeiTaipei 10048, Taiwan
- Department of Kinesiology and Health Science, College of William and MaryWilliamsburg, VA 23185, USA
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationHualien 970, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationHualien 970, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Buddhist Tzu Chi Medical FoundationHualien 970, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical FoundationHualien 970, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical FoundationHualien 970, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondria Related Diseases Research Center, Hualien Tzu Chi HospitalHualien 970, Taiwan
- Graduate Institute of Biomedicine, China Medical UniversityTaichung 406040, Taiwan
- Department of Biotechnology, Asia UniversityTaichung 413, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi UniversityHualien 970, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical UniversityTaichung 404, Taiwan
| |
Collapse
|
11
|
Xiao H, Raza F, Li K, Song J, Zafar H, Yang S, Su J, Qiu M. Cell membrane derived biomimetic nanomedicine for precision delivery of traditional Chinese medicine in cancer therapy. J Control Release 2025; 383:113829. [PMID: 40355044 DOI: 10.1016/j.jconrel.2025.113829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
The rapidly developing modern nanotechnology has brought new vitality to the application of traditional Chinese medicine (TCM), improving the pharmacokinetics and bioavailability of unmodified natural drugs. However, synthetic materials inevitably introduce incompatibilities. This has led to focusing on biomimetic drug delivery systems (DDS) based on biologically derived cell membranes. This "top-down" approach to nanomedicine preparation is simple and effective, as the inherited cell membranes and cell surface substances can mimic nature when delivering drugs back into the body, interacting similarly to the source cells at the biological interface. The concept of biologically derived TCM and biomimetic membranes aligns well with nature, the human body, and medicine, thereby enhancing the in vivo compatibility of TCM. This review focused on the recent progress using biomimetic membranes for TCM in cancer therapy, emphasizing the effective integration of biomimetic nanomedicine and TCM in applications such as cancer diagnosis, imaging, precision treatment, and immunotherapy. The review also provided potential suggestions on the challenges and prospects in this field.
Collapse
Affiliation(s)
- Hang Xiao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kunwei Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinpu Song
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiqi Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
12
|
Yang Z, Wu M, Zhou X, Luo J, Liu Y, Li L. Network pharmacology study on the mechanism of Curcumae Rhizoma in the treatment of non-small cell lung cancer. Medicine (Baltimore) 2025; 104:e42366. [PMID: 40355237 PMCID: PMC12074036 DOI: 10.1097/md.0000000000042366] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 04/18/2025] [Indexed: 05/14/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) poses a significant threat to public health worldwide. Curcumae Rhizoma (CR) has potent therapeutic potential in different cancers. However, the mechanism of CR treating NSCLC remains unclear. In this study, a network pharmacology-based strategy is followed to address the issue. The targets related to CR or NSCLC were obtained from multiple online public databases. Compound-target network was constructed using Cytoscape. Protein-protein interaction (PPI) was analyzed by STRING. Key transcription factors were explored in TRRUST. Gene ontology (GO) function and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis were accomplished in Metascape. The druglikeness of compounds was tested in Molinspiration Cheminformatics Software. Autodock Vina was used for molecular docking. Molecular dynamic (MD) simulation was performed using Gromacs. There were 104 overlapped targets considered as key targets of CR treating NSCLC. The key components of CR, including reynosin, (4S,5S)-13-hydroxygermacrone 4,5-epoxide, and (E)-1,7-bis(4-hydroxyphenyl)-6-hepten-3-one, were screened by topological parameters and bioactivity scores. Central clustered targets in PPI network (epidermal growth factor receptor [EGFR], SRC, JAK2, and mitogen-activated protein kinase 3 [MAPK3]) were identified as critical therapeutic targets of CR. GO and KEGG enrichment analysis suggested that therapeutic effect of CR on NSCLC involved various biological processes, cellular components, and molecular functions, and pathways in cancer, JAK-STAT signaling pathway, and p53 signaling pathway were strongly related. Molecular docking and MD simulation suggested that key compounds in CR had high binding affinity to critical NSCLC targets, like EGFR, JAK2, SRC, and MAPK3, with stable complexes formed. This study revealed key components and mechanism of CR treating NSCLC based on a network pharmacology-driven strategy, providing a reference for in-depth study on treating NSCLC.
Collapse
Affiliation(s)
- Zhirui Yang
- Department of Nuclear Medicine, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Mingquan Wu
- Department of Pharmacy, Sichuan Orthopedic Hospital, Chengdu, Sichuan, China
| | - Xin Zhou
- Department of Nuclear Medicine, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Jin Luo
- Department of Nuclear Medicine, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Yi Liu
- Department of Nuclear Medicine, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Lin Li
- Department of Nuclear Medicine, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Lei Y, Chen C. Bibliometric analysis of traditional Chinese medicine in cancer treatment via immune system modulation (2015-2025). Front Immunol 2025; 16:1581885. [PMID: 40406101 PMCID: PMC12095241 DOI: 10.3389/fimmu.2025.1581885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Objective The application of Traditional Chinese Medicine (TCM) in treating cancer by regulating the immune system has garnered significant attention in the academic community. However, comprehensive quantitative analyses in this field remain limited. This study aims to assess the research progress and key trends over the past decade, providing a framework for future studies. Methods A comprehensive literature search was conducted on the application of TCM in treating cancer by regulating the immune system from 2015 to 2025 using the Web of Science database. The search terms mainly included cancer, Traditional Chinese Medicine, immunity and so on. Data were analyzed and visualized using Origin, R software, VOSviewer, and CiteSpace. Results A total of 2,459 articles were included in the analysis. The number of related publications has steadily increased since 2015. China leads in publication volume and plays a crucial role in international collaboration. The Journal of Ethnopharmacology is the leading journal in this field, publishing a substantial number of highly cited studies. Key research areas include keywords such as "apoptosis," "expression," "inflammation," "extract," "in vitro," "activation," "antioxidant," and "NF-kappa B," focusing on exploring the role, mechanisms, and efficacy of TCM in modulating immune responses. Conclusion Research interest in TCM's role in treating cancer through immune system regulation continues to grow, underscoring its potential in cancer therapy. Current research primarily focuses on the mechanisms by which TCM treats cancer through the modulation of immune cell functions, inhibition of tumor immune evasion, and regulation of immune-related signaling pathways. It also explores its clinical applications and the potential for enhancing the efficacy of immunotherapy.
Collapse
Affiliation(s)
| | - Chunyan Chen
- Department of Pharmacy, Shanghai Public Health Clinical Center, Fudan
University, Shanghai, China
| |
Collapse
|
14
|
Yang Z, Zhao K, Li X, Yanzhang R, Zhang H, Yu Y, Yan M, Fang S, Li T, Li H, Chu X, Han S, Zhang Z, Teng J, Jin G, Guo Z. ZIP8 modulates ferroptosis to drive esophageal carcinoma progression. Cell Death Dis 2025; 16:366. [PMID: 40328750 PMCID: PMC12056185 DOI: 10.1038/s41419-025-07692-z] [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: 08/18/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Ferroptosis, a regulated form of cell death characterized by iron-dependent phospholipid peroxidation, remains poorly understood in the context of esophageal cancer development and its regulatory mechanisms. Through comprehensive bioinformatic analyses, we identified ferroptosis-related pathways as crucial mediators in esophageal cancer progression, with ZIP8 emerging as a key regulatory element. We observed significant upregulation of ZIP8 in esophageal cancer specimens, which correlated with poor clinical outcomes. Functional studies demonstrated that ZIP8 depletion significantly attenuated cellular proliferation in vitro. Mechanistically, elevated ZIP8 expression enhanced zinc-dependent phosphorylation of CREB, leading to upregulation of the ferroptosis suppressor GPX4 and inhibition of this iron-dependent cell death modality. Significantly, we discovered that the natural compound Nobiletin targeted ZIP8, inhibiting Esophageal squamous cell carcinoma (ESCC) cell growth in vitro and in vivo. Our findings demonstrate ZIP8 as a potential therapeutic target in ESCC and suggest that promoting ferroptosis through ZIP8 inhibition may represent a novel anti-cancer strategy for ESCC therapy.
Collapse
Affiliation(s)
- Zhaojie Yang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Kexin Zhao
- China-US (Henan) Hormel Cancer Institute, No.126, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangping Li
- Department of Clinical laboratory, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Ruoping Yanzhang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Huijun Zhang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Yin Yu
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Mingyang Yan
- China-US (Henan) Hormel Cancer Institute, No.126, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shaobo Fang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Department of Medical Imaging, Zhengzhou University People's Hospital& Henan Provincial People's Hospital, Zhengzhou, China
| | - Tao Li
- Healthy Management Center, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Hao Li
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Xiao Chu
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Siyuan Han
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Ziliang Zhang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Junyan Teng
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Guoguo Jin
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China.
- China-US (Henan) Hormel Cancer Institute, No.126, Zhengzhou, Henan, China.
- Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zhiping Guo
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China.
- Healthy Management Center, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China.
- Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
15
|
Men J, Wang J, Lv Z, Wang H, Shi H, Ma Y, Qiao Z, Chen J. Facile fabrication of chitosan-based molecular imprinted microspheres to adsorb selectively, release and anti-bacteria for berberine. Int J Biol Macromol 2025; 306:141592. [PMID: 40024400 DOI: 10.1016/j.ijbiomac.2025.141592] [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: 11/04/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Berberine hydrochloride (Ber), a bioactive compound widely found in the roots, rhizomes, stems and barks of Coptis chinensis, has demonstrated efficacy in treating many diseases, such as cancer, congestive heart failure, Alzheimer's disease, especially inflammatory caused by bacteria. The molecularly imprinted microspheres based on chitosan were fabricated to adsorb selectively, release and anti-bacteria of Ber. The Ber surface molecularly imprinted microspheres (Ber-PSSS@GCS-MIPs) were synthesized using crosslinked chitosan as matrix, Ber as template, and sodium 4-styrene sulfonate (SSS) as functional monomer via a redox surface-initiating system -NH2/-S2O82-. The microspheres were characterized by fourier transform infrared reflection (FTIR), scanning electron microscopy (SEM) and X-ray photoelectron spectroscopy (XPS). Adsorption kinetics, isotherms and imprinting factor were investigated, and the drug release performance and antibacterial activity were evaluated. As a result, via electrostatic interaction and "lock-key" imprinted cavities, the adsorption capacity of Ber-PSSS@GCS-MIPs reaches 185 mg/g at 2 h, significantly higher than 51 mg/g observed for non-imprinted microspheres. The adsorption of Ber-PSSS@GCS-MIPs. follows pseudo-second-order kinetics, with adsorption amount decreasing as temperature increases and salt concentration rises. Ber-PSSS@GCS-MIPs show excellent recognition and selectivity with an imprinting factor of 3.07, a selectivity factor exceeding to 2. The adsorption capacity remains at 82.4 % of three times cycles. The Ber-PSSS@GCS-MIPs loaded drug microspheres attain slow and sustained release for 70 % at 139 h. The relative antibacterial rate of Ber-PSSS@GCS-MIPs loaded Ber is higher than non-imprinted microsphere and control against S. aureus and E. coli.
Collapse
Affiliation(s)
- Jiying Men
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, PR China
| | - Ji Wang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, PR China
| | - Zhenyan Lv
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, PR China
| | - Hongfeng Wang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, PR China
| | - Hongxing Shi
- School of Chemical Engineering, Sichuan University, Chengdu 610065, PR China
| | - Yuan Ma
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, PR China
| | - Zongwen Qiao
- Department of Chemical Engineering, Shanxi Institute of Technology, Xian 710300, PR China
| | - Jianjun Chen
- School of Instrument and Electronics, North University of China, Taiyuan 030051, PR China.
| |
Collapse
|
16
|
Yang C, Ma H, Liang Z, Zhuang Y, Hu L, Zhang K, Huang L, Li M, Zhang S, Zhen Y. Cyclic RGD modified dextran-quercetin polymer micelles for targeted therapy of breast cancer. Int J Biol Macromol 2025; 308:142272. [PMID: 40118409 DOI: 10.1016/j.ijbiomac.2025.142272] [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: 08/02/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Quercetin is a natural flavonoid found in many plants which has various pharmacological activities including antitumor effect. However, the poor water solubility and bioavailability limit the potential benefits of quercetin for patients. Thus, modifying quercetin structure and developing actively targeted drug delivery systems are extremely important for tumor precision therapy. Herein, polymer-drug conjugates dextran-quercetin (D-Q) and cRGD-dextran (R-D) were synthesized by grafting quercetin and polypeptide cRGDfk (Arg-Gly-Asp-(D-Phe)-Lys) to dextran. Then cRGD-modified dextran-quercetin polymer micelles (R-D-Q) were constructed by self-assembling of D-Q and R-D. R-D-Q micelles possessed appropriate particle size (133.4 nm), nearly neutral potential (8.14 mV) and excellent drug-loading efficiency (13.1 %) and achieved higher cytotoxicity, apoptosis induction and penetration to human breast cancer MCF-7 cells than the micelles unmodified with cRGD, which were ascribed to cRGD-integrin mediated transcytosis. R-D-Q micelles effectively suppressed tumor growth in tumor-bearing mice by delivering more quercetin throughout the tumor tissue. And R-D-Q micelles could promote the apoptosis of tumor cells by activating p38 and JNK signal pathways and suppressing ERK signal pathway. In addition, R-D-Q micelles had no damage to normal tissues of mice at therapeutic dose. These results indicate promising prospects for R-D-Q micelles as an effective drug delivery system against tumor.
Collapse
Affiliation(s)
- Chunpeng Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Huiling Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ze Liang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ying Zhuang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Litao Hu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Kexin Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Leixiao Huang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Min Li
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, China; Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
17
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1283-1308. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [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: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
18
|
Li Y, Yang G, Li Q, Zhang Y, Zhang S, Zhou T, Wang X, Liu F, Miao Z, Qi Y, Zhang L, Liu Y, Su H. Guiqi Baizhu decoction enhances radiosensitivity in non-small cell lung cancer by inhibiting the HIF-1α/DNA-PKcs axis-mediated DNA repair. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156591. [PMID: 40054178 DOI: 10.1016/j.phymed.2025.156591] [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: 12/30/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Radiotherapy is one of the main treatments for non-small cell lung cancer (NSCLC), and radiosensitivity is a determinant of its efficacy. Therefore, enhancing the radiosensitivity is of great significance to improve the clinical efficacy of non-small cell lung cancer (NSCLC). PURPOSE This study intended to investigate the radiosensitisation effect and mechanism of Guiqi Baizhu decoction (GQBZD) on non-small cell lung cancer (NSCLC) and the role of hypoxia-inducible factor-1 alpha (HIF-1α)/DNA-dependent protein kinase catalytic subunit (DNA-PKcs) axis-mediated DNA non-homologous end joining (NHEJ) repair in NSCLC radiotherapy. STUDY DESIGN In vivo experimental model was Lewis subcutaneous transplantation tumor model of in C57 black 6 (C57BL/6) mice, and in vitro experimental models were A549, H1299 and H460 cells. METHODS In vivo experimental model was Lewis subcutaneous transplantation tumor model of in C57 black 6 (C57BL/6) mice. After the model was successfully established, the tumor was irradiated locally with 4 Gy X-ray, and 10.465 g/kg Guiqi Baizhu Decoction (GQBZD) was administered by gavage on the second day after irradiation for a total of 10 days. The morphological changes in tumour tissues were observed by HE staining, Ki67 levels in tumour tissues were detected by immunohistochemistry, the apoptosis in tumour cells were detected by Tunel staining. In vitro experimental models were different NSCLC cells (A549, H1299 and H460), irradiated by 2 Gy X-rays and then intervened with 5%, 10% and 20% Guiqi Baizhu Decoction (GQBZD)-containing serum for 24 h. A549 stably-transformed cell lines knocking down and overexpressing HIF-1α were also constructed by lentiviral transfection. The cell proliferation was detected by CCK-8 and clone formation, the apoptosis and cell cycle was detected by flow cytometry. Network pharmacology and transcriptomics to investigate key targets and pathways of GQBZD effects on NSCLC irradiation, further validated by immunofluorescence and Western blot. RESULTS In vivo experiments confirmed that GQBZD combined with irradiation could inhibit the growth of Lewis subcutaneous transplantation tumor, reduce the expression of Ki67 and promote the apoptosis of tumour cells. In vitro experiments confirmed that GQBZD combined with irradiation inhibited the proliferation of different NSCLC cells, promoted NSCLC cell apoptosis and G2/M-phase arrest, and induced the expression of phosphorylated histone H2AX (γ-H2AX) in NSCLC cells, which showed a good radiosensitisation effect. Mechanistically, GQBZD exerts its radiosensitisation effect on NSCLC mainly through the HIF-1α signalling pathway. Meanwhile, under irradiation conditions, the expression of HIF-1α and DNA-PKcs were positively correlated, and HIF-1α had a regulatory effect on DNA-PKcs, promoting DNA-PKcs-dependent non-homologous end joining (NHEJ) repair. In addition, GQBZD combined irradiation down-regulated the expression of HIF-1α, DNA-PKcs, and NHEJ repair-related proteins in NSCLC cells, while reversing the expression of HIF-1α, DNA-PKcs, and NHEJ repair-related proteins in overexpressing HIF-1α A549 cell, thereby enhancing radiosensitivity in NSCLC. CONCLUSION This study provides an in-depth exploration of the radiosensitisation effect of GQBZD and provides an important experimental basis for the study of Chinese medicine in the field of cancer radiosensitisation, and further extends the extensibility of GQBZD on the basis of the previous study.
Collapse
Affiliation(s)
- Yangyang Li
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Gengqiang Yang
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Qiyang Li
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Yiming Zhang
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao 266071, PR China
| | - Shangzu Zhang
- Hubei University of Chinese Medicine,Wuhan 430065, PR China
| | - Ting Zhou
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Xin Wang
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Fuxian Liu
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Zhiming Miao
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Yafeng Qi
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China
| | - Liying Zhang
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China.
| | - Yongqi Liu
- Gansu University of Chinese Medicine, Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Lanzhou 730000, PR China; Key Laboratory of Dunhuang Medicine and Translational Education Ministry of Education, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, PR China.
| | - Haixiang Su
- Gansu Provincial Cancer Hospital, Gansu Provincial Institute of Medical Sciences, Lanzhou, 730050,PR China.
| |
Collapse
|
19
|
Sulaiman C, George BP, Balachandran I, Abrahamse H. Cancer and Traditional Medicine: An Integrative Approach. Pharmaceuticals (Basel) 2025; 18:644. [PMID: 40430464 PMCID: PMC12114720 DOI: 10.3390/ph18050644] [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: 03/25/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Despite numerous advances in treatment, cancer still remains a leading cause of death worldwide. Given the significant health and economic burden this disease imposes, it is important to explore more effective treatment strategies. A major drawback of conventional cancer therapies is the persistence of drug resistance, adverse reactions to chemotherapy, digestive damage, reduced quality of life, and high treatment costs. To address these challenges, researchers have been investigating the utility of using complementary and alternative medicine (CAM) alongside conventional cancer treatments. Some of these CAM approaches have been reported to enhance patients' quality of life and reduce the severity of adverse effects from conventional therapies. This review explores the utility of traditional Chinese, Korean, Indian, Japanese, and South African medicines as supportive therapies for conventional cancer treatments. We also discuss the concept of integrative oncology and its global relevance, as CAM approaches typically emphasize whole-person care, including diet, lifestyle, and mental/emotional well-being. In addition, we identify key active phytochemicals and herbal medicines used in these traditional systems for cancer treatment. Our discussion aims to provide a foundation for future research into integrative oncology, fostering an interdisciplinary and complementary approach to cancer management.
Collapse
Affiliation(s)
- Cheruthazhakkat Sulaiman
- Phytochemistry Division, Centre for Medicinal Plants Research, Arya Vaidya Sala, Kottakkal 676503, Kerala, India; (C.S.); (I.B.)
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| | - Blassan P. George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| | - Indira Balachandran
- Phytochemistry Division, Centre for Medicinal Plants Research, Arya Vaidya Sala, Kottakkal 676503, Kerala, India; (C.S.); (I.B.)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| |
Collapse
|
20
|
Sun T, Wang T, Xia F, Xia L, Sun X, Jiang H, Xu P, Li P. Family Members' Perspectives on Integrative Chinese-Western Medicine in Cancer Care. J Multidiscip Healthc 2025; 18:2361-2373. [PMID: 40321884 PMCID: PMC12047228 DOI: 10.2147/jmdh.s509065] [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: 11/28/2024] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose Family members play a central supporting role in cancer patient management. This study aimed to assess the knowledge, attitudes, and practices (KAP) of cancer patients' family members regarding the use of integrative Chinese-Western medicine in cancer treatment. Proper knowledge is necessary for informed decision-making and providing adequate care and support. Methods This cross-sectional study was conducted between August 2022 and December 2022 and included family members of cancer patients. Results A total of 493 questionnaires were collected. The mean score of knowledge, attitudes, and practices were 5.87 ± 3.63 (possible range: 0-12), 59.90 ± 9.45 (possible range: 16-80), and 21.73 ± 3.27 (possible range: 6-30), respectively. Junior college (university programs lasting 2-3 years) or above (OR=2.87, 95% CI: 1.41-5.88, P=0.004), had family members attending Chinese Medicine/Integrated Chinese and Western Medicine departments (OR=1.94, 95% CI: 1.17-3.24, P=0.011), and possessed knowledge of TCM (OR=2.90, 95% CI: 1.76-4.79, P<0.001) were linked to good knowledge. Knowledge (OR=1.96, 95% CI: 1.23-3.11, P=0.005) and family members attending Chinese Medicine/Integrated Chinese and Western Medicine departments (OR=2.10, 95% CI: 1.27-3.47, P=0.004) were linked to positive attitudes. Attitude (OR=5.65, 95% CI: 3.57-8.93, P<0.001) was linked to positive practices. Conclusion The family members of cancer patients showed limited knowledge and moderate attitudes and practices. Efforts should be made to address the benefits of integrative Chinese-Western medicine and provide more access to education and resources related to TCM. Although research would be necessary, the present study could provide hints regarding the combination of modern and traditional medicine in other countries.
Collapse
Affiliation(s)
- Ting Sun
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, People’s Republic of China
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Ting Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Fan Xia
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People’s Republic of China
| | - Lin Xia
- Department of Internal Medicine-Oncology, Hefei BOE Hospital, Hefei, 230000, People’s Republic of China
| | - Xin Sun
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Haili Jiang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Panlin Xu
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Ping Li
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, People’s Republic of China
| |
Collapse
|
21
|
Jin W, Zhang Y, Pang S, Yao D, Huang Y. Off-label use of sodium cantharidinate and vitamin B6 injection in cancer: a protocol for a systematic review and meta-analysis. Syst Rev 2025; 14:82. [PMID: 40205599 PMCID: PMC11984136 DOI: 10.1186/s13643-025-02826-y] [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: 05/09/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND In China, sodium cantharidinate/vitamin B6 (SC/VB6) injection has been approved since 2002 for the treatment of lung cancer and primary liver cancer. In addition to these authorized indications, clinical application of SC/VB6 is also common in various other types of cancer. However, there is a lack of comprehensive understanding on this topic. Thus, this systematic review and meta-analysis aims to consolidate evidence regarding the efficacy and safety of off-label use of SC/VB6 in oncology. METHODS International databases including PubMed, Embase, Cochrane Library, Web of Science, and CINAHL Plus, as well as Chinese databases including China National Knowledge Infrastructure (CNKI), Chinese Biomedical Literature Database (CBM), and Wanfang, will be searched from the inception to 31 December 2024. Comparative studies that evaluated the add-on effect of SC/VB6 to conventional cancer treatments against the use of conventional treatments alone will be considered in the scope of this review. The primary outcomes are objective response rate and performance status. Secondary outcomes are disease control rate (DCR), progression-free survival (PFS), disease-free survival (DFS), overall survival (OS), and adverse events (AEs). Depending on heterogeneity, data will be synthesized using either the Mantel-Haenszel fixed-effect or the DerSimonian and Laird random-effect model. Subgroup analyses will be conducted for the following variables: type of cancer, study design, SC/VB6 dosage, treatment duration, and combined therapies, provided that each subgroup contains at least two studies. Sensitivity analyses will be performed on efficacy outcomes. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) will be utilized to appraise the overall quality of evidence. DISCUSSION This review will encompass both randomized controlled trials (RCTs) and cohort studies, thereby enabling us to synthesize and assess evidence across experimental and real-world observational settings. Our findings will contribute to a better understanding on the benefit-risk profile regarding the off-label use of SC/VB6 in oncology, guiding the trajectory of future research, and offering a robust scientific foundation to inform clinical and regulatory decision-making process. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42024504977.
Collapse
Affiliation(s)
- Wenzhen Jin
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Yibin Zhang
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Shu Pang
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Dongdong Yao
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China
| | - Yiwen Huang
- Scientific Affairs Department, Clinflash Healthcare Technology (Jiaxing) Co., Ltd, Room 813, 999 West Zhongshan Road, Huawen Plaza, Shanghai, 200051, People's Republic of China.
| |
Collapse
|
22
|
Wu R, Yuan H, Wang Y, Gou X, Hou W, Zhou Z, Wang X, Deng X, Wang C, Wang H, Yan J. Norcantharidin inhibits TOP2A expression via H3K27me3 mediated epigenetic regulation to alleviate the progression of hepatocellular carcinoma. Front Pharmacol 2025; 16:1541298. [PMID: 40271060 PMCID: PMC12015943 DOI: 10.3389/fphar.2025.1541298] [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: 12/07/2024] [Accepted: 03/06/2025] [Indexed: 04/25/2025] Open
Abstract
Background Norcantharidin (NCTD), a bioactive compound derived from traditional Chinese medicine, has demonstrated promising anticancer activity against multiple malignancies, particularly hepatocellular carcinoma (HCC). However, its epigenetic regulatory mechanisms and associated transcriptional consequences remain poorly characterized. Methods In this study, we integrated biochemical assays with a panel of cellular analyses assessing cell viability, proliferation, colony formation, and migratory capacity to investigate NCTD's therapeutic potential in HCC progression. Potential molecular targets of NCTD were systematically identified through integrated network pharmacology approaches. Chromatin immunoprecipitation quantitative PCR (ChIP-qPCR) was performed to quantify H3K27me3 enrichment level at the TOP2A locus in NCTD-treated HCC cells. Molecular docking simulations were employed to examine structural interactions between NCTD and EZH2 (enhancer of zeste homolog 2), while co-immunoprecipitation assays were further conducted to validate protein-protein interactions between EZH2 and protein phosphatase 1 (PP1). Results We identified topoisomerase IIα (TOP2A) as a critical molecular target mediating NCTD's anti-HCC effects. Functional characterization revealed that NCTD significantly attenuated HCC cell proliferation and induced G2/M phase cell cycle arrest through disruption of the TOP2A-p53 signaling axis. Mechanistic investigations demonstrated that NCTD epigenetically suppresses TOP2A transcription via PRC2 (Polycomb Repressive Complex 2)-mediated deposition of the repressive histone mark H3K27me3 at the TOP2A promoter. Structural biology analyses confirmed direct binding of NCTD to EZH2 protein, consequently impairing PP1-mediated dephosphorylation and enhancing PRC2 complex stability. Conclusion Our findings establish that NCTD exerts anticancer effects in HCC through epigenetic silencing of TOP2A. This work not only elucidates a novel pharmacoepigenetic mechanism underlying NCTD's antitumor activity but also provides translational rationale for developing PRC2-targeted therapeutic strategies in HCC management.
Collapse
Affiliation(s)
- Ruibing Wu
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Hengye Yuan
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Yuehua Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xianggang Gou
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wanhua Hou
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Zhongzheng Zhou
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xinran Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiuling Deng
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Changshan Wang
- College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, China
| | - Haisheng Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jia Yan
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Medical Experimental Center of Basic Medical School, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
23
|
Zhao X, Chen C, Feng X, Lei H, Qi L, Zhang H, Xu H, Wan J, Zhang Y, Yang B. Emd-D inhibited ovarian cancer progression via PFKFB4-dependent glycolysis and apoptosis. Chin J Nat Med 2025; 23:431-442. [PMID: 40274346 DOI: 10.1016/s1875-5364(25)60843-0] [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: 12/23/2023] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 04/26/2025]
Abstract
Ovarian cancer poses a significant threat to women's health, necessitating effective therapeutic strategies. Emd-D, an emodin derivative, demonstrates enhanced pharmaceutical properties and bioavailability. In this study, Cell Counting Kit 8 (CCK8) assays and Ki-67 staining revealed dose-dependent inhibition of cell proliferation by Emd-D. Migration and invasion experiments confirmed its inhibitory effects on OVHM cells, while flow cytometry analysis demonstrated Emd-D-induced apoptosis. Mechanistic investigations elucidated that Emd-D functions as an inhibitor by directly binding to the glycolysis-related enzyme PFKFB4. This was corroborated by alterations in intracellular lactate and pyruvate levels, as well as glucose transporter 1 (GLUT1) and hexokinase 2 (HK2) expression. PFKFB4 overexpression experiments further supported the dependence of Emd-D on PFKFB4-mediated glycolysis and SRC3/mTORC1 pathway-associated apoptosis. In vivo experiments exhibited reduced xenograft tumor sizes upon Emd-D treatment, accompanied by suppressed glycolysis and increased expression of Bax/Bcl-2 apoptotic proteins within the tumors. In conclusion, our findings demonstrate Emd-D's potential as an anti-ovarian cancer agent through inhibition of the PFKFB4-dependent glycolysis pathway and induction of apoptosis. These results provide a foundation for further exploration of Emd-D as a promising drug candidate for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin 150081, China
| | - Chao Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xuefei Feng
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Haoqi Lei
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Lingling Qi
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hongxia Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Haiying Xu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Jufeng Wan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin 150081, China.
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| |
Collapse
|
24
|
Yang H, Bi Y, Wang M, Liu Y, Naseem A, Huang J. Xihuang pill suppresses triple-negative breast cancer via regulating AXL/TGF-β1/RhoA/ROCK and ERK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119556. [PMID: 40010552 DOI: 10.1016/j.jep.2025.119556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 01/26/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xihuang pill (XHP), a traditional prescription for treating "Ru yan (breast cancer)" for 270 years, has the efficacies of clearing heat and improving blood circulation. Previous research has demonstrated that XHP yields significant therapeutic effects in the management of triple-negative breast cancer (TNBC). AIM OF THE STUDY To explore and validate the usage of a traditional Chinese medicine formula XHP in combating TNBC by epithelial-mesenchymal transition (EMT) and apoptosis mechanisms. MATERIALS AND METHODS The cytotoxic effects of XHP were evaluated using the CCK-8 assay after incubating with 4T1 and MDA-MB-231 TNBC cell lines. WB assay was conducted to detect the expression of proteins associated with EMT and apoptosis signaling pathways, elucidating the mechanism by which XHP inhibits TNBC. DAPI staining, and mitochondrial membrane potential (MMP) assays (JC-1) were utilized to further confirm the effect of apoptosis in TNBC. Moreover, morphological examination, wound healing assays, and transwell experiments were used to investigate the EMT properties of XHP. Besides, this study constructed 4T1-xenograft model to assess the anti-tumor efficacy of XHP, while the underlying mechanism was revealed by H&E staining. RESULTS XHP suppressed tumor growth and metastasis in Balb/c transplanted tumor model by inducing EMT and apoptosis. XHP modulated the expression of the AXL/TGF-β1/RhoA/ROCK pathway, reducing tumor cell EMT, and influencing the ERK signaling pathway. Moreover, XHP inhibited the expression level of Bcl2, Bax and caspase 9 to promote apoptosis in tumor cells. These phenomena eventually manifested as inhibiting the proliferation and migration of TNBC cells. CONCLUSIONS XHP inhibits TNBC and regulates EMT and apoptosis phenotypes by regulating the AXL/TGF-β1/RhoA/ROCK and ERK pathways. Therefore, it could be further utilized in the discovery of anti-cancer drugs.
Collapse
Affiliation(s)
- Haolin Yang
- Beijing University of Chinese Medicine, Beijing, China
| | - Yu Bi
- Heilongjiang University of Chinese Medicine 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Min Wang
- Heilongjiang University of Chinese Medicine 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Yan Liu
- Heilongjiang University of Chinese Medicine 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Anam Naseem
- Heilongjiang University of Chinese Medicine 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Jinchang Huang
- Beijing University of Chinese Medicine Third Affiliated Hospital, No. 51 Anwai Xiaoguan Street, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
25
|
Zou Y, Chen J, Qu Y, Luo X, Wang W, Zheng X. Evolution of nMOFs in photodynamic therapy: from porphyrins to chlorins and bacteriochlorins for better efficacy. Front Pharmacol 2025; 16:1533040. [PMID: 40170725 PMCID: PMC11959078 DOI: 10.3389/fphar.2025.1533040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Photodynamic therapy (PDT) has gained significant attention due to its non-invasive nature, low cost, and ease of operation. Nanoscale metal-organic frameworks (nMOFs) incorporating porphyrins, chlorins, and bacteriochlorins have emerged as one of the most prominent photoactive materials for tumor PDT. These nMOFs could enhance the water solubility, stability and loading efficiency of photosensitizers (PSs). Their highly ordered porous structure facilitates O2 diffusion and enhances the generation of 1O2 from hydrophobic porphyrins, chlorins, and bacteriochlorins, thereby improving their efficacy of phototherapy. This review provides insights into the PDT effects of nMOFs derived from porphyrins, chlorins, and bacteriochlorins. It overviews the design strategies, types of reactive oxygen species (ROS), ROS generation efficiency, and the unique biological processes involved in inhibiting tumor cell proliferation, focusing on the mechanism by which molecular structure leads to enhanced photochemical properties. Finally, the review highlights the new possibilities offered by porphyrins, chlorins, and bacteriochlorins-based nMOFs for tumor PDT, emphasizing how optimized design can further improve the bioapplication of porphyrin derivatives represented PSs. With ongoing research and technological advancements, we anticipate that this review will garner increased attention from scientific researchers toward porphyrin-based nMOFs, thereby elevating their potential as a prominent approach in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Yutao Zou
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu, China
| | - Jiayi Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yijie Qu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xuanxuan Luo
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaohua Zheng
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
26
|
Fu Z, Wang S, Zhou X, Ouyang L, Chen Z, Deng G. Harnessing the Power of Traditional Chinese Medicine in Cancer Treatment: The Role of Nanocarriers. Int J Nanomedicine 2025; 20:3147-3174. [PMID: 40103746 PMCID: PMC11913986 DOI: 10.2147/ijn.s502104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
For centuries, traditional Chinese medicine (TCM) has had certain advantages in the treatment of tumors. However, due to their poor water solubility, low bioavailability and potential toxicity, their effective delivery to target sites can be a major challenge. Nanocarriers based on the active ingredients of TCM, such as liposomes, polymer nanoparticles, inorganic nanoparticles, and organic/inorganic nanohybrids, are a promising strategy to improve the delivery of TCM, resulting in higher therapeutic outcomes and fewer side effects. Therefore, this article intends to review the application of Chinese medicine nano preparation in tumor. Firstly, we introduce the classification and synthesis of nanometer preparations of Chinese medicine. The second part mainly introduces the different responses of TCM nano-preparations in the course of treatment to introduce how TCM nano-preparations play a role in anti-tumor therapy. The third part focuses on Different response modes of Chinese medicine nano preparations in tumor therapy. The fourth part elucidates the application of Chinese medicine nano preparations in the treatment of cancer. Finally, the research direction to be explored in related fields is put forward.
Collapse
Affiliation(s)
- Ziyu Fu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Shengmei Wang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Xin Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Linqi Ouyang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Zhen Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
| | - Guiming Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China
- The second Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410005, People’s Republic of China
| |
Collapse
|
27
|
Wang J, Fang Q. Peimine inhibits cell proliferation and migration of breast cancer via regulating the O-GlcNAcylation of USP41. Int Immunopharmacol 2025; 149:114108. [PMID: 39923575 DOI: 10.1016/j.intimp.2025.114108] [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: 09/23/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/11/2025]
Abstract
Peimine is a isosteroidal alkaloid with multiple biological activities and has gained widespread clinical applications. This study was designed to investigate the effects of peimine (PM) on breast cancer (BC) and the underlying mechanism. Cell counting kit-8, EdU and transwell migration assays were performed to assess the cell viability, proliferation, and migration of MCF-7 and MDA-MB-231 cells. The interaction between USP41 and O-linked N-acetylglucosamine transferase (OGT) was evaluated by co-immunoprecipitation assay. A xenograft mouse model was established. Results showed that the cell viability of MCF-7 and MDA-MB-231 cells was decreased with the increasing concentration of PM, and the concentration of 20 μM was chosen for followed experiments. Besides, PM suppressed the proliferation and migration of MCF-7 and MDA-MB-231 cells. Moreover, PM treatment decreased the O-linked N-acetylglucosaminylation (O-GlcNAcylation) and OGT protein levels in MCF-7 and MDA-MB-231 cells. Mechanically, USP41 interacted with OGT in MDA-MB-231 cells. Overexpression of OGT enhanced the protein stability of USP41. Final rescue results demonstrated that overexpressing OGT or USP41 reversed the decreases of cell viability, proliferation, and migration in PM-treated MCF-7 and MDA-MB-231 cells; while OGT or USP41 knockout showed opposite results. Animal studies showed that PM treatment inhibited the tumor growth. In summary, PM inhibited cell viability, proliferation, and migration of BC by regulating the O-GlcNAcylation of USP41.
Collapse
Affiliation(s)
- Jinxian Wang
- Department of Preventable Diseases Treatment, Qiqihar City Hospital of Traditional Chinese Medicine, Qiqihar City, Heilongjiang Province 161005, China.
| | - Qiushi Fang
- The Qiqihar City Center for Disease Control and Prevention, Qiqihar City, Heilongjiang Province 161005, China
| |
Collapse
|
28
|
Tan Q, Liu H, Shi Q. Ailanthone Restrains Osteosarcoma Growth and Metastasis by Decreasing the Expression of Regulator of G Protein Signaling 4 and Twist Family BHLH Transcription Factor 1. Chem Biol Drug Des 2025; 105:e70075. [PMID: 40047202 DOI: 10.1111/cbdd.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025]
Abstract
Traditional Chinese medicine Ailanthone (AIL) has been confirmed to possess antimalarial, anti-inflammatory, and anticancer effects. Here, this study aimed to excavate the biological role and mechanism of AIL on osteosarcoma (OS) progression. Levels of Regulator of G protein signaling 4 (RGS4) and Twist Family BHLH Transcription Factor 1 (TWIST1) were detected by qRT-PCR and western blotting. In vitro and tumor formation experiments were conducted for functional analysis. The protein interaction between RGS4 and TWIST1 was verified by using a Co-immunoprecipitation assay. AIL impeded the proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) progression, but induced apoptosis in OS cells. RGS4 was highly expressed in OS tissues and cells and was decreased by AIL in cells. RGS4 silencing suppressed the growth and metastasis of OS cells, and RGS4 overexpression reversed the anticancer action of AIL in OS cells. Mechanistically, RGS4 interacted with TWIST1 and positively regulated its expression. TWIST1 was highly expressed in OS tissues and cells and could be reduced by AIL in cells. Moreover, TWIST1 overexpression abolished RGS4 silencing-triggered growth and metastasis inhibition in OS cells. Importantly, AIL impeded OS growth and metastasis in vivo by regulating RGS4 and TWIST1. Ailanthone restrained OS growth and metastasis by decreasing RGS4 and TWIST1 expression.
Collapse
Affiliation(s)
- Qiang Tan
- Department of Oncology, Affiliated Hospital of Xiangnan University, Chenzhou City, Hunan, China
| | - Hongzhan Liu
- Department of Orthopedics, Affiliated Hospital of Xiangnan University, Chenzhou City, Hunan, China
| | - Qiaojing Shi
- Department of Oncology, Affiliated Hospital of Xiangnan University, Chenzhou City, Hunan, China
| |
Collapse
|
29
|
Park MN, Choi J, Maharub Hossain Fahim M, Asevedo EA, Nurkolis F, Ribeiro RIMA, Kang HN, Kang S, Syahputra RA, Kim B. Phytochemical synergies in BK002: advanced molecular docking insights for targeted prostate cancer therapy. Front Pharmacol 2025; 16:1504618. [PMID: 40034825 PMCID: PMC11872924 DOI: 10.3389/fphar.2025.1504618] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Achyranthes japonica (Miq.) Nakai (AJN) and Melandrium firmum (Siebold and Zucc.) Rohrb. (MFR) are medicinal plants recognized for their bioactive phytochemicals, including ecdysteroids, anthraquinones, and flavonoids. This study investigates the anticancer properties of key constituents of these plants, focusing on the BK002 formulation, a novel combination of AJN and MFR. Specifically, the research employs advanced molecular docking and in silico analyses to assess the interactions of bioactive compounds ecdysterone, inokosterone, and 20-hydroxyecdysone (20-HE) with key prostate cancer-related network proteins, including 5α-reductase, CYP17, DNMT1, Dicer, PD-1, and PD-L1. Molecular docking techniques were applied to evaluate the binding affinities contributions of the bioactive compounds in BK002 against prostate cancer-hub network targets. The primary focus was on enzymes like 5α-reductase and CYP17, which are central to androgen biosynthesis, as well as on cancer-related proteins such as DNA methyltransferase 1 (DNMT1), Dicer, programmed death-1 (PD-1), and programmed death ligand-1 (PD-L1). Based on data from prostate cancer patients, key target networks were identified, followed by in silico analysis of the primary bioactive components of BK002.In silico assessments were conducted to evaluate the safety profiles of these compounds, providing insights into their therapeutic potential. The docking studies revealed that ecdysterone, inokosterone, and 20-hydroxyecdysonec demonstrated strong binding affinities to the critical prostate cancer-related enzymes 5α-reductase and CYP17, contributing to a potential reduction in androgenic activity. These compounds also exhibited significant inhibitory interactions with DNMT1, Dicer, PD-1, and PD-L1, suggesting a capacity to interfere with key oncogenic and immune evasion pathways. Ecdysterone, inokosterone, and 20-hydroxyecdysone have demonstrated the ability to target key oncogenic pathways, and their favorable binding affinity profiles further underscore their potential as novel therapeutic agents for prostate cancer. These findings provide a strong rationale for further preclinical and clinical investigations, supporting the integration of BK002 into therapeutic regimens aimed at modulating tumor progression and immune responses.
Collapse
Affiliation(s)
- Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinwon Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | - Estéfani Alves Asevedo
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Experimental Pathology Laboratory, Midwest Campus, Federal University of São João del-Rei, Divinópolis, Brazil
| | - Fahrul Nurkolis
- Department of Biological Sciences, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
| | | | - Han Na Kang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Rony Abdi Syahputra
- Department of Biological Sciences, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Li S, Zhang L, Zhang W, Chen H, Hong M, Xia J, Zhang W, Luan X, Zheng G, Lu D. Identifying traditional Chinese medicine combinations for breast cancer treatment based on transcriptional regulation and chemical structure. Chin Med 2025; 20:23. [PMID: 39953557 PMCID: PMC11829537 DOI: 10.1186/s13020-025-01074-5] [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: 11/14/2024] [Accepted: 01/24/2025] [Indexed: 02/17/2025] Open
Abstract
Breast cancer (BC) is a prevalent form of cancer among women. Despite the emergence of numerous therapies over the past few decades, few have achieved the ideal therapeutic effect due to the heterogeneity of BC. Drug combination therapy is seen as a promising approach to cancer treatment. Traditional Chinese medicine (TCM), known for its multicomponent nature, has been validated for its anticancer properties, likely due to the synergy effect of the key components. However, identifying effective component combinations from TCM is challenging due to the vast combination possibilities and limited prior knowledge. This study aims to present a strategy for discovering synergistic compounds based on transcriptional regulation and chemical structure. First, BC-related gene sets were used to screen TCM-derived compound combinations guided by synergistic regulation. Then, machine learning models incorporating chemical structural features were established to identify potential compound combinations. Subsequently, the pair of honokiol and neochlorogenic acid was selected by integrating the results of compound combination screening. Finally, cell experiments were conducted to confirm the synergistic effect of the pair against BC. Overall, this study offers an integrated screening strategy to discover compound combinations of TCM against BC. The tumor cell suppression effect of the honokiol and neochlorogenic acid pair validated the effectiveness of the proposed strategy.
Collapse
Affiliation(s)
- Shensuo Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Lijun Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wen Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongyu Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mei Hong
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jianhua Xia
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| | - Xin Luan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Guangyong Zheng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Dong Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
31
|
Xiao C, Zhou J, Xiong W, Ye X. The coexistence characteristics of microplastics and heavy metals in rhizomes of traditional Chinese medicine in mulch planting area. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2025; 47:74. [PMID: 39937393 DOI: 10.1007/s10653-025-02393-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
Rhizomatous traditional Chinese medicines (RTCMs) are widely crushed into powder and swallowed directly as medicine and food or health products to treat various diseases; however, they may contain toxic microplastics (MPs) and heavy metals. Currently, there are no reports on the detection of MPs and MP-heavy metal synergies in RTCMs. In this study, we selected eight representative RTCMs to investigate the abundance, types, sizes, and polymers of MP and heavy metals and to assess the level of contamination of MPs and synergies between MPs and heavy metals in RTCMs. The abundance of MPs in different RTCM ranged from 20.83 to 43.65 items/g. The dominant type was fragment (95.43%), and the dominant particle size was < 0.5 mm (73.72%) in MPs. Polyurethane (PU) (29.21%) and acrylics (ACR 13.53%) were the dominant polymers of MP. MP polymers showed obvious correlations with type and particle size: PU was enriched in 0-50-mm and 100-300-mm fragments, whereas ethylene vinyl acetate and ACR were enriched in 0-30-mm fibers. The heavy metals arsenic (As), lead (Pb), and chromium (Cr) were found to be more susceptible to synergistic contamination with MPs in RTCMs compared to other heavy metals. The estimated daily intake (EDI) of the MPs and heavy metals for RG (Rehmannia glutinosa) and RAY (Rhizoma atractylodis) were higher than others. The results showed that MP pollution is common in RTCMs and carries the potential risk of heavy metal or MP poisoning in humans who consume RTCMs.
Collapse
Affiliation(s)
- Cong Xiao
- School of Civil Engineering, Architecture and Enivironment, Hubei University of Technology, Wuhan, 430068, China.
- Hubei Key Laboratory of Environmental Soil and Ecological Restoration for River-Lakes, Wuhan, 430068, China.
| | - Jiabin Zhou
- School of Civil Engineering, Architecture and Enivironment, Hubei University of Technology, Wuhan, 430068, China
| | - Wen Xiong
- School of Civil Engineering, Architecture and Enivironment, Hubei University of Technology, Wuhan, 430068, China
- Hubei Key Laboratory of Environmental Soil and Ecological Restoration for River-Lakes, Wuhan, 430068, China
| | - Xiaochuan Ye
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| |
Collapse
|
32
|
Xing F, Lv H, Xiang W, Wang L, Zong Q, Lv G, Liu C, Feng Q, Wang H, Yang W. Traditional medicine Bazi Bushen potentiates immunosurveillance of senescent liver cancer cells via cGAS-STING signaling activation in macrophages. Cancer Lett 2025; 627:217544. [PMID: 39929434 DOI: 10.1016/j.canlet.2025.217544] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 05/19/2025]
Abstract
Senescent cancer cells often evade immune clearance to exert profound effects on cancer progression and therapy resistance. Improving immunosurveillance to eliminate senescent cancer cells is a crucial measure to enhance anti-cancer therapy. Bazi Bushen (BZBS) is a traditional medicine with the function of relieving fatigue and delaying ageing, but its role in tumor treatment remains poorly understood. Herein, we find that BZBS promotes immunosurveillance of both chemotherapy- and oncogene-induced senescent liver cancer cells, further leading to enhanced chemotherapy efficacy and dramatic tumor repression in mice. Mechanistically, BZBS induces mitochondrial DNA leakage by mitochondrial damage to further activate cGAS-STING signaling in macrophages. Subsequently, cGAS-STING signaling activation in macrophages recruits CD8+ T cells into tumor and promotes the anti-tumor activity of CD8+ T cells to eradicate senescent cancer cells. Furthermore, host STING is responsible for BZBS-mediated immunosurveillance of senescent liver cancer cells in mice. Therefore, our findings unveil the role of traditional medicine BZBS in activating cGAS-STING signaling and potentiating senescence immunosurveillance to enhance anti-cancer therapy.
Collapse
Affiliation(s)
- Fuxue Xing
- Department of Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Hongwei Lv
- Department of Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China; National Center for Liver Cancer, Naval Medical University, Shanghai, 201805, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University, Shanghai, 200438, China; Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai, 200438, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China
| | - Wei Xiang
- Department of Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Liang Wang
- National Center for Liver Cancer, Naval Medical University, Shanghai, 201805, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qianni Zong
- National Center for Liver Cancer, Naval Medical University, Shanghai, 201805, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University, Shanghai, 200438, China
| | - Guishuai Lv
- National Center for Liver Cancer, Naval Medical University, Shanghai, 201805, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University, Shanghai, 200438, China
| | - Chunying Liu
- National Center for Liver Cancer, Naval Medical University, Shanghai, 201805, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qiyu Feng
- Department of Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China.
| | - Hongyang Wang
- Department of Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China; National Center for Liver Cancer, Naval Medical University, Shanghai, 201805, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University, Shanghai, 200438, China; Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai, 200438, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| | - Wen Yang
- Department of Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China; National Center for Liver Cancer, Naval Medical University, Shanghai, 201805, China; International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute/Hospital, Naval Medical University, Shanghai, 200438, China; Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Shanghai, 200438, China; Key Laboratory of Signaling Regulation and Targeting Therapy of Liver Cancer, Ministry of Education, Shanghai, 200438, China.
| |
Collapse
|
33
|
Jin Z, Yun L, Cheng P. Tanshinone I reprograms glycolysis metabolism to regulate histone H3 lysine 18 lactylation (H3K18la) and inhibits cancer cell growth in ovarian cancer. Int J Biol Macromol 2025; 291:139072. [PMID: 39710022 DOI: 10.1016/j.ijbiomac.2024.139072] [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: 10/22/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Salvia miltiorrhiza, the anticancer properties of these components are multifaceted, encompassing the inhibition of tumor growth, prevention of the metastatic spread of cancer cells, enhancement of the sensitivity of cancer cells to chemotherapy and radiation therapy, and the suppression of angiogenesis, which is crucial for tumor growth and survival. In the context of our recent study, we have discovered that tanshinone I, one of the active components of Salvia miltiorrhiza, possesses the ability to inhibit the proliferation of ovarian cancer cells, both in laboratory settings and within living organisms. To further understand the molecular mechanisms behind this effect, we conducted a comprehensive transcriptomic analysis. Our findings indicated that tanshinone I exerts its inhibitory action by downregulating the expression of genes associated with glycolysis. Specifically, tanshinone I decreased the expression of glycolysis-related genes such as HK2 (hexokinase 2), PFK (phosphofructokinase), ENO2 (enolase 2), and LDHA (lactate dehydrogenase A). Inhibiting lactate production by tanshinone I application reduced the level of histone H3 lysine 18 lactylation (H3K18la), which reduced the expression of tumor-associated genes, such as TTK, PDGFRβ, YTHDF2 and RUBCNL. In addition, tanshinone I alleviated the immunosuppressive tumor microenvironment. In summary, tanshinone I blocks glycolysis to regulate histone H3 lysine 18 lactylation (H3K18la), which inhibits ovarian cancer cell growth, revealing the anticancer mechanism of tanshinone I.
Collapse
Affiliation(s)
- Zhou Jin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Yun
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Peng Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
34
|
Jia WD, Bai X, Ma QQ, Bian M, Bai CM, Li D, Li LF, Wei CX, Yu LJ. Synthesis, molecular docking studies of formononetin derivatives as potent Bax agonists for anticancer activity. Nat Prod Res 2025; 39:423-437. [PMID: 37921074 DOI: 10.1080/14786419.2023.2269592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/21/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023]
Abstract
Formononetin as a Bax agonist exhibits anticancer effects. To identify novel Bax agonist, 18 new structurally modified formononetin derivatives were synthesised and their anticancer activities were evaluated in the A549 and Beas-2b cell lines. The results indicated that 7a elicited the most potent inhibitory effect against the A549 cell line, with an IC50 value of 0.87 μM, and no obvious toxicity to Beas-2b cells. These results indicated that 7a was 40-fold and 6.94-fold more efficacious than Formononetin and Doxorubicin, respectively. Additionally, western blot and immunofluorescence assays demonstrated that 7a downregulated the protein expression of Bcl-2 and upregulated the expressions of Bax to promote A549 apoptosis, the obtained results also suggested that 7a had the potential to be developed into a lead compound that can be applied in the prevention and treatment of lung cancer.
Collapse
Affiliation(s)
- Wei-Dong Jia
- Inner Mongolia Minzu University, Tongliao, P.R. China
| | - Xue Bai
- Inner Mongolia Minzu University, Tongliao, P.R. China
- College of Public Health, Inner Mongolia Minzu University, Tongliao, China
| | - Qian-Qian Ma
- Inner Mongolia Minzu University, Tongliao, P.R. China
- College of Public Health, Inner Mongolia Minzu University, Tongliao, China
| | - Ming Bian
- Inner Mongolia Minzu University, Tongliao, P.R. China
- College of Public Health, Inner Mongolia Minzu University, Tongliao, China
| | - Chun-Mei Bai
- Inner Mongolia Minzu University, Tongliao, P.R. China
| | - Di Li
- Inner Mongolia Minzu University, Tongliao, P.R. China
| | - Li-Fei Li
- First Clinical Medical of Inner, Mongolia Minzu University, Tongliao, P.R. China
| | - Cheng-Xi Wei
- Inner Mongolia Minzu University, Tongliao, P.R. China
| | - Li-Jun Yu
- Inner Mongolia Minzu University, Tongliao, P.R. China
- College of Public Health, Inner Mongolia Minzu University, Tongliao, China
| |
Collapse
|
35
|
Song P, Shen N, Wu Z, He S. Baicalein Inhibits Tumor Property of Hepatocellular Carcinoma Cells Through the Inactivation of the E2F Transcription Factor 1/Mediator Complex Subunit 7 Axis. Chem Biol Drug Des 2025; 105:e70063. [PMID: 39935236 DOI: 10.1111/cbdd.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025]
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy with poor prognosis. Baicalein, a natural compound, can regulate multiple cellular processes in various cancer types. In this study, we investigated the role of baicalein in regulating HCC and explored its potential mechanism. The expression of mediator complex subunit 7 (MED7) and E2F transcription factor 1 (E2F1) was analyzed by quantitative real-time polymerase chain reaction or Western blotting assay. Cell proliferation was assessed by cell colony formation assay and 5-ethynyl-2'-deoxyuridine assay. Cell migration was analyzed by transwell assay and wound-healing assay. Cell invasion was analyzed by transwell assay. Angiogenic ability of HCC cells was assessed by tube formation assay. Dual-luciferase reporter assay and chromatin immunoprecipitation assay were performed to validate the association between E2F1 and MED7. The xenograft mouse model assay was conducted to determine the effects of baicalein and E2F1 overexpression on tumor formation. Immunohistochemistry assay was used to determine positive expression rates of proteins. Upregulation of MED7 and E2F1 expression was observed in both HCC tissues and cells. Knockdown of MED7 suppressed HCC cell proliferation, migration, invasion, and tube formation. Transcriptional activation of MED7 by E2F1 was demonstrated in HCC cells. Overexpression of MED7 mitigated the effects induced by E2F1 depletion in HCC cells. Additionally, baicalein treatment effectively inhibited the tumor property of HCC cells by decreasing E2F1 expression in both in vitro and in vivo models. Baicalein inhibited the tumor property of HCC cells through the inactivation of the E2F1/MED7 axis, highlighting its potential clinical application in the treatment of HCC.
Collapse
Affiliation(s)
- Pinghui Song
- Department of General Surgery, Shaanxi 215 Hospital of Nuclear Industry, Xianyang City, Shaanxi, China
| | - Naiying Shen
- Department of General Surgery, Shaanxi 215 Hospital of Nuclear Industry, Xianyang City, Shaanxi, China
| | - Zhongkun Wu
- Department of General Surgery, Shaanxi 215 Hospital of Nuclear Industry, Xianyang City, Shaanxi, China
| | - Sha He
- Department of Interventional, Shaanxi 215 Hospital of Nuclear Industry, Xianyang City, Shaanxi, China
| |
Collapse
|
36
|
Han Y, Sun Z. Anticancer potential of osthole: targeting gynecological tumors and breast cancer. Pharmacol Rep 2025; 77:87-102. [PMID: 39617816 DOI: 10.1007/s43440-024-00685-3] [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: 10/14/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/21/2025]
Abstract
Gynecological tumors, such as ovarian, endometrial, and cervical cancers, alongside breast cancer, represent significant malignancies that pose serious threats to women's health worldwide. Standard treatments, including surgery, chemotherapy, radiotherapy, and targeted therapies, are commonly utilized in clinical practice. However, challenges such as high recurrence rates, drug resistance, and adverse side effects underscore the urgent need for more effective therapeutic options. Osthole, a natural coumarin compound derived from Chinese herbal medicine, has demonstrated remarkable antitumor activity against various cancers. Emerging evidence indicates that osthole can inhibit the proliferation, invasion, and metastasis of gynecological and breast cancer cells through various mechanisms, including inducing apoptosis and autophagy, regulating the tumor microenvironment, inhibiting tumor angiogenesis, and enhancing the sensitivity of cancer cells to chemotherapy and radiotherapy. This review highlights the recent advancements in osthole research within the context of gynecological and breast cancers, focusing on its molecular mechanisms, and offers a theoretical foundation for its potential development as an anticancer agent.
Collapse
Affiliation(s)
- Yingqi Han
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Lixia District, Jinan, Shangdong Province, 250014, China
| | - Zhengao Sun
- Department of Reproduction and Genetics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 42 Wenhua West Road, Lixia District, Jinan, Shangdong Province, 250014, China.
| |
Collapse
|
37
|
Wu Z, Song B, Peng F, Zhang Q, Wu S. Zangsiwei prevents particulate matter-induced lung inflammation and fibrosis by inhibiting the TGF-β/SMAD pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118752. [PMID: 39232997 DOI: 10.1016/j.jep.2024.118752] [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: 06/27/2024] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zangsiwei(ZSW) is a traditional Tibetan medicine from China consisting of extracts of Rhododendron anthopogonoides Maxim, Gentiana Tourn, Corydalis hendersonii Hemsl and Berberis kansuensis C.K.Schneid. Traditionally, ZSW has been used by Tibetan physicians to treat chronic respiratory diseases. The role of ZSW in particulate matter-induced lung inflammation and fibrosis remains unclear. AIM OF THE STUDY Combining non-targeted metabolomics, network pharmacology, and molecular docking to explore the mechanism of ZSW in the treatment of particulate matter-induced lung inflammation and fibrosis, and validated by in vivo and in vitro experiments. MATERIALS AND METHODS The serum metabolite profile post-ZSW administration was first identified utilizing non-targeted metabolomics. Network pharmacology and molecular docking were employed to predict potential bioactive components and their corresponding targets. The in silico predictions were subsequently validated through in vivo studies in mice exposed to PM2.5 and silica dust, as well as in vitro studies utilizing human lung epithelial cells (A549) and lung fibroblasts (MRC5). RESULTS Metabolomic analysis identified specific serum metabolites that were associated with ZSW treatment. Network pharmacology and molecular docking identified key targets involved in the Transforming growth factor-β (TGF-β)/SMAD pathway, which were subsequently validated through in vivo experiments demonstrating a reduction in lung inflammation and fibrosis in ZSW-treated mice. In vitro studies demonstrated that ZSW exerts protective effects against PM2.5-induced cytotoxicity and modulates fibrotic markers in a dose-dependent manner. This is consistent with the inhibition of the TGF-β/SMAD pathway. CONCLUSION Our integrated approach, which combines non-targeted metabolomics, network pharmacology, and molecular docking, followed by rigorous in vivo and in vitro validation, establishes ZSW as a potential therapeutic agent for particulate matter-induced lung inflammation and fibrosis.
Collapse
Affiliation(s)
- Zhijian Wu
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Boyang Song
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Fei Peng
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Quan Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China
| | - Shangjie Wu
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Centre for Evidence-based Medicine, Changsha, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease in Hunan Province, Changsha, Hunan, 410011, China.
| |
Collapse
|
38
|
Lei Z, Luan F, Zou J, Zhang X, Zhai B, Xin B, Sun J, Guo D, Wang J, Shi Y. Traditional uses, phytochemical constituents, pharmacological properties, and quality control of Pseudostellaria heterophylla (Miq.) Pax. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118871. [PMID: 39368760 DOI: 10.1016/j.jep.2024.118871] [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: 04/24/2024] [Revised: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
ETHNOPHARMACOLOGIC RELEVANCE Pseudostellaria heterophylla (Miq.) Pax belongs to the Caryophyllaceae family, which is widely used in traditional Chinese medicine in Asia. P. heterophylla was first documented in the classical text Bencao Congxin, also known as "Haier Shen". As a renowned folk medicine with a long history of medicinal application in China, this plant is frequently employed to address spleen deficiency and fatigue, loss of appetite, and weakness after illness. In recent years, P. heterophylla has gained significant global attention as an important medicinal plant, attributable to its pharmacological activities on the immune and endocrine systems, as well as its diverse applications. AIM OF THE WORK This review aims to deliver a comprehensive and analytical overview of the ethnopharmacology, phytochemistry, pharmacology, pharmacokinetics, toxicology, and quality control of P. heterophylla, while also offering novel insights and opportunities for future research. MATERIALS AND METHODS Relevant information regarding P. heterophylla was gathered from various databases, including Web of Science, PubMed, ACS Publications, Google Scholar, Baidu Scholar, and CNKI, in addition to The Catalogue of Life, the Flora of China database, and The World Flora Online. All published articles in multiple languages have been included and properly cited. The chemical structure of the compound was illustrated utilizing ChemDraw 19.0 software. RESULTS P. heterophylla has been traditionally employed to address a range of ailments, including cancer, cardiovascular diseases, diabetes, and respiratory disorders. More than 289 active constituents have been identified in P. heterophylla, comprising cyclic peptides, polysaccharides, saponins, alkaloids, flavonoids, nucleosides, and amino acids. Pharmacological investigations have demonstrated that P. heterophylla and its active constituents exhibit a broad spectrum of biological activities, including anti-cancer, immunomodulatory, antioxidant, hypoglycemic, anti-inflammatory effects, modulation of intestinal flora, enhancement of cognitive function, and inhibition of tyrosine kinase activity. Furthermore, it is extensively utilized in the functional food and cosmetics industries. CONCLUSION As a dual-purpose resource for both food and medicine, P. heterophylla possesses significant health care functions and considerable edible and medicinal value, with promising prospects for future development and utilization. However, numerous investigations into the biological activities of P. heterophylla are primarily focused on its extracts and bioactive constituents, and the mechanisms underlying the actions of these extracts and components remain unclear, with a dearth of studies on clinical efficacy and safety. Consequently, further detailed in vitro and in vivo studies investigating the mechanisms of action of pure active compounds of P. heterophylla are warranted, along with additional clinical investigations to ascertain the safety and efficacy of the plant for human use.
Collapse
Affiliation(s)
- Ziwen Lei
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Fei Luan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Junbo Zou
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Xiaofei Zhang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Bingtao Zhai
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Bao Xin
- School of Public Health, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Jing Sun
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Dongyan Guo
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China
| | - Jingyuan Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China.
| | - Yajun Shi
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, PR China.
| |
Collapse
|
39
|
Liu M, Zheng L, Zhang Y, Tian J. Mechanistic insights into pachymic acid's action on triple-negative breast Cancer through TOP2A targeting. Sci Rep 2025; 15:2856. [PMID: 39843552 PMCID: PMC11754797 DOI: 10.1038/s41598-025-87286-z] [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: 10/11/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen and progesterone receptors, and lack of human epidermal growth factor receptor 2 (HER2) expression. Traditional Chinese medicine (TCM) has demonstrated promising efficacy in treating TNBC. This study explored the mechanisms of pachymic acid (PA) on TNBC by merging network pharmacology with experimental validation. We acquired Microarray data of TNBC from the Gene Expression Omnibus (GEO). The related targets of PA were predicted and screened using the following 6 databases: Swiss Target Prediction, HERB (Herbal Medicine Database), ETCM (Encyclopedia of Traditional Chinese Medicine), BATMAN (Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicine), HIT (Herb Ingredients' Targets Database), and PharmMapper. The STRING interaction network analysis tool was used to create Protein-Protein Interaction (PPI) networks. Enrichment analysis included Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We conducted a pan-cancer analysis, tumor immune microenvironment analysis, and molecular docking. We performed cell experimental, included cytotoxicity assay, apoptosis analysis, proliferation assay, and migration and invasion assays. PA has potential for treating TNBC with the target of TOP2A, and platinum drug resistance possibly serving as the KEGG pathway through which PA exerts its therapeutic effects. PA is involved in processes such as nuclear division, chromosome segregation, mitotic nuclear division, condensed chromosome formation, and protein C-terminus binding. PA probably exert its therapeutic effects through the tumor immune microenvironment, involving elements such as Dendritic cells activated, Eosinophils, Macrophages M0, Macrophages M1, and T cells CD4 memory activated. The therapeutic effects of PA may vary across different subtypes of TNBC such as TNBC-BL1, TNBC-Metaplastic, and TNBC-BL2. This study provides compelling evidence that PA holds significant promise as a therapeutic agent for TNBC, primarily through its action on TOP2A and its influence on the TNBC.
Collapse
Affiliation(s)
- Ming Liu
- Evidence-based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou City, No.199 Donggang West Road, 730000, Gansu Province, China
| | - Li Zheng
- Department of Pharmacy, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Yang Zhang
- Department of Traditional Chinese medicine, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Jinhui Tian
- Evidence-based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou City, No.199 Donggang West Road, 730000, Gansu Province, China.
- Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou City, Gansu Province, China.
| |
Collapse
|
40
|
Zhou C, Zhong R, Zhang L, Yang R, Luo Y, Lei H, Li L, Cao J, Yuan Z, Tan X, Xie M, Qu H, He Z. Exploring the mechanism of rosmarinic acid in the treatment of lung adenocarcinoma based on bioinformatics methods and experimental validation. Discov Oncol 2025; 16:47. [PMID: 39812944 PMCID: PMC11735722 DOI: 10.1007/s12672-025-01784-0] [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: 08/10/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Rosmarinic acid (RosA) is a natural polyphenol compound that has been shown to be effective in the treatment of inflammatory disease and a variety of malignant tumors. However, its specific mechanism for the treatment of lung adenocarcinoma (LUAD) has not been fully elucidated. Therefore, this study aims to clarify the mechanism of RosA in the treatment of LUAD by integrating bioinformatics, network pharmacology and in vivo experiments, and to explore the potential of the active ingredients of traditional Chinese medicine in treating LUAD. METHODS Firstly, the network pharmacology was used to screen the RosA targets, and LUAD-related differential expressed genes (DEGs) were acquired from the GEO database. The intersection of LUAD regulated by RosA (RDEGs) was obtained through the Venn diagram. Secondly, GO and KEGG enrichment analysis of RDEGs were performed, and protein-protein interaction networks (PPIs) were constructed to identify and visualize hub RDEGs. Then, molecular docking between hub RDEGs and RosA was performed, and further evaluation was carried out by using bioinformatics for the predictive value of the hub RDEGs. Finally, the mechanism of RosA in the treatment of LUAD was verified by establishing a xenograft model of NSCLC in nude mouse. RESULTS Bioinformatics and other analysis showed that, compared with the control group, the expressions of MMP-1, MMP-9, IGFBP3 and PLAU in LUAD tissues were significantly up-regulated, and the expressions of PPARG and FABP4 were significantly down-regulated, and these hub RDEGs had potential predictive value for LUAD. In vivo experimental results showed that RosA could inhibit the growth of transplanted tumors in nude mice bearing tumors of lung cancer cells, reduce the positive expression of Ki67 in lung tumor tissue, and hinder the proliferation of lung tumor cells. Upregulated expression of PPARG and FABP4 by activating the PPAR signaling pathway increases the level of ROS in lung tumor tissues and promotes apoptosis of lung tumor cells. In addition, RosA can also reduce the expression of MMP-9 and IGFBP3, inhibit the migration and invasion of lung tumor tissue cells. CONCLUSIONS This study demonstrated that RosA could induce apoptosis by regulating the PPAR signaling pathway and the expression of MMP-9, inhibit the proliferation, migration and invasion of lung cancer cells, thereby exerting anti-LUAD effects. This study provides new insight into the potential mechanism of RosA in treating LUAD and provides a new therapeutic avenue for treatment of LUAD.
Collapse
Affiliation(s)
- Chaowang Zhou
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Ruqian Zhong
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Lei Zhang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Renyi Yang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Yuxin Luo
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Huijun Lei
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Liang Li
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Jianzhong Cao
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Zhiying Yuan
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Xiaoning Tan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China
| | - Mengzhou Xie
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Haoyu Qu
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Zuomei He
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China.
| |
Collapse
|
41
|
Ji Y, Li L, Li W, Li L, Ma Y, Li Q, Chen X, Zhao W, Zhu H, Huo J, Wu M. Xiaoai Jiedu recipe reduces cell survival and induces apoptosis in hepatocellular carcinoma by stimulating autophagy via the AKT/mTOR pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119135. [PMID: 39586558 DOI: 10.1016/j.jep.2024.119135] [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: 09/28/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Xiaoai Jiedu recipe (XJR) is a traditional Chinese medicine formulation used in clinical settings to treat liver cancer. It has shown promising effectiveness by combining herbal and animal-derived ingredients, offering a new approach to cancer treatment. However, its mechanism of action is poorly understood. AIM OF THE STUDY The molecular processes underlying the inhibitory effects of the XJR on hepatocellular cancer (HCC) were investigated. MATERIALS AND METHODS The primary chemical components of XJR and associated disease targets relevant to HCC were anticipated and compiled using a database. The potential targets and processes by which XJR influenced HCC were investigated using GO and KEGG enrichment analyses, as well as protein-protein interaction (PPI) networks. Transmission electron microscopy, laser confocal microscopy, and Western blotting were used to evaluate autophagy, while CCK-8 assays measured cell viability and Western blotting and flow cytometry evaluated apoptosis. In vivo assays were conducted employing an HCC xenograft mouse model. RESULTS Network pharmacology analysis identified 456 intersecting targets between XJR and HCC. The top five active components are quercetin, cholesterol, jaceosidine, eupafolin, and oleanolic acid. The key targets include TP53, AKT1, IL6, EGFR, SRC, HSP90AA1, TNF, IL1B, MYC, and CASP3. Additionally, the autophagy pathway was found to be one of the main pathways through which XJR intervenes in HCC. The increased quantity of autophagosomes and autolysosomes, the overexpression of Beclin1 and LC3A/B-II proteins, and the downregulation of P62 all suggest that XJR stimulated autophagy in HCC cells. Functional tests employing pathway-specific activators and inhibitors and siRNA-based knockdown demonstrated that XJR promoted autophagy by blocking AKT/mTOR signaling. Furthermore, XJR reduced the viability of HCC cells and promoted apoptosis by upregulating apoptosis proteins. Autophagy inhibitors and Beclin1 silencing reversed these effects. Research conducted in vivo showed that XJR activated autophagy through the AKT/mTOR axis, thereby markedly reducing tumor growth and inducing tumor cell demise. CONCLUSIONS These studies show that XJR activates autophagy in both cellular and animal models to induce apoptosis and decrease HCC cell proliferation, as shown by network pharmacology and verification assays. Further, these findings provide experimental evidence that the anti-tumor activity of XJR involves autophagy stimulation.
Collapse
Affiliation(s)
- Yi Ji
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Li Li
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Wenting Li
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Liu Li
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Yanxia Ma
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qingfeng Li
- School of Acupuncture-Moxibustion and Tuina, School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xi Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Wenyue Zhao
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China
| | - Hengzhou Zhu
- Department of Oncology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China.
| | - Jiege Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| | - Mianhua Wu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210023, China.
| |
Collapse
|
42
|
Fang Y, Tang MX, Liu X. Traditional Chinese medicine trade among RCEP countries: structural characteristics and determinants. Front Public Health 2025; 12:1508839. [PMID: 39839391 PMCID: PMC11747611 DOI: 10.3389/fpubh.2024.1508839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Background With the increasing global focus on health and the growing popularity of natural therapies, Traditional Chinese Medicine (TCM) products, including extracts, crude drugs, and herbal preparations, are widely utilized as both primary and complementary medicines worldwide. The Regional Comprehensive Economic Partnership (RCEP), spanning 15 countries across East Asia, Southeast Asia, and Oceania, offers a vast market for TCM. However, limited research has been conducted on the complex trade relations among RCEP members. Methods The structural features and nodes attributes are analyzed using Social Network Analysis (SNA). Influencing factors are studied through the Quadratic Assignment Procedure (QAP) model. We also compiled a list of medicinal plants among RCEP member countries and the main TCM export products. Results The scale of TCM trade among RCEP members has fluctuated over time, with a temporary spike during the COVID-19 pandemic, followed by a gradual normalization. The trade network does not exhibit small-world properties, indicating a relatively balanced trade relationship. Due to its resource advantages, China occupies a central role acting as a dominant producer and leading exporter. Vietnam's export performance has been excellent in recent years, with the highest annual growth rate. Emerging markets, such as Myanmar, warrant closer attention. Economic size and population significantly positively affect trade value, while geographic distance and land adjacency have no significant impact. Trade activity is positively influenced by cultural and linguistic similarities, and countries with higher levels of economic freedom tend to engage in more trade. Tonifying TCM products with antioxidant and immune-boosting properties are more widely recognized in international markets. Conclusion RCEP has established an excellent trade platform for the export of TCM. The factors influencing TCM trade are predominantly long-term and structural, rather than being driven by the occurrence of any single, isolated event. Member states should strengthen collaboration in standardization, technology coordination, and knowledge sharing to establish a mutually beneficial trade ecosystem for TCM.
Collapse
Affiliation(s)
- Yue Fang
- School of Economics and Management, Anhui University of Chinese Medicine, Hefei, China
- Anhui Institute of Conservation and Development of Traditional Chinese Medicine Resources, Hefei, China
| | - Meng Xue Tang
- School of Economics and Management, Anhui University of Chinese Medicine, Hefei, China
| | - Xu Liu
- School of Economics and Management, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
43
|
Guo J, Wu Z, Chang X, Huang M, Wang Y, Liu R, Li J. Network Pharmacology Analysis and In Vitro Validation of the Active Ingredients and Potential Mechanisms of Gynostemma Pentaphyllum Against Esophageal Cancer. Comb Chem High Throughput Screen 2025; 28:500-513. [PMID: 38243957 DOI: 10.2174/0113862073280183240108113853] [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: 10/26/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Esophageal cancer (EC) is one of the deadliest malignancies worldwide. Gynostemma pentaphyllum Thunb. Makino (GpM) has been used in traditional Chinese medicine as a treatment for tumors and hyperlipidemia. Nevertheless, the active components and underlying mechanisms of anti-EC effects of GpM remain elusive. OBJECTIVE This study aims to determine the major active ingredients of GpM in the treatment of EC and to explore their molecular mechanisms by using network pharmacology, molecular docking, and in vitro experiments. METHODS Firstly, active ingredients and potential targets of GpM, as well as targets of EC, were screened in relevant databases to construct a compound-target network and a protein-protein interaction (PPI) network that narrowed down the pool of ingredients and targets. This was followed by gene ontology (GO) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Next, molecular docking, ADME and toxicity risk prediction, cell viability assays, in vitro scratch assays, Transwell cell invasion assays, and Western blotting analysis were subsequently applied to validate the results of the network analysis. RESULTS The screening produced a total of 21 active ingredients and 167 ingredient-related targets for GpM, along with 2653 targets for EC. The PPI network analysis highlighted three targets of interest, namely AKT1, TP53, and VEGFA, and the compound-target network identified three possible active ingredients: quercetin, rhamnazin, and isofucosterol. GO and EKGG indicated that the mechanism of action might be related to the PI3K/AKT signaling pathway as well as the regulation of cell motility and cell migration. Molecular docking and pharmacokinetic analyses suggest that quercetin and isoprostanoid sterols may have therapeutic value and safety for EC. The in vitro experiments confirmed that GpM can inhibit EC cell proliferation, migration, and invasion and suppress PI3K and AKT phosphorylation. CONCLUSION Our findings indicate that GpM exerts its anti-tumor effect on EC by inhibiting EC cell migration and invasion via downregulation of the PI3K/AKT signaling pathway. Hence, we have reason to believe that GpM could be a promising candidate for the treatment of EC.
Collapse
Affiliation(s)
- Jianxin Guo
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050011, China
| | - Zhongbing Wu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050011, China
| | - Xiaoyue Chang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050011, China
| | - Ming Huang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050011, China
| | - Yu Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050011, China
| | - Renping Liu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050011, China
| | - Jing Li
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050011, China
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, 05001l, China
| |
Collapse
|
44
|
Zhang Y, Zhang M, Song H, Dai Q, Liu C. Tumor Microenvironment-Responsive Polymer-Based RNA Delivery Systems for Cancer Treatment. SMALL METHODS 2025; 9:e2400278. [PMID: 38803312 DOI: 10.1002/smtd.202400278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/30/2024] [Indexed: 05/29/2024]
Abstract
Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment-responsive polymer-based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
Collapse
Affiliation(s)
- Yahan Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing, 102206, China
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Qiong Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
45
|
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: 5] [Impact Index Per Article: 5.0] [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
|
46
|
Zhang S, Guo L, Tao R, Liu S. Ferroptosis-targeting drugs in breast cancer. J Drug Target 2025; 33:42-59. [PMID: 39225187 DOI: 10.1080/1061186x.2024.2399181] [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: 05/19/2024] [Revised: 07/07/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
In 2020, breast cancer surpassed lung cancer as the most common cancer in the world for the first time. Due to the resistance of some breast cancer cell lines to apoptosis, the therapeutic effect of anti-breast cancer drugs is limited. According to recent report, the susceptibility of breast cancer cells to ferroptosis affects the progress, prognosis and drug resistance of breast cancer. For instance, roblitinib induces ferroptosis of trastuzumab-resistant human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells by diminishing fibroblast growth factor receptor 4 (FGFR4) expression, thereby augmenting the susceptibility of these cells to HER2-targeted therapies. In tamoxifen-resistant breast cancer cells, Fascin exacerbates their resistance by repressing solute carrier family 7 member 11 (SLC7A11) expression, which in turn heightens their responsiveness to tamoxifen. In recent years, Chinese herbs extracts and therapeutic drugs have been demonstrated to elicit ferroptosis in breast cancer cells by modulating a spectrum of regulatory factors pertinent to ferroptosis, including SLC7A11, glutathione peroxidase 4 (GPX4), acyl-CoA synthetase long chain family member 4 (ACSL4), and haem oxygenase 1 (HO-1). Here, we review the roles and mechanisms of Chinese herbal extracts and therapeutic drugs in regulating ferroptosis in breast cancer, providing potential therapeutic options for anti-breast cancer.
Collapse
Affiliation(s)
- Shuxian Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Lijuan Guo
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, China
| | - Shuangping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| |
Collapse
|
47
|
Wang Z, Zhang Y, Shen Y, Zhu C, Qin X, Gao Y. Liquidambaric acid inhibits cholangiocarcinoma progression by disrupting the STAMBPL1/NRF2 positive feedback loop. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156303. [PMID: 39706065 DOI: 10.1016/j.phymed.2024.156303] [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: 06/24/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Abnormal antioxidant capacity in cancer cells is intimately linked to tumor aggressiveness. Modulating oxidative stress status and inhibiting ferroptosis represents a novel anticancer therapeutic strategy. STAM Binding Protein Like 1 (STAMBPL1), a deubiquitinase, is implicated in various malignancies, yet its function in inhibiting ferroptosis and therapeutic potential for cholangiocarcinoma (CCA) remains unexplored. PURPOSE This study elucidates STAMBPL1's function in ferroptosis and evaluates liquidambaric acid (LDA) as its inhibitor for therapeutic applications. METHODS Using bioinformatics, WB, IHC, the expression and prognostic value of STAMBPL1 in CCA tissue was detected. The carcinogenic capacity of STAMBPL1 and LDA were assessed through CCK-8, EdU, cloning, transwell, scratch, apoptosis, and cell cycle assays. Flow cytometry and fluorescence microscopy, as well as transmission electron microscopy (TEM), examines the effects of STAMBPL1 and LDA on intracellular reactive oxygen species (ROS) and changes in mitochondrial membrane potential. The tumorigenic ability of STAMBPL1 and LDA in vivo was evaluated through subcutaneous tumor model and lung metastasis model. The underlying mechanism of STAMBPL1 was explored using immunoprecipitation coupled with Mass spectrometry (IP/MS), Co-immunoprecipitation (Co-IP), GST pull-down, DNA pull-down, and Dual-luciferase reporter assays. Molecular docking simulations, SPR, DARTS and CETSA predict the putative binding site of LDA on STAMBPL1 protein. Rescue experiments further confirmed the above conclusions. RESULTS This study unveils the upregulation and oncogenic role of STAMBPL1 in CCA. Functionally, STAMBPL1 notably enhances CCA cell proliferation and metastasis while impeding ferroptosis. STAMBPL1 stabilizes NRF2, a pivotal regulator of antioxidant enzymes, through K63 deubiquitination. Elevated NRF2, stabilized by STAMBPL1 overexpression, triggers GPX4 activation and reactive oxygen species (ROS) elimination. Particularly, sites 251-436 of STAMBPL1 interact with sites 228-605 of NRF2, facilitating DUB activity and eliminating ubiquitin molecules attached to NRF2, thus protecting it from proteasome-mediated degradation. Moreover, NRF2, acting as a transcription factor, binds to the promoter region of STAMBPL1 and activates its transcription, thus forming STAMBPL1/NRF2 positive feedback loop and regulating redox homeostasis. Molecular docking and in vitro/in vivo experiments identified that LDA binds to and inhibits STAMBPL1, thereby disrupting the STAMBPL1/NRF2 positive feedback loop, consequently suppressing CCA progression. CONCLUSION This study firstly reveals that STAMBPL1 promotes cholangiocarcinoma progression by upregulating NRF2, indicating that targeting the STAMBPL1/NRF2 axis is a novel therapeutic strategy. Additionally, our findings firstly suggest that LDA can bind to STAMBPL1, inhibiting NRF2 deubiquitination and offering significant therapeutic potential.
Collapse
Affiliation(s)
- Zhihuai Wang
- The Department of Hepato-biliary-pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China; Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Yinjie Zhang
- The Department of Hepato-biliary-pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China; Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Yuhang Shen
- The Department of Hepato-biliary-pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China; Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Chunfu Zhu
- The Department of Hepato-biliary-pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China; Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China
| | - Xihu Qin
- The Department of Hepato-biliary-pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China; Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China.
| | - Yuan Gao
- The Department of Hepato-biliary-pancreatic Surgery, The Institute of Hepatobiliary and Pancreatic Diseases, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, PR China; Changzhou Medical Center, Nanjing Medical University, Changzhou, PR China.
| |
Collapse
|
48
|
Wei J, Lei G, Chen Q, Huang W, Ning H, Yang M, Dong J, Hu L, Peng S, Gong H, Yuan M, Yuan P. Casticin inhibits proliferation of Non-small cell lung cancer cells through regulating reprogramming of glucose metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156278. [PMID: 39647464 DOI: 10.1016/j.phymed.2024.156278] [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: 07/11/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, with poor prognosis due to its rapid progression and resistance to existing therapies. Metabolic reprogramming, particularly alterations in glucose metabolism, is a key mechanism underlying tumor growth and progression, providing potential targets for novel therapeutic strategies. Casticin (CAS), a bioactive flavonoid, has shown anticancer effects in various cancers, but its specific role in NSCLC metabolism remains unclear. PURPOSE This study aims to investigate the effects of casticin on the proliferation and glucose metabolism of NSCLC cells, and to explore its underlying mechanisms. STUDY DESIGN AND METHODS We used both in vitro and in vivo models. (18)F-FDG PET/MR imaging was employed to assess the impact of casticin on glucose metabolism in A549 xenograft mice. NSCLC cell lines (A549 and H157) were treated with casticin to evaluate its effects on cell viability, glycolysis, oxidative phosphorylation, and fatty acid oxidation. Key metabolic enzyme expressions were analyzed using molecular detection techniques, and in vivo validation was performed using a subcutaneous xenograft mouse model. RESULTS Casticin significantly inhibited glucose metabolism and cell proliferation in a dose-dependent manner, while promoting oxidative phosphorylation without affecting lipid metabolism. The drug suppressed glycolysis by downregulating the expression of key glycolytic enzymes (GLUT1, HK2, GPI, ALDOA, ENO2, PKM2, and MCT4) through the regulation of HIF-1α. Overexpression of HIF-1α in both in vitro and in vivo models reversed the inhibitory effects of casticin, indicating that HIF-1α plays a central role in its mechanism of action. CONCLUSION Casticin inhibits NSCLC cell proliferation by suppressing glycolytic reprogramming via HIF-1α regulation. These findings highlight the potential of casticin as an anticancer therapeutic, particularly in targeting glucose metabolism in NSCLC.
Collapse
Affiliation(s)
- Jingyi Wei
- Department of nuclear medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, PR China; Shaanxi University of Chinese Medicine, Shaanxi Xianyang 710046, PR China
| | - Guangyan Lei
- Department of thoracic surgery, Tumor Hospital of Shaanxi Province, Xi'an, Shaanxi 710061, PR China
| | - Qiang Chen
- Cancer Center, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, PR China; MOE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, PR China
| | - Wen Huang
- Department of nuclear medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, PR China; Medical School of Yan'an University, Yan'an, Shaanxi 716000, PR China
| | - Hui Ning
- Shaanxi University of Chinese Medicine, Shaanxi Xianyang 710046, PR China
| | - Meng Yang
- Shaanxi University of Chinese Medicine, Shaanxi Xianyang 710046, PR China
| | - Jiaqi Dong
- Shaanxi University of Chinese Medicine, Shaanxi Xianyang 710046, PR China
| | - Longquan Hu
- Department of nuclear medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, PR China
| | - Shujia Peng
- Department of General Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, PR China
| | - Hui Gong
- Department of nuclear medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, PR China; Hospital of Unit 94162, Air Force of the CPLA, Xi'an, Shaanxi 710613, PR China
| | - Menghui Yuan
- Department of nuclear medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, PR China.
| | - Peng Yuan
- Department of nuclear medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, PR China.
| |
Collapse
|
49
|
Wang Y, Guan WX, Zhou Y, Zhang XY, Zhao HJ. Red ginseng polysaccharide promotes ferroptosis in gastric cancer cells by inhibiting PI3K/Akt pathway through down-regulation of AQP3. Cancer Biol Ther 2024; 25:2284849. [PMID: 38051132 PMCID: PMC10761076 DOI: 10.1080/15384047.2023.2284849] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/10/2023] [Indexed: 12/07/2023] Open
Abstract
OBJECTIVE This study aims to investigate the effect of red ginseng polysaccharide (RGP) on gastric cancer (GC) development and explore its mechanism. METHODS GC cell lines AGS were treated with varying concentrations of RGP (50, 100, and 200 μg/mL). AGS cells treated with 200 μg/mL RGP were transfected with aquaporin 3 (AQP3) overexpression vector. Cell proliferation, viability, and apoptosis were evaluated by MTT, colony formation assay, and flow cytometry, respectively. Real-time quantitative reverse transcription PCR (qRT-PCR) was used to detect the expression of AQP3. The levels of Fe2+, malondialdehyde, and lactate dehydrogenase were measured using their respective detection kits, and the reactive oxygen species levels was determined by probe 2',7'-dichlorodihydrofluorescein diacetate. The expression of ferroptosis-related protein and PI3K/Akt pathway-related protein were assessed by western blot. In vivo experiments in nude mice were performed and the mice were divided into four groups (n = 5/group) which gavage administrated with 150 mg/kg normal saline, and 75, 150, 300 mg/kg RGP, respectively. Their tumor weight and volume were recorded. RESULTS RGP treatment effectively inhibited the proliferation and viability of AGS cells in a dosage-dependent manner and induced apoptosis. It induced ferroptosis in AGS cells, as well as inhibiting the expression of PI3K/Akt-related proteins. AQP3 overexpression could reversed the effect of RGP treatment on ferroptosis. Confirmatory in vivo experiments showed that RGP could reduce the growth of implanted tumor, with increased RGP concentration resulting in greater tumor inhibitory effects. CONCLUSION RGP might have therapeutic potential against GC, effectively inhibiting the proliferation and viability of AGS cells.
Collapse
Affiliation(s)
- Yan Wang
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Gastrointestinal Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Wen-Xian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan Zhou
- Department of Gastrointestinal Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Xiao-Yu Zhang
- Department of Gastrointestinal Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Hai-Jian Zhao
- Department of Gastrointestinal Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
50
|
Cheng Y, Yu G, Du C, Chen Z, Liu X. Yanghe Decoction promotes ferroptosis through PPARγ-dependent autophagy to inhibit the malignant progression of triple-negative breast cancer. Prostaglandins Other Lipid Mediat 2024; 175:106909. [PMID: 39284544 DOI: 10.1016/j.prostaglandins.2024.106909] [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: 06/23/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous subtype of breast cancer that displays highly aggressive with poor prognosis. Yanghe Decoction (YHD) has been used in the treatment of breast cancer for many years. We aimed to explore the effects of YHD on the malignant phenotypes of MDA-MB-231 cells and the potential mechanism related to PPARγ, autophagy and ferroptosis. The serum of rat containing different concentrations of YHD were collected to culture MDA-MB-231 cells. Cell viability and proliferation were assessed by the CCK-8 assay and EDU staining. Wound healing- and transwell assays were used to detect the capacities of MDA-MB-231 cell migration and invasion. Additionally, the levels of lipid peroxidation, Fe2+ and the expression of ferroptosis-related proteins were evaluated. The expression of PPARγ and autophagy-related proteins was assessed using immunofluorescence staining or western blot assay. Then, the PPARγ inhibitor (GW9662), autophagy inhibitor (3-MA) and autophagy inducer (rapamycin; Rap) were used to further study the potential mechanism of YHD on TNBC. Results indicated that contained-YHD serum significantly decreased the viability, proliferation, migration and invasion of TNBC cells. Moreover, YHD promoted lipid peroxidation level, elevated Fe2+ content and downregulated GPX4, SLC7A11 and SLC3A2 expression. Besides, autophagy was induced and PPARγ was upregulated by YHD in MDA-MB-231 cells. Furthermore, GW9662 alleviated the impacts of YHD on autophagy of MDA-MB-231 cells. Rap reversed the effects of GW9662 on lipid peroxidation, ferroptosis, proliferation, migration and invasion of MDA-MB-231 cells. 3-MA had the similar effects to GW9662. Collectively, YHD suppressed the malignant progression of MDA-MB-231 cells by inducing ferroptosis through PPARγ-dependent autophagy.
Collapse
Affiliation(s)
- YangZi Cheng
- Department of Medical Oncology, Xi'an Ninth Hospital, Xi'an, Shaanxi 710054, China.
| | - GuiPing Yu
- Department of Medical Oncology, Xi'an Ninth Hospital, Xi'an, Shaanxi 710054, China
| | - Chen Du
- Department of Medical Oncology, Xi'an Ninth Hospital, Xi'an, Shaanxi 710054, China
| | - ZhaoHui Chen
- Department of Medical Oncology, Xi'an Ninth Hospital, Xi'an, Shaanxi 710054, China
| | - XuFeng Liu
- Department of Medical Oncology, Xi'an Ninth Hospital, Xi'an, Shaanxi 710054, China
| |
Collapse
|