1
|
Liu M, Zou G, Lu M, Fu J, Chen H, Pan C, Liu HM, Fu L. Mechanism of Rabdosia rubescens extract against gastric cancer microenvironment by SIRT1/NF-κB/p53 pathway and promoting tumor-associated macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119935. [PMID: 40345273 DOI: 10.1016/j.jep.2025.119935] [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: 02/07/2025] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional action of Rabdosia rubescens (Hemsl.) H. Hara is heat-clearing and detoxifying, relieve sore throat, dissipate binds and disperse swelling. DLC, as an extract prepared from Rabdosiae Rubescentis Herba, could regulate the polarization of tumor associated macrophages (TAMs). For TAMs play an important role in the tumor microenvironment. It is worthy to further explore the mechanism of DLC on the polarized function of macrophages. AIM OF THE STUDY The aim of this study is to investigate the activity and molecular mechanisms of DLC on dissipating binds and dispersing swelling by modulating the gastric cancer microenvironment and macrophage polarization. MATERIALS AND METHODS We conducted comprehensive qualitative and quantitative chromatographic analyses to characterize the main components of DLC. To evaluate its anti-tumor effects, immunofluorescence, MTT assay, plate cloning, transcriptomics analysis, western blotting, and siRNA knockdown experiments were performed to assess DLC's action on gastric cancer cell proliferation. Additionally, we utilized Trypan blue staining, a THP-1 and MGC-803 co-culture model, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and a mouse xenograft model with five distinct dosage groups to systematically investigate DLC's effects on macrophage polarization. RESULTS Key compounds in DLC were identified. The vivo tests demonstrated the tumor inhibition rate of the 5 g/kg DLC group reached 66.99 %, surpassing that of the 5-fluorouracil group (59.94 %). Mechanistically, DLC upregulated SIRT1 expression and suppressed NF-κB pathway, thereby preventing p65 from translocating into nuclear and modulating downstream p53/MDM2/USP7 signaling. Moreover, DLC enhanced M1 macrophage factors such as TNF-α, IL-6 while inhibiting M2 marker TGF-β, effectively repolarizing M2 TAMs toward an M1 phenotype. This effect was associated with suppressed protein expression of HIF-1α, p-p65, and p-PI3K. CONCLUSION This study provides insights into DLC's mechanisms in regulating tumor microenvironment remodeling and promoting macrophage polarization toward an anti-tumor phenotype. These results provide a solid basis for DLC's potential clinical treament in gastric cancer, highlighting its promise as a natural therapeutic agent.
Collapse
Affiliation(s)
- Mengran Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Guona Zou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Mengyao Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiayue Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Han Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Chengxue Pan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| | - Ling Fu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou, 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
2
|
Zhang C, Wang G, Xu J, Wang W, Sun P, Hu S. A review of the role of CSCs and CSC-EXOs in increasing drug resistance in breast cancer and future applications. Crit Rev Oncol Hematol 2025; 212:104774. [PMID: 40412576 DOI: 10.1016/j.critrevonc.2025.104774] [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: 04/12/2025] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Breast cancer (BCa) remains a major global health challenge due to its complex etiology, varied clinical manifestations, and therapy resistance, which contributes to nearly 90 % of cancer-related deaths. A key factor in treatment failure is the presence of BCa stem cells (BCSCs), which drive drug resistance and tumor recurrence. Understanding BCSC formation, regulation, and role in therapeutic resistance is crucial for developing targeted therapies. Additionally, exosomes (EXOs) secreted by BCSCs play a critical role in cancer progression and drug resistance. These vesicles facilitate communication between cancer and stromal cells by transferring RNA and proteins, influencing treatment response. For instance, EXOs from stromal cells can enhance BCa cell survival under chemotherapy and radiation. This study explores BCSC mechanisms, their contribution to drug resistance, and emerging therapeutic strategies. We also examine how BCa-derived and BCSC-derived EXOs promote drug tolerance and tumor growth. Finally, we discuss future treatment approaches, current research limitations, and potential solutions to advance BCa therapy. Novel interventions may overcome resistance and improve patient outcomes by targeting BCSCs and EXO-mediated pathways. However, further research is needed to translate these findings into practical clinical applications.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Liver Transplant Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Liver Transplant Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jianjun Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Liver Transplant Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenqiang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Liver Transplant Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ping Sun
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Liver Transplant Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
3
|
Banerjee B, Sharma T, Banerjee S, Banerjee S, Mondal A. Synthesis and Anticancer Activity of Metformin-Phenolic Acid Conjugates. Chem Biodivers 2025:e202403199. [PMID: 39956923 DOI: 10.1002/cbdv.202403199] [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: 12/03/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
The leading cause of death worldwide is cancer. Several studies suggest phenolic acids and metformin as potential cancer treatment options because of their biological and therapeutic properties. So, we synthesized some novel metformin-phenolic acid conjugates. We used an acid-base neutralization method to extract the metformin-free base. N,N'-dicyclohexylcarbodiimide-4-dimethylaminopyridine coupling of phenolic/aromatic acids (benzoic acid, cinnamic acid, caffeic acid, ferulic acid, gallic acid, para-hydroxybenzoic acid, para coumaric acid, protocatechuic acid, salicylic acid, and vanillic acid) with metformin was performed to produce metformin phenolic acid conjugates (M1-M10). We evaluated the structures using proton nuclear magnetic resonance, carbon-13 nuclear magnetic resonance, Fourier-transform infrared, and mass spectroscopy. All newly synthesized metformin phenolic acid conjugates were evaluated for their in vitro anticancer activity. Metformin phenolic acid conjugates were synthesized and showed a range of inhibitory effects. The metformin-caffeic acid conjugate [(E)-3-(3,4-dihydroxyphenyl)-N-(N,N-dimethylcarbamimidoyl)carbamimidoyl)acrylamide] (M3) (IC50: 5.47 ± 2.72 and 4.42 ± 2.15 µg/mL) showed the best anticancer activity against MDA-MB-468 and A549 cancer cell lines.
Collapse
Affiliation(s)
- Biswajit Banerjee
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Siksha' O' Anusandhan, Deemed to be University, Bhubaneswar, India
| | - Tripti Sharma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences and Research, Chhatrapati Shivaji Maharaj University, Navi Mumbai, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Technology, Eminent College of Pharmaceutical Technology, Kolkata, India
| | - Arijit Mondal
- Department of Pharmaceutical Chemistry, Anand College of Education, Debra, India
| |
Collapse
|
4
|
Cheng X, Sun J, Chen S, Wang N, Tang W, Xia Z, Shu Y, Gao S, Wang Z, Wang X, Shao R, Cao J. Parishin B blocking TRIB3-AKT1 interaction inhibits breast cancer lung metastasis. Front Pharmacol 2025; 15:1517708. [PMID: 39881875 PMCID: PMC11775015 DOI: 10.3389/fphar.2024.1517708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Background TRIB3 has been reported to mediate breast cancer (BC) proliferation and metastasis by interacting with AKT1, and blocking the interaction between TRIB3 and AKT1 can inhibit the progression of BC. Besides, inhibiting TRIB3 to turn "cold tumor" hot has also been proved to be an effective therapeutic strategy for BC. Thus, this study aim to find drugs that can bind to TRIB3 to inhibit BC progression, and further elucidate its mechanism. Methods The possible inhibitors of TRIB3 were screened by high-throughput molecular docking, CETSA, and CO-IP assay. Then, the effect of TRIB3 inhibitor anti BC was assessed by CCK-8 assay, flow cytometry, plate colony formation assay, and transwell assay; and the RNA-seq was empolyed to study the potential mechanism of Parishin B (PB) anti-BC. Finally, the effect of TRIB3 inhibitor on BC lung metastasis in vivo was evaluated. Results PB was screened as a possible inhibitor of TRIB3, and CETSA and CO-IP assay indicated that PB could target TRIB3 and block TRIB3-AKT1 interaction. In addition, PB exhibited good anti-BC activity without drug toxicity in normal breast cells by experiments in vitro, and RNA-seq analysis suggested PB could inhibit the proliferation and invasion of BC cells related with cell cycle. It was also proved that PB could inhibit BC lung metastasis in vivo. Conclusion The study demonstrated PB can bind to TRIB3 to inhibit BC proliferation and lung metastasis by blocking TRIB3-AKT1 interaction and regulating cell cycle, providing a therapeutic agent for the treatment of BC.
Collapse
Affiliation(s)
- Xiongtao Cheng
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jianguo Sun
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shouhong Chen
- Department of Oncology, Guangzhou Concord Cancer Center, Guangzhou, Guangdong, China
| | - Nan Wang
- Department of Oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Weijing Tang
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zijian Xia
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yuhong Shu
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xinxia Wang
- Department of Pharmacy, Shanghai Jiahui International Hospital, Shanghai, China
| | - Rongzi Shao
- Department of Pharmacy, The 960th Hospital of PLA Joint Logistics Support Force, Jinan, China
| | - Jianxiong Cao
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|