1
|
Wang Y, Jiang N, Wu S, Lin G, Lu W, Shang B, Zhu X, Han W, Li J, Chen Y. An injectable hydrogel for hemostasis and tumor suppression in intraoperative breast cancer. BIOMATERIALS ADVANCES 2025; 172:214219. [PMID: 39987716 DOI: 10.1016/j.bioadv.2025.214219] [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: 07/22/2024] [Revised: 01/14/2025] [Accepted: 02/02/2025] [Indexed: 02/25/2025]
Abstract
In the period between surgery and systemic therapy for breast cancer, residual tumor cells may proliferate, leading to tumor recurrence. Additionally, intraoperative wound bleeding may cause surgical failure or the spread of tumor cells. This study introduces an innovative injectable hydrogel composed of oxidized hyaluronic acid (OHA) loaded 5-fluorouracil (5-FU) and N-carboxyethyl chitosan (CEC), designed for intraoperative hemostasis and tumor suppression in intraoperative breast cancer. The CEC/OHA injectable hydrogel was synthesized through a Schiff base reaction between the aldehyde group of OHA and the amino group of CEC, incorporating 5-FU during hydrogel formation. This CEC/OHA injectable hydrogel demonstrated hemostatic effects comparable to gelatin sponges in both an in vivo rat liver hemorrhage model and an in vitro rat tail amputation model. When loaded with 5-FU, the injectable hydrogel effectively inhibited the proliferation of MDA-MB-231 breast cancer cells in vitro, significantly inhibited tumor growth and recurrence in vivo, and did not induce significant damage or inflammatory response in any major organ. This CEC/OHA & 5-FU injectable hydrogel is envisioned as a complementary therapeutic regimen during the intraoperative period in breast cancer surgery to prevent hemostasis and tumor recurrence.
Collapse
Affiliation(s)
- Yue Wang
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Nan Jiang
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Shuhan Wu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Guangshuai Lin
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Wudang Lu
- Xi'an Libang Pharmaceutical Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Bin Shang
- Xi'an Libang Pharmaceutical Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Xulong Zhu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Wei Han
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Jianhui Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China.
| | - Yongmei Chen
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, Shaanxi 710021, China.
| |
Collapse
|
2
|
Cui D, Zhang C, Zhang L, Zheng J, Wang J, He L, Jin H, Kang Q, Zhang Y, Li N, Sun Z, Zheng W, Wei J, Zhang S, Feng Y, Tan W, Zhong Z. Natural anti-cancer products: insights from herbal medicine. Chin Med 2025; 20:82. [PMID: 40490812 DOI: 10.1186/s13020-025-01124-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 05/05/2025] [Indexed: 06/11/2025] Open
Abstract
Herbal medicine exhibits a broad spectrum of potent anti-cancer properties, including the enhancement of tumor immune responses, reversal of multidrug resistance, regulation of autophagy and ferroptosis, as well as anti-proliferative, pro-apoptotic, and anti-metastatic effects. This review systematically explores recent advances (primarily documented since 2019) in research on key anti-cancer compounds derived from herbal medicine, such as apigenin, artemisinin, berberine, curcumin, emodin, epigallocatechin gallate (EGCG), ginsenosides, icariin, resveratrol, silibinin, triptolide, and ursolic acid (UA). These studies were sourced from scientific databases, including PubMed, Web of Science, Medline, Scopus, and Clinical Trials. The review focuses on the significant role that these natural products play in modern oncology, exploring their efficacy, mechanisms of action, and the challenges and prospects of integrating them into conventional cancer therapies. Furthermore, it highlights cutting-edge approaches in cancer research, such as the utilization of gut microbiota, omics technologies, synthetic derivatives, and advanced drug delivery systems (DDS). This review underscores the potential of these natural products to advance the development of novel anti-cancer treatments and support contemporary medicine. Additionally, recent multi-omics findings reveal how these compounds reshape transcriptional and metabolic networks, further broadening their therapeutic scope. Many natural products exhibit synergy with first-line chemotherapies or targeted therapies, thereby enhancing treatment efficacy and reducing side effects. Advanced nano-formulations and antibody-drug conjugates have also substantially improved their bioavailability, making them promising candidates for future translational research.
Collapse
Affiliation(s)
- Dianxin Cui
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 6/F, 3 Sassoon Road, Pokfulam, Hong Kong S.A.R., 999077, China
| | - Lili Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Jingbin Zheng
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Jie Wang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Luying He
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Haochun Jin
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Na Li
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Zhenlong Sun
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Wenying Zheng
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Jinchao Wei
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Siyuan Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 6/F, 3 Sassoon Road, Pokfulam, Hong Kong S.A.R., 999077, China.
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao S.A.R., 999078, China.
| |
Collapse
|
3
|
Sun L, Liu Y, Sun Q, Wang G, Du B, Liu B, Gao T, Zhao P, Yang Y, Rong R. Polysaccharides from traditional Chinese medicine and their nano-formulated delivery systems for cancer immunotherapy. Carbohydr Polym 2025; 357:123416. [PMID: 40158963 DOI: 10.1016/j.carbpol.2025.123416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has evolved into a new generation strategy in the field of anti-tumor treatment. Polysaccharides derived from Traditional Chinese Medicine (TCM) are gaining recognition as powerful immunomodulators in cancer therapy, noted for their multi-target and multi-pathway actions. Owing to their beneficial properties such as water solubility, biocompatibility, and chemical structure modifiability, TCM polysaccharides can also serve as carriers for hydrophobic drugs in the development of innovative drug delivery systems, enhancing synergistic antitumor effects. In this article, we summarize the diverse mechanisms of immunoregulation by TCM polysaccharides in tumor therapy. The applications of these polysaccharides as both active ingredients and drug carriers within nanodelivery systems for cancer immunotherapy are also introduced. Additionally, extensive research on TCM polysaccharides in clinical settings has been collected. Furthermore, discussions are presented on the development prospects and challenges faced by these polysaccharides in the field of tumor immunotherapy. Our goal is to improve researchers' comprehension of TCM polysaccharides in cancer immunotherapy, providing promising strategies to optimize cancer treatment and benefit diverse patient populations.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yuting Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Guimei Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Bodong Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Collaborative Innovation Center for Antiviral Traditional Chinese Medicine in Shandong Province, Jinan 250355, PR China; Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
4
|
Guo J, Yang X, Yang J, Du F, Liu S. Electroacupuncture Promotes the Proliferation and Differentiation of Enteric Neural Precursor Cells via the PTEN/PI3K/Akt/mTOR Signaling Pathway in Diabetic Mice. Neurogastroenterol Motil 2025; 37:e70040. [PMID: 40190044 DOI: 10.1111/nmo.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 02/15/2025] [Accepted: 03/11/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Enteric neuronal loss significantly contributes to gastrointestinal (GI) motility disorders. Electroacupuncture (EA) can promote the regeneration of lost enteric neurons in diabetic mice, but its mechanisms are not fully understood. Nestin+/Ngfr+ cells can function as enteric neural precursor cells (ENPCs) to proliferate and differentiate into enteric neurons in adult mice. However, EA's effects on ENPCs remain unknown. The study aimed to investigate whether EA reversed enteric neuronal loss via regulation of ENPCs and its molecular basis. MATERIALS AND METHODS The study utilized conventional C57BL/6J mice and ENPC-tracing transgenic mice. Streptozotocin-induced type 1 diabetic mouse, PI3K inhibitor, and PTEN inhibitor models were used. GI motility was evaluated by defecation frequency, fecal water content, and whole gut transit test. The alterations of enteric neurons, ENPCs, and PTEN/PI3K/Akt/mTOR signaling were detected by Western blot and immunofluorescence. RESULTS EA increased defecation frequency and fecal water content, reduced whole gut transit time, and increased the number of enteric neurons. Notably, EA inhibited ENPC apoptosis and facilitated ENPC proliferation and differentiation with a preferential into ChAT enteric neurons. Additionally, PTEN was decreased and PI3K/Akt/mTOR signaling was activated with EA. However, LY294002 (PI3K inhibitor) inhibited EA's effects on ENPCs, while BpV(HOpic) (PTEN inhibitor) partially rescued these inhibitory effects. CONCLUSIONS EA alleviates diabetic enteric neuropathy by regulating ENPC dynamics through the PTEN/PI3K/Akt/mTOR signaling pathway. Notably, EA-mediated anti-apoptotic and pro-proliferative effects on ENPCs, and their preferential cholinergic differentiation establish EA as a multimodal therapy that bridges neuromodulation with precursor cell biology, offering an alternative strategy for GI motility disorders.
Collapse
Affiliation(s)
- Jinlu Guo
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Yang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingze Yang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Du
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Liu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Liu WM, Chen CY, Ma HQ, Zhang QQ, Zhou X, Wu YL, Huang WJ, Qi XS, Zhang YX, Tang D, Sun HY, Wu HP, Jiao YF, He ZY, Yu WF, Yan HX. Inhibition of liver cancer cell growth by metabolites S-adenosylmethionine and nicotinic acid originating from liver progenitor cells. J Gastroenterol 2025; 60:754-769. [PMID: 40019515 PMCID: PMC12095412 DOI: 10.1007/s00535-025-02226-y] [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: 10/09/2024] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), the most common form of liver cancer, presents a challenging malignancy with scarce treatment options. Liver progenitor cells (LPCs) play a pivotal role in both liver regeneration and the progression of liver cancer, yet the specific functions of LPCs from different origins in liver cancer remain to be fully elucidated. METHODS We explored the liver progenitor-like cells derived from human hepatocytes (HepLPCs) on the proliferation of HCC both in vitro and in vivo. The mitochondrial function was assessed through electron microscopy and functional experiments. Transcriptomic sequencing and western blot unveiled the fundamental mechanisms at play, whereas metabolomic sequencing pinpointed crucial effector molecules involved in the paracrine secretion of HepLPCs. RESULTS By employing a co-culture system of HepLPCs and HCC cells, we found that HepLPCs markedly inhibited HCC growth by prompting mitochondrial dysfunction, which further led to the co-inhibition of the Notch1 and JAK1/STAT3 signaling pathways through paracrine actions involving S-adenosylmethionine (SAM) and Nicotinic acid (NA). CONCLUSIONS This study has uncovered that HepLPCs have a suppressive influence on the proliferation of HCC cells. This is achieved through the impairment of mitochondrial function and the inhibition of key signaling pathways, namely, Notch1 and JAK1/STAT3, which are critical drivers of cancer progression. The secretion of the metabolites SAM and NA by HepLPCs appears to be instrumental in mediating these effects. These findings provide a solid foundation for identifying new therapeutic targets and clarifying the mechanisms through which HepLPCs can be harnessed to effectively treat HCC.
Collapse
Affiliation(s)
- Wen-Ming Liu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China
| | - Cai-Yang Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong-Qian Ma
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China
| | - Qiu-Qiu Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China
| | - Xu Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu-Ling Wu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei-Jian Huang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao-Shu Qi
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu-Xin Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China
| | - Han-Yong Sun
- Department of Liver Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong-Ping Wu
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Ying-Fu Jiao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China
| | - Zhi-Ying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China.
| | - He-Xin Yan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
- Shanghai Engineering Research Center of Peri-Operative Organ Support and Function Preservation (20DZ2254200), Renji Hospital, Shanghai, China.
| |
Collapse
|
6
|
Xi J, Ji C, Sun H, Wu Y, Shi C, Li S, Yang T, Shen Y, Li Y, Fan Y, Zhao Q, Liu S, Xie T, Chen G. Research progress on new physical therapies for cancer (Review). Oncol Lett 2025; 29:313. [PMID: 40337606 PMCID: PMC12056479 DOI: 10.3892/ol.2025.15059] [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: 12/28/2024] [Accepted: 04/04/2025] [Indexed: 05/09/2025] Open
Abstract
Currently, the clinical treatment of cancer is mainly based on surgery, chemotherapy and radiotherapy, but there are still problems associated with these treatments, such as disease recurrence and adverse reactions. The complexity and harmful nature of cancer mean that combining multiple treatment methods is an inevitable response. Therefore, it is of theoretical and practical significance to expand upon and study the aforementioned classic and traditional measures. With the advancement of technology, physical therapy has become important in the current research and treatment of cancer, and the physical factors related to cancer deserve in-depth study and discussion. The present review aimed to describe the mechanisms of action of pressure, temperature, photo-, sound and other physical therapies for cancer, which may provide new avenues for cancer treatment.
Collapse
Affiliation(s)
- Jingyi Xi
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Cheng Ji
- Department of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Haixin Sun
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yurun Wu
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Chengjie Shi
- Department of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Shasha Li
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Tao Yang
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yuxiang Shen
- Department of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yulin Li
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yaoxuan Fan
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Qichao Zhao
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Shuiping Liu
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Tian Xie
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Gongxing Chen
- Department of Pharmacy Experiment, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| |
Collapse
|
7
|
Li FL, Wang BB, Zeng KF, Chen HY, Wu XH, Wang Y, Lin HC, Li WL, Zhao XD. Exploring the Anticancer Effects of Xianliu Jieduan Fang on Colitis-Associated Colorectal Cancer Through Network Pharmacology and Experimental Validation. Biomed Chromatogr 2025; 39:e70102. [PMID: 40324811 DOI: 10.1002/bmc.70102] [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: 03/28/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
This study evaluated the therapeutic effects of Xianliu Jieduan Fang (XLJDF) on colitis-associated colorectal cancer (CAC) and explored its molecular mechanisms through network pharmacology and experimental validation. Using an AOM/DSS-induced CAC mouse model, we evaluated XLJDF's efficacy. Active components were identified by UHPLC-QE-HRMS. Targets were predicted using SwissTargetPrediction and PubChem, while disease genes were obtained from GeneCards, DisGeNET, and TTD. Core targets and pathways were analyzed via Cytoscape and Metascape. Mechanisms were validated through molecular docking and experiments. XLJDF improved colon pathology and identified 68 active compounds, including nine key components like Kaempferol and Luteolin. Network analysis revealed 959 targets with 29 core genes (AKT1, CTNNB1, GSK3B, etc.). KEGG analysis showed XLJDF primarily acts through Wnt signaling, regulating apoptosis and cell migration. Experimental validation confirmed XLJDF inhibits Wnt/β-catenin pathway by preventing GSK3β inactivation. XLJDF exerts anti-CAC effects via a multi-component, multi-target network. Our study identifies key active compounds and demonstrates that XLJDF suppresses the Wnt/β-catenin pathway by preventing GSK3β inactivation, thereby inhibiting β-catenin stabilization.
Collapse
Affiliation(s)
- Fang-Lan Li
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bei-Bei Wang
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ke-Feng Zeng
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao-Yang Chen
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xi-Hua Wu
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yun Wang
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Hong-Cheng Lin
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wei-Lin Li
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiang-Dong Zhao
- Department of Anorectal, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
8
|
Gong YX. Research progress of effective components of traditional Chinese medicine in intervening apoptosis of renal tubular epithelial cells in diabetic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119874. [PMID: 40280372 DOI: 10.1016/j.jep.2025.119874] [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: 12/11/2024] [Revised: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Apoptosis of renal tubular epithelial cells (RTECs) is a critical pathological feature of diabetic kidney disease (DKD), a primary contributor to end-stage renal disease (ESRD). Traditional Chinese medicine (TCM) has shown potential in modulating RTECs apoptosis and alleviating DKD progression, making it a promising area for further investigation. AIM OF THE STUDY This study aims to summarize the apoptotic pathways implicated in DKD, analyze existing research on the effects of TCM monomers and compounds on RTECs apoptosis, and elucidate the molecular mechanisms underlying these effects. Additionally, this study emphasizes the significant role of TCM in mitigating DKD progression. MATERIALS AND METHODS Relevant literature was systematically retrieved from ancient Chinese medicine texts and modern scientific databases, including CNKI, Web of Science, and PubMed, using keywords such as "Traditional Chinese Medicine", "Diabetic Kidney Disease", "Diabetic Nephropathy", "Renal Tubular Epithelial Cells", and "Apoptosis". The collected information was synthesized and analyzed. RESULTS This review systematically analyzed 187 relevant studies, focusing on the mechanisms and clinical applications of 16 TCM monomers and 20 TCM compounds in DKD treatment. Key bioactive compounds, such as berberine, astragaloside IV, and tanshinone IIA, have demonstrated renoprotective effects by mitigating oxidative stress and inflammation, as well as regulating critical signaling pathways, including PI3K/Akt, NF-κB, and TGF-β/Smad, to suppress RTECs apoptosis and decelerate DKD progression. Additionally, several TCM compounds have shown significant efficacy in clinical studies, reducing proteinuria and enhancing renal function, thereby reinforcing the therapeutic potential of TCM in DKD management. CONCLUSIONS RTECs apoptosis is a critical pathological feature of DKD. TCM exhibits significant therapeutic potential by intervening in this process through multiple pathways. This study highlights the ability of TCM to exert anti-apoptotic and renoprotective effects by modulating oxidative stress, inflammatory responses, and multiple cellular signaling pathways. The multi-component and multi-target characteristics of TCM offer a promising avenue for the development of novel therapeutic strategies. However, further rigorous research and high-quality clinical trials are required to validate its efficacy and elucidate its mechanisms of action.
Collapse
Affiliation(s)
- Yu Xin Gong
- Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
9
|
Mandriota G, Avugadda SK, Sadeghi E, Silvestri N, Marotta R, Gavilán H, Olsson U, Giannini C, Tsai YH, Samia ACS, Pellegrino T. Magnetic nanosheets: from iron oxide nanocubes to polydopamine embedded 2D clusters and their multi-purpose properties. NANOSCALE HORIZONS 2025; 10:1140-1158. [PMID: 40191967 PMCID: PMC11973963 DOI: 10.1039/d4nh00566j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/19/2025] [Indexed: 04/10/2025]
Abstract
We here develop stable bidimensional magnetic nanoclusters (2D-MNCs) of iron oxide nanocubes (IONCs) arranged in thin nanosheets of closed-packed nanocubes. The assembly occurs by means of a two-step approach: in the first one, the ionic surfactant, sodium dodecyl sulfate (SDS), acts as a transient water transfer agent and as 2D clustering agent to induce formation of a monolayer of nanocubes arranged in thin nanosheets. Next, the addition of dopamine followed by solution basification, induces the in situ polymerization of dopamine with a tunable shell tickness depending on the dopamine amount, which helps to compact the clusters and ensures the long term water stability of the clusters. TEM, cryo-EM, and SAXS techniques helped to reveal structural features of the 2D-clusters. The pH-dependent degradation properties of polydopamine, enable to disassemble the clusters in acidic tumour microenviroment leading to a four-fold increase in the magnetic particle imaging signal and a concomitant increase of the magnetic heat losses of these clusters, makes them appealing in magnetic hyperthermia, while the shortening of T2 relaxation time suggests their use as contrast in magnetic resonance imaging. Finally, with crystal violet dye, used as drug molecule, the feasibility to release payloads pre-encapsulated with the polydopamine polymer shell has been also shown.
Collapse
Affiliation(s)
- Giacomo Mandriota
- Italian Institute of Technology, via Morego 30, 16163, Genoa, Italy.
| | | | - Ehsan Sadeghi
- Italian Institute of Technology, via Morego 30, 16163, Genoa, Italy.
- Chemical and Chemical Industry Department, via Dodecaneso, 31, Genoa, 16146, Italy
| | - Niccolò Silvestri
- Italian Institute of Technology, via Morego 30, 16163, Genoa, Italy.
| | - Roberto Marotta
- Italian Institute of Technology, via Morego 30, 16163, Genoa, Italy.
| | - Helena Gavilán
- Italian Institute of Technology, via Morego 30, 16163, Genoa, Italy.
| | - Ulf Olsson
- Physical Chemistry, Lund University, Box 124, Lund SE-22100, Sweden
| | - Cinzia Giannini
- Institute of Crystallography, National Research Council, via Amendola 122/O, 70126, Bari, Italy
| | - Yu Hsin Tsai
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| | - Anna Cristina S Samia
- Department of Chemistry, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| | - Teresa Pellegrino
- Italian Institute of Technology, via Morego 30, 16163, Genoa, Italy.
| |
Collapse
|
10
|
Lin M, Liu D, Gong Y, Shu L, Wang H, Zhang G, Li J, Gao Z, Sun J, Chen X. Bioactive Assembly Cofactor-Assisted Ursolic Acid Helix for Enhanced Anticancer Efficacy via In Situ Virus-like Transition. J Am Chem Soc 2025; 147:17010-17021. [PMID: 40354555 DOI: 10.1021/jacs.5c01214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Natural bioactive pentacyclic triterpenoids, such as ursolic acid (UA), hold significant potential as anticancer agents. However, their clinical application is limited by their poor solubility and bioavailability. Herein, we developed a novel polypeptoid assembly cofactor-assisted nanoplatform designed to enhance UA's therapeutic efficacy through in situ self-assembly within the tumor microenvironment (TME). Bioactive polypeptoid polyelectrolytes, inspired by natural molecular chaperones, were utilized as assembly cofactors to guide UA's co-assembly into stimuli-responsive nanostructures. These polypeptoids provide precise control over the assembly process, improving stability and enabling reversible, pH-responsive transformations. Acid-responsive groups and the target molecule lactobionic acid further promote the specificity and efficacy of UA delivery. Under neutral conditions, the assemblies retain a helical fibrous structure, while in the acidic TME, they transform into virus-like clusters composed of assembly subunits, facilitating deeper tumor penetration. Once internalized, these nanoparticles escape into the cytoplasm and accumulate around the mitochondria, where the oxidation of thioether bonds triggers the release of UA and polypeptoids, causing mitochondrial damage and apoptosis. Some nanoparticles reassemble into fibrous structures intracellularly, extending their retention in tumor cells and potentially leading to mitochondria damage. Notably, the nanoplatform demonstrates excellent synergistic effects, achieving significantly higher therapeutic efficiency compared with individual components, including UA and polypeptoids. In vivo studies further confirmed the effectiveness, demonstrating significant tumor growth suppression and reduced metastasis. By integrating the therapeutic UA with bioactive polypeptoids under precise control, this synergistic platform represents a highly efficient and targeted approach to cancer therapy, offering a promising new opportunity for natural compounds for advanced nanomedicine.
Collapse
Affiliation(s)
- Min Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Dandan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Yiyu Gong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Lilei Shu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Helin Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Guojing Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Jiayi Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Zixin Gao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Jing Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Xuesi Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022 Changchun, China
| |
Collapse
|
11
|
Yuan Z, Mao F, Liu Z, Xing S, Yang L, Wu W, Xu W, Deng J, Wang G. Characteristic of motor-related cortical network during stimulating at the affected or bilateral limbs of stroke patients by acupuncture. Brain Res Bull 2025; 227:111393. [PMID: 40398702 DOI: 10.1016/j.brainresbull.2025.111393] [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: 03/16/2025] [Revised: 04/29/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND The compensatory pattern between the two hemispheres after stroke has been the focus of research. Some evidence suggests bilateral stimulation more effectively engages networks across both hemispheres compared to the affected side only. OBJECTIVES To explore whether the stimulating at bilateral limbs of stroke patients by acupuncture may better engage compensatory reorganization between the hemispheres compared to stimulating at the hemiplegic limb. METHODS Conscious patients with hemiplegia were screened. Brain activity was assessed by the functional near-infrared spectroscopy(fNIRS) in three states: no treatment, acupuncture on the affected side, and then acupuncture on both sides. Brain activation and directed functional connectivity(FC) was analyzed between the two acupuncture strategies. RESULTS Acupuncture of bilateral limbs resulted in stronger activation in the primary motor cortex(M1) of the ipsilesional hemisphere than acupuncture of the affected side only. And no significantly enhanced activation of the contralesional hemisphere was observed after acupuncture on the healthy limb. Besides, the FCs from the ipsilesional premotor cortex to the contralesional sensory-related area were significantly enhanced, and the FCs from the sensory area to motor area within the ipsilesional hemisphere were also significantly enhanced. Additionally, FCs from contralesional M1 to ipsilesional motor area were attenuated. CONCLUSION Stimulating at bilateral limbs by acupuncture could lead to greater brain network remodeling in the motor-related areas compared to stimulating solely at the affected side, and not through more stimulation.
Collapse
Affiliation(s)
- Ziwen Yuan
- Department of Rehabilitation, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China; Research and Transformation Center of Intelligent Rehabilitation Assistance Devices, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fei Mao
- Department of Rehabilitation, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Research and Transformation Center of Intelligent Rehabilitation Assistance Devices, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zimo Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shubo Xing
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Liu Yang
- Department of Rehabilitation, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China; Research and Transformation Center of Intelligent Rehabilitation Assistance Devices, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenping Wu
- Department of Rehabilitation, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Research and Transformation Center of Intelligent Rehabilitation Assistance Devices, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weiwei Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jingyuan Deng
- Department of Rehabilitation, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Research and Transformation Center of Intelligent Rehabilitation Assistance Devices, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Gang Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
12
|
Zheng H, Chen Y, Luo W, Han S, Sun M, Lin M, Wu C, Gao L, Xie T, Kong N. Integration of active ingredients from traditional Chinese medicine with nano-delivery systems for tumor immunotherapy. J Nanobiotechnology 2025; 23:357. [PMID: 40382641 PMCID: PMC12085060 DOI: 10.1186/s12951-025-03378-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
Tumor immune escape presents a significant challenge in cancer treatment, characterized by the upregulation of immune inhibitory molecules and dysfunction of immune cells. Tumor immunotherapy seeks to restore normal anti-tumor immune responses to control and eliminate tumors effectively. The active ingredients of traditional Chinese medicine (TCM) demonstrate a variety of anti-tumor activities and mechanisms, including the modulation of immune cell functions and inhibiting tumor-related suppressive factors, thereby potentially enhancing anti-tumor immune responses. Furthermore, nano-delivery systems function as efficient carriers to enhance the bioavailability and targeted delivery of TCM active ingredients, augmenting therapeutic efficacy. This review comprehensively analyzes the impact of TCM active ingredients on the immune system and explores the synergistic application of nano-delivery systems in combination with TCM active ingredients for enhancing tumor immunotherapy.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yiquan Chen
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Wei Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Shiqi Han
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Mengjuan Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Min Lin
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
| | - Chenghan Wu
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
| | - Lili Gao
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China.
| | - Tian Xie
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
13
|
Abdel-Badeea WIE, Abdel-Wahab A, Mahmoud AA, Abdel-Razik ARH, Attia EZ, Abdelmohsen UR, Hassanin KMA. Syzygium cumini Fruit Extract and Quercetin Ameliorate Cadmium-Induced Ovarian Apoptosis in Rats Via miRNA- 204 - 5p-Mediated Bcl- 2 Upregulation and Bax/Caspase 9/Caspase 3 Downregulation. Biol Trace Elem Res 2025:10.1007/s12011-025-04632-y. [PMID: 40360878 DOI: 10.1007/s12011-025-04632-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
The correlation between exposure to environmental pollutants and adverse impacts on fertility has been documented. Cadmium (Cd) is one of the most toxic elements that impairs female reproductive capacity. Syzygium cumini is a plant that contains polyphenols with strong antioxidant properties. Our study aimed to investigate the possible protective effects of Syzygium cumini fruit extract (SCFE) and quercetin (QU) against Cd-induced ovarian toxicity and apoptosis in rats assisted by measuring oxidative stress markers, hormonal profile, intrinsic apoptotic pathway, miRNA-204-5p, and histopathology. For that, 45 adult female rats were divided randomly into five groups; control, cadmium chloride (CdCl2), CdCl2 + QU, CdCl2 + SCFE200, and CdCl2 + SCFE400, and the experiment lasted 35 days. Cadmium exposure induced irregular estrus cycle, and oxidative challenges, as evidenced by increased malondialdehyde and decreased antioxidant indicators that include superoxide dismutase, catalase and reduced glutathione, and hormonal imbalances, including reduced follicle-stimulating hormone, luteinizing hormone, anti-mullarian hormone, estrogen, and progesterone levels. Additionally, Cd triggered ovarian apoptosis through upregulating miRNA- 204 - 5p expression that in turn downregulated Bcl- 2 and upregulated the members of intrinsic mitochondrial apoptotic pathway including Bax and caspases 3, 9. The histopathological findings, morphometric and lesion scoring in ovarian, and uterine tissues confirmed the negative impacts of Cd. Interestingly, treatment with QU or SCFE, especially at the higher dose (400 mg/kg), significantly ameliorated these Cd-induced adverse effects, suggesting their potential as protective agents against ovarian toxicity.
Collapse
Affiliation(s)
- Walaa I E Abdel-Badeea
- Biochemistry Department, Faculty of Veterinary Medicine, Minia University, 61519, Minia, Egypt.
| | - Ahmed Abdel-Wahab
- Physiology Department, Faculty of Veterinary Medicine, Minia University, 61519, Minia, Egypt
| | - Ahmed A Mahmoud
- Chemistry Department, Faculty of Sciences, Minia University, 61519, Minia, Egypt
| | | | - Eman Z Attia
- Pharmacognosy Department, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
- Pharmacognosy Department, Faculty of Pharmacy, National Minia University, 61111, New Minia, Egypt
| | - Usama R Abdelmohsen
- Pharmacognosy Department, Faculty of Pharmacy, Minia University, 61519, Minia, Egypt
- Pharmacognosy Department, Faculty of Pharmacy, Deraya University, 61111, New Minia, Egypt
| | - Kamel M A Hassanin
- Biochemistry Department, Faculty of Veterinary Medicine, Minia University, 61519, Minia, Egypt
| |
Collapse
|
14
|
Feng W, Yang K, Zou Y, Xiao Z, Qian R, Qian R. Progress of ursolic acid on the regulation of macrophage: summary and prospect. Front Immunol 2025; 16:1576771. [PMID: 40421013 PMCID: PMC12104263 DOI: 10.3389/fimmu.2025.1576771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/15/2025] [Indexed: 05/28/2025] Open
Abstract
Ursolic acid (UA), a prevalent pentacyclic triterpenoid found in numerous fruits and herbs, has garnered significant attention for its vital role in anti-inflammatory processes and immune regulation. The study of immune cells has consistently been a focal point, particularly regarding macrophages, which play crucial roles in antigen presentation, immunomodulation, the inflammatory response, and pathogen phagocytosis. This paper reveals the underlying regulatory effects of UA on the function of macrophages and the specific therapeutic effects of UA on a variety of diseases. Owing to the superior effect of UA on macrophages, different types of macrophages in different tissues have been described. Through the multifaceted regulation of macrophage function, UA may provide new ideas for the development of novel anti-inflammatory and immunomodulatory drugs. However, to facilitate its translation into actual medical means, the specific mechanism of UA in macrophages and its clinical application still need to be further studied.
Collapse
Affiliation(s)
- Wenjing Feng
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kehong Yang
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Ying Zou
- Department of Anatomy, Anatomy Teaching Center of Hunan University of Chinese Medicine, Changsha, China
| | - Zhaohua Xiao
- Xiangya Hospital, Central South University, Changsha, China
| | - Rongkang Qian
- Department of Integrated Traditional Chinese and Western Medicine, Qian Rongkang Clinic, Loudi, China
| | - Ronghua Qian
- Key Laboratory of Vascular Biology and Translational Medicine of Hunan Province, Medical School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
15
|
Wei LH, Chen X, Shen AL, Fang Y, Xie QR, Guo Z, Sferra TJ, Chen YQ, Peng J. Pien Tze Huang Attenuates Cell Proliferation and Stemness Promoted by miR-483-5p in Hepatocellular Carcinoma Cells. Chin J Integr Med 2025:10.1007/s11655-025-4126-0. [PMID: 40342037 DOI: 10.1007/s11655-025-4126-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 05/11/2025]
Abstract
OBJECTIVE To investigate the effect of miR-483-5p on hepatocellular carcinoma (HCC) cells proliferation and stemness, as well as the attenuating effect of Pien Tze Huang (PZH). METHODS Differentially expressed miRNA between HepG2 cells and hepatic cancer stem-like cells (HCSCs) were identified by a miRNA microarray assay. miR-483-5p mimics were transfected into HepG2 cells to explore the effects of miR-483-5p on cell proliferation and stemness. HepG2 cells and HCSCs were treated with PZH (0, 0.25, 0.50 and 0.75 mg/mL) to explore the effects of PZH on the proliferation and stemness, both in non-induced state and the state induced by miR-483-5p mimics. RESULTS miR-483-5p was significantly up-regulated in HCSCs and its overexpression increased cell proliferation and stemness in HepG2 cells (P<0.05). PZH not only significantly inhibited proliferation in HepG2 cells, but also significantly suppressed the cell proliferation and self-renewal of HCSCs (P<0.05). The effects of miR-483-5p mimics on proliferation and stemness of HepG2 cells were partially abolished by PZH. CONCLUSIONS miR-483-5p promotes proliferation and enhances stemness of HepG2 cells, which were attenuated by PZH, demonstrating that miR-483-5p is a potential molecular target for the treatment of HCC and provide experimental evidence to support clinical use of PZH for patients with HCC.
Collapse
Affiliation(s)
- Li-Hui Wei
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Xi Chen
- Department of Oncology, the 900th Hospital of the Chinese People's Liberation Army Joint Service Support Force, Fuzhou, 350003, China
| | - A-Ling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Yi Fang
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Qiu-Rong Xie
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Zhi Guo
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Thomas J Sferra
- Department of Pediatrics, Rainbow Babies and Children's Hospital and Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - You-Qin Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
- Department of Pediatrics, Rainbow Babies and Children's Hospital and Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| |
Collapse
|
16
|
Uchibori Y, Suekuni M, Kokaji Y, Yoshida K, Kiyono T, Kasahara Y, Fujita M. AmNA-Modified Antisense Oligonucleotide Targeting MCM8 as a Cancer-Specific Chemosensitizer for Platinum Compounds. Cancer Sci 2025; 116:1405-1416. [PMID: 40098305 PMCID: PMC12044654 DOI: 10.1111/cas.70024] [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: 12/18/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
MCM8 and MCM9 participate in homologous recombination with long-tract gene conversion to repair double-strand breaks caused by replication stress, which is generally higher in cancer cells than in normal cells. MCM8 is highly expressed in certain cancer cells, where it is necessary for maintaining cell growth, migration, and invasion, although the molecular mechanisms remain unclear. Knockdown with siRNAs or knockout of MCM8 or MCM9 selectively sensitizes cancer cells to cisplatin. Thus, drugs inhibiting MCM8 or MCM9 could serve as novel anti-neoplastic agents and/or chemosensitizers that selectively sensitize cancer cells to platinum compounds. The present study describes the development of an amido-bridged nucleic acid (AmNA)-modified gapmer antisense oligonucleotide (ASO) targeting MCM8, called ASO 8-3419. In vitro, ASO 8-3419 inhibited MCM8 expression in several human cell lines and selectively sensitized cancer cells to cisplatin. Moreover, ASO 8-3419 modestly suppressed the growth of several cancer cell lines whose proliferation has been reported to depend on MCM8. In vivo, ASO 8-3419 inhibited the expression of MCM8 in xenografted tumors of colon cancer-derived HCT116 cells in nude mice and increased tumor sensitivity to cisplatin with minimal toxicity. These findings suggest that AmNA-modified, MCM8-specific ASOs hold promise as novel anti-cancer agents.
Collapse
Affiliation(s)
- Yuki Uchibori
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical SciencesKyushu UniversityHigashi‐kuFukuokaJapan
| | - Masaki Suekuni
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical SciencesKyushu UniversityHigashi‐kuFukuokaJapan
| | - Yuko Kokaji
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical SciencesKyushu UniversityHigashi‐kuFukuokaJapan
| | - Kazumasa Yoshida
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical SciencesKyushu UniversityHigashi‐kuFukuokaJapan
| | - Tohru Kiyono
- Project for Prevention of HPV‐Related Cancer, Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaChibaJapan
| | - Yuuya Kasahara
- National Institutes of Biomedical InnovationHealth and Nutrition (NIBIOHN)IbarakiOsakaJapan
- Graduate School of Pharmaceutical SciencesOsaka UniversitySuitaOsakaJapan
| | - Masatoshi Fujita
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical SciencesKyushu UniversityHigashi‐kuFukuokaJapan
| |
Collapse
|
17
|
Tian Y, Zhang H, Xiang Z, Xu C, Xue H, Xu Q. Periodic tryptophan protein 1 promotes colorectal cancer growth via ribosome biogenesis. Int J Clin Oncol 2025; 30:944-955. [PMID: 40057905 DOI: 10.1007/s10147-025-02733-1] [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: 10/07/2024] [Accepted: 02/25/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Periodic tryptophan protein 1 (PWP1) is a member of the WD-40 family, located at the nucleus. The study of PWP1 in malignant tumors is at the initial stage. Its role and mechanism in colorectal cancer (CRC) remain unclear. METHODS KEGG pathway analysis were used for bioinformatics analysis. The protein expression of PWP1 in the tissue microarrays was detected by immunohistochemical methods. Colony and CCK-8 were used for PWP1 function in vitro, and the orthotopic model was used to assess PWP1 function in vivo. The growth of CRC was tracked by the Living Imaging System. Immunofluorescence was used for the quantification of nascent rRNAs. Polysome fractionation analysis was used to detect mRNA abundance. RESULTS GEO database was used to identify differential genes with elevated expression in CRC and followed by a KEGG analysis, which revealed that the ribosome synthesis pathway was enriched in CRC, with PWP1 displaying the most significant differential expression. Subsequently, the results of both in vitro and in vivo experiments demonstrated that PWP1 knockdown inhibited the proliferation of CRC. The results of immunofluorescence demonstrated that PWP1 knockdown suppressed de novo rRNA synthesis. Then, the differential proteins were examined, and this revealed that the most significantly downregulated proteins were those associated with DNA replication and mismatch repair functions following PWP1 interference. Moreover, ribosome profiling demonstrated significant downregulation of mRNAs associated with above functions. CONCLUSION We found that the PWP1-ribosome synthesis pathway is instrumental in achieving precise regulation of downstream signaling pathways, which in turn promote CRC growth.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Zeyu Xiang
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatologyschool of Medicineshanghai Institute of Digestive Disease, Ministry of Healthrenji Hospitalshanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Qing Xu
- Department of Gastrointestinal Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
18
|
Lin CH, Jiang WP, Itokazu N, Huang GJ. Chlorogenic acid attenuates 5-fluorouracil-induced intestinal mucositis in mice through SIRT1 signaling-mediated oxidative stress and inflammatory pathways. Biomed Pharmacother 2025; 186:117982. [PMID: 40106967 DOI: 10.1016/j.biopha.2025.117982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Mucositis, a common side effect of the chemotherapeutic drug 5-Fluorouracil (5-FU), causes severe and aggravating effects on mucosal cells in the oral cavity and intestine. This study in mice aimed to assess the antioxidant, anti-inflammatory, and mucosal protective properties of chlorogenic acid in mitigating 5-FU-induced intestinal mucositis. To investigate these potential protective effects, we developed a mouse model by administering an initial intraperitoneal (i.p.) injection of 5-FU, followed by daily i.p. injections of chlorogenic acid (10 and 20 mg/kg) for 10 consecutive days. Chlorogenic acid mitigated intestinal histopathological damage, reduced proinflammatory mediators and malondialdehyde (MDA) levels, and increased the glutathione (GSH) level by 5-FU. Chlorogenic acid treatment led to a significant reduction in the expression of inflammation-related proteins decreased oxidative stress-related proteins and, attenuated the expression of apoptosis and autophagy-related proteins in small intestinal tissues. Additional investigations are necessary to verify our findings and enhance our comprehension of how SIRT1 inhibition (EX-527) counteracts the anti-inflammatory effects of chlorogenic acid in intestinal tissues. In conclusion, our mice study has shown that chlorogenic acid exerts its protective effects on 5-FU-induced intestinal tissue damage, by reducing oxidative stress and inflammation through the modulation of multiple signaling pathways, including the TLR4/NF-κB/MAPK, AMPK/ SIRT1, and PI3K/AKT axis. These findings highlight the potential of chlorogenic acid as a therapeutic agent for mucositis, given its anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- Che-Hsuan Lin
- Department of Otolaryngology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Otolaryngology, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Wen-Ping Jiang
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Nanae Itokazu
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama 3620806, Japan
| | - Guan-Jhong Huang
- Department of Food Nutrition and Healthy Biotechnology, Asia University, Asia University, Taichung 413, Taiwan; Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
19
|
García-Beltrán A, Lozano Melero A, Martínez Martínez R, Porres Foulquie JM, López Jurado Romero de la Cruz M, Kapravelou G. A Systematic Review of the Beneficial Effects of Berry Extracts on Non-Alcoholic Fatty Liver Disease in Animal Models. Nutr Rev 2025; 83:819-841. [PMID: 39365946 PMCID: PMC11986334 DOI: 10.1093/nutrit/nuae132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024] Open
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in Western countries and is strongly associated with several metabolic disorders. Plant-derived bioactive extracts, such as berry extracts, with high antioxidant capacity have been used for the treatment and prevention of this pathology. Moreover, they promote circular economy and sustainability. OBJECTIVE To study the beneficial effects of extracts from different parts of berry plants in animal models of NAFLD. DATA SOURCES A systematic research of the MEDLINE (via PubMed), Cochrane, and Scopus databases was conducted to identify relevant studies published after January 2011. In vivo animal studies of NAFLD were included in which berry extracts of different parts of the plant were administered and significantly improved altered biomarkers related to the pathology, such as lipid metabolism and hepatic steatosis, glucose and glycogen metabolism, and antioxidant and anti-inflammatory biomarkers. DATA EXTRACTION Of a total of 203 articles identified, 31 studies were included after implementation of the inclusion and exclusion criteria. DATA ANALYSIS Most of the studies showed a decrease in steatosis and a stimulation of genes related to β-oxidation and downregulation of lipogenic genes, with administration of berry extracts. Berry extracts also attenuated inflammation and oxidative stress. CONCLUSIONS Administration of berry extracts seems to have promising potential in the design of enriched foodstuffs or nutraceuticals for the treatment of NAFLD.
Collapse
Affiliation(s)
- Alejandro García-Beltrán
- Department of Physiology, Biomedical Research Center, Instituto mixto de Deporte y Salud, University of Granada, 18007 Granada, Spain
| | - Aida Lozano Melero
- Department of Physiology, Biomedical Research Center, Instituto mixto de Deporte y Salud, University of Granada, 18007 Granada, Spain
| | - Rosario Martínez Martínez
- Department of Physiology, Biomedical Research Center, Instituto mixto de Deporte y Salud, University of Granada, 18007 Granada, Spain
| | | | | | - Garyfallia Kapravelou
- Department of Physiology, Faculty of Health Sciences, Campus of Melilla, University of Granada, 52005 Granada, Spain
| |
Collapse
|
20
|
Lu X, Friedrich LJ, Efferth T. Natural products targeting tumour angiogenesis. Br J Pharmacol 2025; 182:2094-2136. [PMID: 37680009 DOI: 10.1111/bph.16232] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
Tumour angiogenesis is the formation of new blood vessels to support the growth of a tumour. This process is critical for tumour progression and metastasis, making it an attractive approach to cancer therapy. Natural products derived from plants, animals or microorganisms exert anti-angiogenic properties and can be used to inhibit tumour growth and progression. In this review, we comprehensively report on the current status of natural products against tumour angiogenesis from four perspectives until March 2023: (1) the role of pro-angiogenic factors and antiangiogenic factors in tumour angiogenesis; (2) the development of anti-tumour angiogenesis therapy (monoclonal antibodies, VEGFR-targeted small molecules and fusion proteins); (3) the summary of anti-angiogenic natural agents, including polyphenols, polysaccharides, alkaloids, terpenoids, saponins and their mechanisms of action, and (4) the future perspectives of anti-angiogenic natural products (bioavailability improvement, testing of dosage and side effects, combination use and discovery of unique natural-based compounds). Our review aims to better understand the potential of natural products for drug development in inhibiting tumour angiogenesis and further aid the effective transition of these outcomes into clinical trials. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lara Johanna Friedrich
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
21
|
Liu L, Wu M, Chen Y, Cheng Y, Liu S, Zhang X, Xie Q, Cao L, Wei L, Fang Y, Jafri A, Sferra TJ, Shen A, Li L. Downregulating FGGY carbohydrate kinase domain containing promotes cell senescence by activating the p53/p21 signaling pathway in colorectal cancer. Int J Mol Med 2025; 55:81. [PMID: 40116125 PMCID: PMC11964412 DOI: 10.3892/ijmm.2025.5522] [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: 07/04/2024] [Accepted: 01/14/2025] [Indexed: 03/23/2025] Open
Abstract
Carbohydrate kinases serve an oncogenic role in several types of cancer; however, the function of FGGY carbohydrate kinase domain containing (FGGY) in colorectal cancer (CRC) remains unknown. The present study investigated the function and possible molecular mechanisms of FGGY in CRC. The results showed that elevated levels of FGGY mRNA and protein were observed in CRC tissues, and a higher expression of FGGY was associated with advanced N stage and reduced overall survival time in patients with CRC. Silencing FGGY inhibited the viability of CRC cells by inducing cell cycle arrest and promoting apoptosis in vitro, thereby attenuating tumor growth in a xenograft mouse model. FGGY knockdown also enriched the senescence‑associated heterochromatin foci (SAHF) pathway and p53 pathway, as further confirmed by enhancing senescence‑associated β‑galactosidase (SA‑β‑gal) activity, with increased levels of SAHF‑associated proteins HP1γ and trimethylation of H3K9 (H3k9me3) in CRC cells, as well as upregulation of p53 and its downstream protein p21. Furthermore, p53 knockout rescued FGGY knockdown‑mediated reductions in cell viability, SA‑β‑gal activity, and the levels of HP1γ and H3k9me3 in CRC cells. These findings indicated that FGGY could act as a newly identified potential oncogene in CRC, partially through regulating the p53/p21 signaling pathway and altering cell senescence.
Collapse
Affiliation(s)
- Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Department of Scientific Research, Affiliated Sanming Integrated Medicine Hospital of Fujian University of Traditional Chinese Medicine, Sanming, Fujian 365001, P.R. China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Sijia Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xinran Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Liujing Cao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yi Fang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Anjum Jafri
- Department of Genetics and Genome Sciences, Histology Core, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thomas J. Sferra
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Li Li
- Shengli Clinical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Health Management, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
22
|
Wang XR, Luan JX, Guo ZA. Mechanism of Astragaloside IV in Treatment of Renal Tubulointerstitial Fibrosis. Chin J Integr Med 2025; 31:474-480. [PMID: 38850482 DOI: 10.1007/s11655-024-3805-6] [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] [Accepted: 12/07/2023] [Indexed: 06/10/2024]
Abstract
Tubulointerstitial fibrosis (TIF) is one of the key indicators in evaluating the renal function of patients. Mild TIF can cause a vicious cycle of renal tubular glomerular injury and aggravate renal disease. Therefore, studying the mechanisms underlying TIF is essential to identify therapeutic targets, thereby protecting the renal function of patients with timely intervention. Astragaloside IV (AS-IV) is a Chinese medicine component that has been shown to inhibit the occurrence and progression of TIF via multiple pathways. Previous studies have reported that AS-IV protected against TIF by inhibiting inflammation, autophagy, endoplasmic reticulum stress, macrophages, and transforming growth factor-β1, which laid the foundation for the development of a new preventive and therapeutic option for TIF.
Collapse
Affiliation(s)
- Xin-Ru Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jing-Xiang Luan
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhao-An Guo
- Department of Nephrology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
23
|
Bai L, Liu X, Yuan Z, Xu G, Li X, Wan Z, Zhu M, Liang X, Li P, Lan Q, Yu H, Tang G, Huang M, Peng S, Lin J, Wang X, Luo Y, Wei G. Activation of IL-2/IL-2R pathway by Hedyotis diffusa polysaccharide improves immunotherapy in colorectal cancer. Int J Biol Macromol 2025; 306:141013. [PMID: 39954887 DOI: 10.1016/j.ijbiomac.2025.141013] [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: 09/02/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Colorectal cancer (CRC) is a prevalent and highly malignant tumor with a limited response to immune checkpoint inhibitor-based immunotherapy. There is an urgent need for novel immunomodulatory agents to enhance the immunotherapeutic response in CRC. Hedyotis diffusa, known for its immunomodulatory properties, has long been utilized as an adjunct in cancer treatment, positioning it as a potential source for discovering new tumor immunomodulators. In this study, we identified a polysaccharide derived from Hedyotis diffusa (HDP), comprising six monosaccharides: rhamnose, arabinose, galactose, glucose, xylose, and mannose. When combined with PD-1 and CTLA-4 inhibitors, HDP can boost systemic immunity in mice to enhance the effectiveness of immune checkpoint inhibitors in CRC therapy. HDP significantly increases the infiltration of CD4+ and CD8+ T cells into tumor microenvironment and upregulates the expression of key effector molecules derived from cytotoxic T cells. Mechanistic studies reveal that HDP activates the IL-2/IL-2R axis by upregulating IL-2 production and the expression of IL-2 receptor subunits, thereby promoting T cell proliferation. Collectively, this research introduces an innovative strategy to improve the efficacy of tumor immunotherapy by harnessing the immunomodulatory potential of polysaccharides. It also directs a roadmap for developing HDP as a promising immunomodulator for CRC treatment.
Collapse
Affiliation(s)
- Liangliang Bai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xiaoxia Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Ze Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Gaopo Xu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xuan Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Zhongxian Wan
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei 445000, China
| | - Mingxuan Zhu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaoxia Liang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Peisi Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Qiqian Lan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Huichuan Yu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Guannan Tang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Mingzhe Huang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Shaoyong Peng
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Jinxing Lin
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China
| | - Xiaolin Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Yanxin Luo
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Gang Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
24
|
Xue F, Xie L, Zhang X, Gao Y, Guo J, Liu X, Zhu H, Liu X. Simultaneous Determination of 14 Bioactive Components in Fangji Huangqi Tang by UHPLC-QqQ-MS Technique. Biomed Chromatogr 2025; 39:e70073. [PMID: 40174933 DOI: 10.1002/bmc.70073] [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: 12/23/2024] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
Fangji Huangqi Tang (FHT) is a traditional prescription frequently utilized in clinical practice, with a wide range of clinical applications and good therapeutic effects. Quality control of FHT is difficult because Chinese medicine compounds usually contain a vast array of components characterized by significant structural diversity. A quick and accurate method to determine the content of active constituents in FHT was essential, by which the purpose of quality control and efficacy assessment could be achieved. A method utilizing UHPLC-QqQ-MS technology in multiple reaction monitoring (MRM) mode was established to quantify 14 bioactive components in FHT simultaneously. These analytes included tetrandrine, fangchinoline, calycosin, calycosin-7-glucoside, medicarpin, formononetin, atractylenolide I, atractylenolide II, atractylenolide III, liquiritigenin, isoliquiritigenin, liquiritin, isoliquiritin, and glycyrrhizic acid. And to our knowledge, the content of calycosin, medicarpin, formononetin, and atractylenolide II in FHT was reported for the first time in this paper. The method was thoroughly validated for stable and reliable application regarding specificity, linearity, precision, stability, repeatability, and accuracy. The established method allowed the simultaneous determination of 14 bioactive components with diverse structures and trace amounts in FHT, ultimately achieving the quality control and assessment of FHT for its safe and appropriate clinical use.
Collapse
Affiliation(s)
- Fangfang Xue
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lintong Xie
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xia Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yifei Gao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jizhen Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xue Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
25
|
Ling Y, Liang X, Yan K, Zeng G, Zhu X, Jiang J, Lu S, Wang X, Zhou Y, Li Z, Mai W, Wang D, Chen J. Bimetallic Ca/Zn Nanoagonist Remould the Immunosuppressive Hepatocellular Carcinoma Microenvironment Following Incomplete Microwave Ablation via Pyroptosis and the STING Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500670. [PMID: 40305756 DOI: 10.1002/advs.202500670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/18/2025] [Indexed: 05/02/2025]
Abstract
During the treatment of solid tumors, local therapeutic approaches carry the risk of incomplete radical cure, which may lead to rapid tumor growth. Incomplete microwave ablation (iMWA) can induce tumors to exhibit highly invasive and uncontrollable growth, which is related to the immunosuppressive microenvironment. A multifunctional bimetallic Ca/Zn nanoagonist (PZH/Zn@CaNA) with a biomimetic liposome-modified surface to tumor tissues after iMWA is developed. In response to the acidic tumor microenvironment, the released traditional Chinese medicine preparation Pien Tze Huang (PZH) reduced protein expressions of the JAK2-STAT3 signaling pathway, thereby slowing down the proliferation and growth of hepatocellular carcinoma (HCC). Furthermore, the bimetallic ions Ca2⁺ and Zn2⁺ can cascade to enhance the killing effect of oxidative stress, generating substantial amounts of reactive oxygen species. This process induces pyroptosis and releases significant quantities of damage associated molecular patterns, thereby triggering immune activation mechanisms related to the STING pathway that reshape the immunosuppressive HCC microenvironment resulting from iMWA. This strategy markedly differs from previous chemoimmunotherapies, which not only effectively addressed the problem of conventional drugs showing heterogeneous distribution in tumor regions, but also verified the critical role played by PZH/Zn@CaNA in inhibiting iMWA-induced rapid tumor growth, regulating oxidative stress and remodeling the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Yuan Ling
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Xiayi Liang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Kangning Yan
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Guichun Zeng
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Xiaoqi Zhu
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Jinghang Jiang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Shaolong Lu
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Xiaobo Wang
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Yuying Zhou
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Zhaoshen Li
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Wei Mai
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| | - Duo Wang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Jie Chen
- Department of Hepatobiliary Surgery, Department of Medical Ultrasound, Department of Traditional Chinese Medicine, Guangxi Medical University Cancer Hospital, Guangxi Medical University. No. 71 Hedi Road, Nanning, Guangxi, 530021, China
| |
Collapse
|
26
|
Zhang W, Li R, Lu D, Wang X, Wang Q, Feng X, Qi S, Zhang X. Phospholipids and peroxisomes in ferroptosis: the therapeutic target of acupuncture regulating vascular cognitive impairment and dementia. Front Aging Neurosci 2025; 17:1512980. [PMID: 40365351 PMCID: PMC12070441 DOI: 10.3389/fnagi.2025.1512980] [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: 10/17/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Ferroptosis, since its conceptualization in 2012, has witnessed an exponential growth in research interest over recent years. It is regulated by various cellular metabolic pathways during chronic cerebral ischemia and hypoxia, including reactive oxygen species (ROS) generation, iron accumulation, abnormalities in glutathione metabolism, and disruptions in lipid and glucose metabolism. With the deepening and widespread research, ferroptosis has emerged as a critical pathway in the pathogenesis of vascular cognitive impairment and dementia (VCID). This unique cell death pathway caused by iron-dependent phospholipid peroxidation is strongly related to VICD. We examine the impact of phospholipid composition on neuronal susceptibility to ferroptosis, with a particular focus on the critical role of polyunsaturated fatty acids (PUFAs) in this process. Intriguingly, peroxisomes, as key regulators of lipid metabolism and oxidative stress, influence the susceptibility of neuronal cells to ferroptosis through the synthesis of plasmalogens and other lipid species. In this Review, we provide a critical analysis of the current molecular mechanisms and regulatory networks of acupuncture for ferroptosis, the potential functions of acupuncture in peroxisomal functions and phospholipid metabolism, and its neuroprotective effects in VCID, together with a potential for therapeutic targeting. As such, this highlights the theoretical basis for the application of acupuncture in VCID through multi-target regulation of ferroptosis. This review underscores the potential of acupuncture as a non-pharmacological therapeutic approach in VCID, offering new insights into its role in modulating ferroptosis and associated metabolic pathways for neuroprotection.
Collapse
Affiliation(s)
- Wenyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruiyu Li
- Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, China
| | - Donglei Lu
- Sports Training Academy of Tianjin University of Sport, Tianjin, China
| | - Xinliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuxuan Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuyang Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Sai Qi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuezhu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
27
|
Su Y, Chen R, Wang B, Wang T, Tao J, Diao Q, Jiang T, Zhao X. Erythrocyte membrane camouflaged celastrol and bilirubin self-assembly for rheumatoid arthritis immunotherapy based on STING inhibition and RONS clearance. J Nanobiotechnology 2025; 23:318. [PMID: 40287703 PMCID: PMC12032812 DOI: 10.1186/s12951-025-03389-9] [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: 02/26/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Activation of cGAS-STING signaling pathway and accumulation of reactive oxygen and nitrogen species (RONS) are important issues facing the treatment of rheumatoid arthritis (RA). Here, we report a biomimetic nano-Chinese medicine (HA-RM-Cel-BR) for RA immunotherapy based on STING inhibition of celastrol (Cel) and RONS clearance of bilirubin (BR). HA-RM-Cel-BR is constructed by the carrier-free self-assembly of active ingredients Cel and BR from traditional Chinese medicine, and then camouflaged by hyaluronic acid (HA)-modified red blood cell membranes (RM). HA-RM-Cel-BR prolongs circulation time through RM camouflage, targets inflamed joints by HA modification, and remodels the joint immune microenvironment by STING inhibition and RONS clearance. More importantly, HA-RM-Cel-BR shows excellent therapeutic effects on RA rat model, and significantly reduces hepatotoxicity associated with Cel. Our work provides a new strategy for RA immunotherapy with traditional Chinese medicine ingredients.
Collapse
Affiliation(s)
- Yanguo Su
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Rui Chen
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Bingjie Wang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Teng Wang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Jiaojiao Tao
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Qijie Diao
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Tianze Jiang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Xia Zhao
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
28
|
Zhang BT, Song X, Cho CS(W, Wong CK, Wang D. Deciphering Anticancer Mechanisms of Calycosin in Lung Adenocarcinoma Through Multi-Omics: Targeting SMAD3-Mediated NOTCH Signaling in the Tumor Microenvironment. Cancers (Basel) 2025; 17:1455. [PMID: 40361382 PMCID: PMC12071042 DOI: 10.3390/cancers17091455] [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: 03/23/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE Lung adenocarcinoma (LUAD) remains a leading cause of cancer-related mortality, particularly in advanced stages. This study investigates the anticancer mechanisms of calycosin, an isoflavonoid derived from Astragalus membranaceus, in LUAD. METHODS Using integrative approaches including bulk and single-cell RNA sequencing, network pharmacology, and molecular docking, we identified SMAD3 as a critical biomarker associated with LUAD staging and prognosis. RESULTS Calycosin targets SMAD3, modulating the NOTCH signaling pathway in monocytes/macrophages to suppress tumor growth, invasion, and immune evasion. Enrichment analyses revealed significant involvement of NOTCH signaling components in SMAD3-correlated genes, particularly in advanced-stage LUAD. Single-cell RNA sequencing further demonstrated NOTCH pathway enrichment in tumor-associated monocytes/macrophages. Additionally, KMT2A was identified as a key transcriptional regulator in these cells. CONCLUSIONS These findings highlight the potential effects of calycosin and provide novel insights into targeting the tumor-immune microenvironment in LUAD.
Collapse
Affiliation(s)
- Bi-Tian Zhang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China;
| | - Xiaoyu Song
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China;
| | | | - Chun-Kwok Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China;
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dongjie Wang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China;
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
29
|
Wu L, Wu J, Wang X, Xu Y, Lin Z, Chen J, Wu X. Natural product-based nanotechnological formulations for colorectal cancer treatment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04175-y. [PMID: 40274619 DOI: 10.1007/s00210-025-04175-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
Colorectal cancer is one of the most common malignancies affecting the gastrointestinal tract. A silent onset often marks it and carries a poor prognosis. Studies have shown that natural products can suppress the growth of colorectal cancer and exert therapeutic effects at the molecular level. However, unfavorable physicochemical properties frequently hinder their clinical application, such as low solubility, limited bioavailability, short half-life, and rapid systemic clearance. As scientific and technological progress continues, increasing attention has been directed toward nanotechnology-based approaches. Techniques involving nanoparticles, liposomes, and micelles are being explored to improve drug delivery. These advancements provide a promising foundation for overcoming the limitations associated with natural products. This review systematically examines the application of nano-formulations for natural ingredients to offer meaningful insights into their. potential use in treating colorectal cancer.
Collapse
Affiliation(s)
- Lanfang Wu
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China
| | - Jiali Wu
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China
| | - Xinyu Wang
- , Shanghai Wei Er Lab, Shanghai, 201707, China
| | - Youfa Xu
- , Shanghai Wei Er Lab, Shanghai, 201707, China
| | - Zhizhe Lin
- , Shanghai Wei Er Lab, Shanghai, 201707, China
| | - Jianming Chen
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China.
- , Shanghai Wei Er Lab, Shanghai, 201707, China.
| | - Xin Wu
- Fujian University of Traditional Chinese Medicine, No. 1 Qiuyang Road, Fuzhou, 350122, China.
- , Shanghai Wei Er Lab, Shanghai, 201707, China.
| |
Collapse
|
30
|
Zhu L, Huang R, Feng JR, Zhang M, Huang XJ, Chen Z, Wang W, Chen Y. Shexiang Tongxin Dropping Pills attenuate ischemic microvascular dysfunction via suppressing P66Shc-mediated mitochondrial respiration deficits. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119664. [PMID: 40154895 DOI: 10.1016/j.jep.2025.119664] [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: 02/28/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ischemic stroke (IS) disrupts mitochondrial energy metabolism, leading to cerebral microvascular dysfunction (CMD). Shexiang Tongxin Dropping Pills (STDP) is a traditional Chinese medicinal formulation that has been clinically used for treating microcirculatory dysfunction. We have previously reported its ability to improve cerebral microcirculatory abnormalities. Nevertheless, the protective effects of STDP on cerebral microvascular mitochondria in the context of energy metabolism repair remain underinvestigated. AIM OF THE STUDY This study aims to investigate the potential mechanisms by which STDP ameliorates IS-induced CMD through the restoration of mitochondrial function. MATERIALS AND METHODS An ischemic stroke/reperfusion model was established by occluding and subsequently reperfusing the middle cerebral artery (MCAO/R) in C57BL/6 J mice. Laser speckle contrast imaging, Y-maze, rotarod tests and TTC staining were employed to evaluate the anti-ischemic stroke effects of STDP. Histological examination of cell adhesion proteins (ICAM 1, VCAM 1) and tight junction proteins (VE-cadherin, occludin) was conducted to assess the effects of STDP on the cerebral microvascular endothelium. In vitro, a bEnd.3 cell model was established through oxygen-glucose deprivation followed by reoxygenation (OGD/R). The cytoprotective capability of STDP was assessed by quantifying endothelial permeability, reactive oxygen species (ROS) levels, and cell viability. Mendelian randomization (MR) analysis and bioinformatic studies were performed to elucidate the causal associations between mitochondrial biological function and IS. Mitochondrial membrane potential (MMP) was assessed using a tetramethylrhodamine ethyl ester perchlorate fluorescent probe, while ATP production was quantified using a commercially available assay kit. Mitochondrial respiration was evaluated by measuring the oxygen consumption rate (OCR). Finally, the verification of important targets in mouse brain slices and bEnd.3 cells was conducted through immunoblotting and immunofluorescence. RESULTS STDP significantly restored cerebral blood flow and neurological function, and reduced infarct volume in MCAO/R mice. Furthermore, STDP markedly alleviated inflammation and hyperpermeability of the cerebral microvascular endothelium in MCAO/R mice, as evidenced by the suppression of ICAM-1 and VCAM-1 expression, along with the upregulation of VE-cadherin and occludin protein levels. Moreover, STDP not only mitigated hyperpermeability and excessive production of ROS induced by OGD/R in bEnd.3 cells but also enhanced the protective effects of the ROS scavenger N-acetylcysteine on bEnd.3 cells. Results of MR analysis and bioinformation studies demonstrated that the disruption of mitochondrial respiration is a critical pathogenic factor in IS-induced CMD. Our data confirmed that STDP effectively restored MMP and ATP production in OGD/R-treated bEnd.3 cells. Furthermore, STDP significantly enhanced basal respiration, maximal OCR, and spare respiratory capacity in bEnd.3 cells compared to the OGD/R group. Mechanistically, STDP markedly increased endothelial cystathionine γ-lyase (CSE)-mediated hydrogen sulfide (H2S) production and S-sulfhydration of P66shc, resulting in reduced protein expression and phosphorylation levels of P66Shc. This inhibition prevented its translocation into mitochondria, thereby restoring mitochondrial respiration. CONCLUSION STDP facilitated CSE expression and promoted H2S production, contributing to the inactivation of P66shc by suppressing its expression and increasing its sulfhydration. This process impeded P66Shc translocation to mitochondria, subsequently restoring mitochondrial respiration and alleviating IS-induced cerebral microvascular endothelial dysfunction.
Collapse
Affiliation(s)
- Li Zhu
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China; School of Pharmaceutics, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China
| | - Ru Huang
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China; School of Pharmaceutics, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China
| | - Jing-Rui Feng
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China; School of Pharmaceutics, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China
| | - Miao Zhang
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China; School of Pharmaceutics, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China
| | - Xiao-Jie Huang
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China; School of Pharmaceutics, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China
| | - Zeyu Chen
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China
| | - Wei Wang
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China; School of Pharmaceutics, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China.
| | - Yang Chen
- Science and Technology Innovation Center, NMPA Key Laboratory for Research of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China; Chinese Medicine Guangdong Laboratory, Zhuhai, 519031, China.
| |
Collapse
|
31
|
An D, Jiang X, Yang Y. Sesamin Exerts Anti-Tumor Activity in Nasopharyngeal Carcinoma Through Inducing Autophagy and Reactive Oxygen Species Production. FRONT BIOSCI-LANDMRK 2025; 30:26038. [PMID: 40302321 DOI: 10.31083/fbl26038] [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: 08/07/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 05/02/2025]
Abstract
BACKGROUND Sesamin can suppress many cancers, but its effect on nasopharyngeal carcinoma (NPC) is unclear. Herein, we set out to pinpoint the possible changes in NPC due to Sesamin. METHODS The biological function of NPC cells exposed to Sesamin/N-acetyl-L-cysteine (NAC)/3-Methyladenine (3-MA) was detected, followed by evaluation of reactive oxygen species (ROS) production (dichlorodihydrofluorescein diacetate staining) and mitochondrial membrane potential (MMP) (flow cytometry). Proteins pertinent to apoptosis (cleaved caspase-3, cleaved poly (ADP-ribose) polymerase 1 (PARP1)), cell cycle (Cyclin B1), and autophagy (microtubule-associated protein light chain 3 (LC3)-I, LC3-II, Beclin-1, P62) were quantified by Western blot. After the xenografted tumor model in mice was established, the tumor volume and weight were recorded, and Ki-67 and cleaved caspase-3 levels were determined by immunohistochemical analysis. RESULTS Sesamin inhibited viability, proliferation, cell cycle progression and migration, induced apoptosis, increased ROS production, and decreased MMP in NPC cells. Sesamin elevated cleaved caspase-3/caspase-3, cleaved PARP1/PARP1, and Beclin-1 expressions as well as LC3-II/LC3-I ratio, while diminishing Cyclin B1 and P62 levels. NAC and 3-MA abrogated Sesamin-induced changes as above in NPC cells. Sesamin inhibited the increase of the xenografted tumor volume and weight, down-regulated Ki-67, and up-regulated cleaved caspase-3 in xenografted tumors. CONCLUSION Sesamin exerts anti-tumor activity in NPC, as demonstrated by attenuated tumor proliferation and xenografted tumor volume and weight, as well as induced apoptosis in tumor tissues, consequent upon the promotion of autophagy and reactive oxygen species production.
Collapse
Affiliation(s)
- Deqiang An
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Xianyao Jiang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Yucheng Yang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| |
Collapse
|
32
|
Manavi MA, Mohammad Jafari R, Shafaroodi H, Dehpour AR. The Keap1/Nrf2/ARE/HO-1 axis in epilepsy: Crosstalk between oxidative stress and neuroinflammation. Int Immunopharmacol 2025; 153:114304. [PMID: 40117806 DOI: 10.1016/j.intimp.2025.114304] [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: 10/08/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Epilepsy is a complex neurological disorder characterized by recurrent seizures, which are driven by multifaceted pathophysiological mechanisms, including oxidative stress and neuroinflammation. Despite advancements in anti-seizure medications (ASMs), a significant proportion of patients remain resistant to treatment, highlighting the need for novel therapeutic strategies. This review focuses on the Kelch-like ECH-associated protein 1 (Keap1) / Nuclear factor erythroid 2-related factor 2 (Nrf2) / Antioxidant Response Element (ARE) / Heme Oxygenase-1 (HO-1) axis as a promising target for neuroprotection in epilepsy. We explored the intricate interactions between Keap1 and Nrf2 under homeostatic conditions and how oxidative stress disrupts this balance, triggering Nrf2 activation. This review details the subsequent process of Nrf2 nuclear translocation, its binding to AREs, and the induction of cytoprotective gene expression, which collectively orchestrate a robust cellular defense response. Special emphasis is placed on HO-1, a key effector of Nrf2-mediated neuroprotection, highlighting its enzymatic function and protective mechanisms, including antioxidant, anti-inflammatory, and anti-apoptotic effects. Additionally, the review examines HO-1's role in mitigating seizure-induced neuronal damage. However, challenges remain, including variability in therapeutic responses, gaps in long-term clinical validation, and the need for standardized protocols. Future research should focus on biomarkers for personalized treatment, advanced imaging, and genetic tools to explore the Keap1/Nrf2/ARE/HO-1 axis in greater depth. Future studies should focus on overcoming the challenges of translating preclinical findings into clinical applications and exploring the long-term effects of targeting this pathway in epilepsy treatment.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran university of medical sciences, Tehran, Iran; Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Razieh Mohammad Jafari
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran
| | - Hamed Shafaroodi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran.
| |
Collapse
|
33
|
Wu R, Wu S, Zeng H, Ye Q, Qian F, Ma C, Zhang W, Ye J. UPLC-QTOF-MS-Based Quantification and Antiplatelet Activity Evaluation of Herb Pair Interactions Between Red Ginseng and Trogopterus Feces. Chem Biodivers 2025:e202500770. [PMID: 40265573 DOI: 10.1002/cbdv.202500770] [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: 03/06/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/24/2025]
Abstract
Red ginseng (RG) and Trogopterus Feces (TF) are one of the drug pairs in "nineteen medicaments of mutual antagonism" and not recommended for drug compatibility in Chinese Pharmacopoeia. In addition, RG functions to "tonify Qi" and "nourish the blood," while TF can "promote blood circulation" and "relieve pain." Therefore, the two drugs are often combined to treat blood stasis syndrome in the clinical practice of traditional Chinese medicine (TCM). Nevertheless, the combined application of RG and TF in TCM formulas has attracted much attention but still lacks in-depth research. To bridge this gap, our investigation employed UPLC-QTOF-MS technology coupled with in vitro pharmacological models to systematically examine both compositional alterations and functional consequences of RG-TF compatibility. Key findings revealed that co-decoction of these two herbal medicines induced a significant reduction in the content of bioactive ginsenosides derived from RG, while concurrently diminishing the antiplatelet aggregation efficacy observed in the single-herb decoction of RG. This chemical-pharmacological correlation provides compelling evidence for the incompatibility of RG with TF and elucidates the biochemical basis of their mutual antagonism documented in classical TCM literature.
Collapse
Affiliation(s)
- Ruijun Wu
- School of Pharmacy, Naval Medical University, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shiyu Wu
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huawu Zeng
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Qing Ye
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Fei Qian
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chi Ma
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weidong Zhang
- School of Pharmacy, Naval Medical University, Shanghai, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ji Ye
- School of Pharmacy, Naval Medical University, Shanghai, China
| |
Collapse
|
34
|
Wang H, Guo J, Zhang Y, Fu Z, Yao Y. Closed-loop rehabilitation of upper-limb dyskinesia after stroke: from natural motion to neuronal microfluidics. J Neuroeng Rehabil 2025; 22:87. [PMID: 40253334 PMCID: PMC12008995 DOI: 10.1186/s12984-025-01617-9] [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: 12/04/2024] [Accepted: 03/27/2025] [Indexed: 04/21/2025] Open
Abstract
This review proposes an innovative closed-loop rehabilitation strategy that integrates multiple subdomains of stroke science to address the global challenge of upper-limb dyskinesia post-stroke. Despite advancements in neural remodeling and rehabilitation research, the compartmentalization of subdomains has limited the effectiveness of current rehabilitation strategies. Our approach unites key areas-including the post-stroke brain, upper-limb rehabilitation robotics, motion sensing, metrics, neural microfluidics, and neuroelectronics-into a cohesive framework designed to enhance upper-limb motion rehabilitation outcomes. By leveraging cutting-edge technologies such as lightweight rehabilitation robotics, advanced motion sensing, and neural microfluidic models, this strategy enables real-time monitoring, adaptive interventions, and personalized rehabilitation plans. Furthermore, we explore the potential of closed-loop systems to drive neural plasticity and functional recovery, offering a transformative perspective on stroke rehabilitation. Finally, we discuss future directions, emphasizing the integration of emerging technologies and interdisciplinary collaboration to advance the field. This review highlights the promise of closed-loop strategies in achieving unprecedented integration of subdomains and improving post-stroke upper-limb rehabilitation outcomes.
Collapse
Affiliation(s)
- Honggang Wang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Junlong Guo
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Yangqi Zhang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Ze Fu
- Institute of Biological and Medical Technology, Harbin Institute of Technology (Weihai), Weihai, 264200, China
| | - Yufeng Yao
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
35
|
Li L, Chen M, Reis RL, Kundu SC, Xiao B, Shi X. Advancements of nanoscale drug formulations for combination treatment of colorectal cancer. Int J Pharm 2025; 674:125508. [PMID: 40132771 DOI: 10.1016/j.ijpharm.2025.125508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Combination chemotherapy is widely utilized in treating colorectal cancer (CRC), particularly for patients who are ineligible for surgery or those with metastatic CRC (mCRC). While this therapeutic method has demonstrated efficacy in managing CRC and mCRC, its broader clinical application is limited due to the unique physical properties, mechanisms of action, and pharmacokinetics of different chemotherapeutic drugs. Consequently, achieving satisfactory treatment outcomes proves to be challenging. Nanotechnology has given rise to innovative drug systems that are precise, controllable, and highly efficient in drug delivery. These nanoscale drug delivery systems can integrate the advantageous aspects of various therapeutic modalities, including chemotherapy, gene therapy, and immunotherapy. This review aims to explain the application of nano-drug delivery system in the treatment of colorectal cancer. Through its unique physical/chemical properties and biological functions, it can solve the limitations of traditional therapy and achieve more accurate, efficient and safe treatment. The advantages/disadvantages, physical and chemical characteristics of various drug delivery systems are described in detail, and suggestions on selecting reasonable NDDSs according to different drug combination methods are given to achieve the best therapeutic effect. This review paper presents an exhaustive summary of the diverse range of drugs utilized in chemotherapy, in addition to outlining strategies for effectively integrating chemotherapy with other treatment modalities. Furthermore, it delves into the principle of selecting carriers for various drug combinations.
Collapse
Affiliation(s)
- Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Maohua Chen
- Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Bo Xiao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
36
|
Singh S, Subudhi M, Moorthy A V, Suresh A, Sharma P. Ursolic acid induces apoptosis and disrupts host-parasite interactions in Theileria annulata-infected cells. Int J Parasitol Drugs Drug Resist 2025; 28:100593. [PMID: 40273613 PMCID: PMC12051632 DOI: 10.1016/j.ijpddr.2025.100593] [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: 12/02/2024] [Revised: 03/21/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
This study explores the anti-proliferative and anti-parasitic properties of ursolic acid (UA) in Theileria annulata-infected bovine (TA) cells. Dose-response analyses determined an IC50 value of approximately 5 μg/mL for UA, demonstrating selective cytotoxicity toward infected cells with minimal impact on healthy cells. UA treatment induced pronounced morphological alterations and apoptosis in TA cells, as evidenced by light microscopy and a time-dependent increase in cell mortality. Notably, UA exhibited consistent efficacy against both buparvaquone (BPQ)-sensitive and BPQ-resistant TA cell lines, highlighting its broad-spectrum anti-parasitic potential. Mechanistic investigations revealed that UA triggers DNA damage, elevates reactive oxygen species (ROS) levels, disrupts mitochondrial function, and induces sub-G1 phase arrest, culminating in apoptosis primarily via the intrinsic pathway. Mass spectrometry-based proteomic profiling identified significant perturbations in host cell pathways, including DNA repair mechanisms, cell cycle regulation, and signaling networks, alongside direct interference with parasite metabolic processes. Western blot analysis further confirmed UA-mediated modulation of host cell signaling pathways and chromatin organization. Given the rising incidence of drug-resistant T. annulata strains, the development of novel therapeutic strategies is imperative. These findings highlight UA's multifaceted mechanism of action, targeting both parasitic and host cellular processes, and position it as a promising candidate for the treatment of bovine theileriosis.
Collapse
Affiliation(s)
- Sakshi Singh
- National Institute of Animal Biotechnology, Hyderabad, 500032, India; Graduate Studies, Regional Centre for Biotechnology (RCB), Faridabad, 121001, India
| | - Madhusmita Subudhi
- National Institute of Animal Biotechnology, Hyderabad, 500032, India; Graduate Studies, Regional Centre for Biotechnology (RCB), Faridabad, 121001, India
| | - Vengatachala Moorthy A
- National Institute of Animal Biotechnology, Hyderabad, 500032, India; Graduate Studies, Regional Centre for Biotechnology (RCB), Faridabad, 121001, India
| | - Akash Suresh
- National Institute of Animal Biotechnology, Hyderabad, 500032, India
| | - Paresh Sharma
- National Institute of Animal Biotechnology, Hyderabad, 500032, India.
| |
Collapse
|
37
|
Akabane M, Kawashima J, Woldesenbetm S, Macedo AB, Altaf A, Aucejo F, Popescu I, Kitago M, Poultsides GA, Sasaki K, Imaoka Y, Ruzzenente A, Endo I, Pawlik TM. Enhancing outcome prediction in patients with colorectal liver metastases undergoing hepatectomy: the synergistic impact of FIB-4 index and tumor burden score across KRAS profiles. HPB (Oxford) 2025:S1365-182X(25)00546-5. [PMID: 40287297 DOI: 10.1016/j.hpb.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The prognostic value of Fibrosis-4 (FIB-4) index, concerning KRAS status (wild-type [wtKRAS] vs. mutated [mutKRAS]) remains unclear in post-hepatectomy colorectal liver metastases (CRLM). We evaluated the combined impact of FIB-4 and Tumor Burden Score (TBS) on overall survival (OS)/recurrence-free survival (RFS), stratified by KRAS status. METHODS CRLM patients undergoing hepatectomy (2000-2020) were analyzed, grouped by TBS/FIB-4. RESULTS Among 828 patients, 196 had high FIB-4. High TBS had worse 5-year OS (P < 0.001). In wtKRAS, high TBS correlated with worse OS (P < 0.001), but not in mutKRAS. High FIB-4 correlated with worse OS (P = 0.01). Sub-stratification showed no OS difference by FIB-4 in wtKRAS, but a difference in mutKRAS (P = 0.03). Multivariable analysis identified mutKRAS (HR: 1.90), high TBS (HR: 1.62), and FIB-4 (HR: 1.15) as mortality risk factors. The TBS-FIB-4-KRAS index had highest predictive accuracy. For RFS, TBS and FIB-4 independently stratified outcomes. High TBS was associated with worse RFS in wtKRAS (P < 0.001) but not in mutKRAS. High FIB-4 decreased RFS in mutKRAS (P = 0.001) but not in wtKRAS. FIB-4 was associated with a 10% increased recurrence risk. CONCLUSION TBS and FIB-4, alongside KRAS status, should be considered to improve outcome predictions.
Collapse
Affiliation(s)
- Miho Akabane
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Jun Kawashima
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Selamawit Woldesenbetm
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Amanda B Macedo
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Abdullah Altaf
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Federico Aucejo
- Department of General Surgery, Cleveland Clinic Foundation, OH, USA
| | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | | | - Kazunari Sasaki
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Yuki Imaoka
- Department of Surgery, Stanford University, Stanford, CA, USA
| | | | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University, Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
38
|
Teske K, Erickson NA, Huck A, Dzamukova M, Fulde M, Heinbokel T, Horst D, Klymiuk N, Pastille E, Mekes-Adamczyk A, Löhning M, Gruber AD, Glauben R, Mundhenk L. The impact of disease and species differences on the intestinal CLCA4 gene expression. J Mol Med (Berl) 2025:10.1007/s00109-025-02538-9. [PMID: 40220130 DOI: 10.1007/s00109-025-02538-9] [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: 10/21/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025]
Abstract
The human chloride channel regulator, calcium-activated (CLCA) 4 is discussed as a driver of epithelial-to-mesenchymal transition as well as a biomarker for colorectal cancer (CRC) and ulcerative colitis. In contrast to humans, the Clca4 gene is duplicated in the mouse, a common model species to study gene functions. However, the relevance of the functional murine Clca4 variants in healthy and diseased intestine is largely unknown. Here, we characterized the spatiotemporal expression patterns of the murine Clca4a and Clca4b genes in the healthy intestinal tract as well as in dextran sulfate sodium (DSS)-induced colitis and colitis-associated colon cancer (CAC) mouse model using RT-qPCR and in situ-hybridization. Similarly, we analyzed expression of the human CLCA4 in healthy, inflamed and cancerous intestinal tracts at single cell level. Murine Clca4a and -4b but not the human CLCA4 were detected in small intestine enterocytes of the respective species. Conversely, healthy colonocytes expressed the human CLCA4 and its murine ortholog Clca4a but not the murine Clca4b. Under inflammatory conditions, de novo expression of Clca4b was observed with both murine homologs abundantly expressed in enterocytes adjacent to ulcerations. Neoplastic colonocytes expressed none or only minimal amounts of the CLCA4 homologs both in humans and mice, whereas adjacent non-neoplastic colonocytes strongly up-regulated the human or both murine homologs, respectively. Our results suggest marked species- and homolog-specific differences in the expression patterns of the three CLCA4 homologs. Moreover, all three seem to play a role in reactive, non-neoplastic colonocytes adjacent to ulcerated and neoplastic lesions. KEY MESSAGES: Human CLCA4 and murine Clca4a, but not Clca4b, are expressed in healthy colonocytes. Inflammation leads to a de novo expression of the murine Clca4b in colonocytes. Human and murine CLCA4 homologs are absent from neoplastic enterocytes. Human and murine CLCA4s are highly expressed in tumor-adjacent, reactive colonocytes.
Collapse
Affiliation(s)
- K Teske
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - N A Erickson
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- MF 3 - Animal Experimental Research and 3r - Methods Development, Research Infrastructure and Information Technology, Robert Koch Institute, Berlin, Germany
| | - A Huck
- Medical Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - University Medicine Berlin, Berlin, Germany
| | - M Dzamukova
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center Berlin, a Leibniz Institute, Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - M Fulde
- Center of Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - T Heinbokel
- Institute of Pathology, Charité - University Medicine Berlin, Berlin, Germany
| | - D Horst
- Institute of Pathology, Charité - University Medicine Berlin, Berlin, Germany
| | - N Klymiuk
- Large Animal Models in Cardiovascular Research, Internal Medical Department I, Technical University of Munich, Munich, Germany
- Center for Innovative Medical Models, Ludwig-Maximilians-University Munich, Munich, Germany
| | - E Pastille
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - A Mekes-Adamczyk
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - M Löhning
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center Berlin, a Leibniz Institute, Berlin, Germany
- Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - A D Gruber
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - R Glauben
- Medical Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - University Medicine Berlin, Berlin, Germany
| | - L Mundhenk
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
39
|
Hoseinpour F, Hashemnia M, Cheraghi H, Salari Asl MM, Zare F, Zanjani IA. Protective effect of the hydroethanolic extract of camelthorn (Alhagi maurorum) on benign prostatic hyperplasia induced by testosterone in rats. BMC Complement Med Ther 2025; 25:136. [PMID: 40221684 PMCID: PMC11992843 DOI: 10.1186/s12906-025-04862-6] [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: 09/05/2024] [Accepted: 03/14/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND One of the Iranian medicinal plants is Alhagi maurorum, which belongs to the Fabaceae family. The plant is used to treat different conditions, such as aphthous ulcers, cardiac pains, hemorrhoids, kidney stones, dysuria, etc. Given that A. maurorum is characterized by its richness in flavonoids and phenolic compounds, and it is used to treat urinary tract disorders, this study aimed to investigate the protective effects of its hydroethanolic extract in a rat model of benign prostatic hyperplasia (BPH). METHODS After preparing the hydroethanolic extract, phytochemical analysis, including GC-MS, was conducted. Adult male Wistar rats (n:35) were randomly divided into five groups (n:7): A sham surgery was conducted on the first group, while the other four groups underwent castration through the scrotal route. Seven days after the surgery, benign prostatic hyperplasia (BPH) was induced in all groups except the first one, using a subcutaneous injection of testosterone propionate at a dosage of 10 mg/kg/day. The treatment duration lasted 28 days, during which the animals received oral treatments as follows: 1. Sham control: normal saline, 2. Positive control (BPH group): normal saline, 3. Comparative control: finasteride at 5 mg/kg/day, 4. T1 group: A. maurorum extract at 200 mg/kg/day, and 5. T2 group: A. maurorum extract at 400 mg/kg/day. At the end of the experiment, following an overnight fast and after administering anesthesia with ketamine and xylazine, blood was collected through cardiac puncture, and sera were harvested for hormone assay. Finally after euthanizing the rats, the ventral prostatic lobes were dissected for biochemical, histopathological, and gene expression analyses. RESULTS GC-MS analysis showed the presence of nine components. A. maurorum extract and/or Finasteride led to a significant reduction of the prostate weight and prostatic index, serum, and prostatic levels of testosterone. These compounds led to the downregulation of 5-α reductase and androgen receptor genes expression, boosted total antioxidant capacity, and declined prostatic malondialdehyde levels. Intervention using the extract, comparable to Finasteride, led to BPH-induced histopathological enhancements in the prostate. CONCLUSION Treatment using A. maurorum extract resulted in significant protection of the prostate against BPH, which is attributable to its antioxidant and androgen-modulating characteristics.
Collapse
Affiliation(s)
- Fatemeh Hoseinpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah-Sheikh-E- Tousi Blvd, Kermanshah, 6715685414, Iran.
| | - Mohammad Hashemnia
- Department of Pathobiology, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Hadi Cheraghi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Mohammad Mohsen Salari Asl
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah-Sheikh-E- Tousi Blvd, Kermanshah, 6715685414, Iran
| | - Farshad Zare
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah-Sheikh-E- Tousi Blvd, Kermanshah, 6715685414, Iran
| | - Iman Ahmadi Zanjani
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah-Sheikh-E- Tousi Blvd, Kermanshah, 6715685414, Iran
| |
Collapse
|
40
|
Nobari S, Shojaeian A, Jalilian FA, Kalvandi G, Amini R. Modulation of the tumor microenvironment by zerumbone and 5-fluorouracil in colorectal cancer by target in cancer-associated fibroblasts. Discov Oncol 2025; 16:505. [PMID: 40208502 PMCID: PMC11985878 DOI: 10.1007/s12672-025-02253-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND This research aimed at the evaluation of the incremental effect(s) of combination therapy on colorectal cancer-associated fibroblasts (CAFs) in vivo and in vitro models of CRC. Drug resistance decreases the effectiveness of treatment for colorectal cancer (CRC) patients. CAFs, the main cells in the tumor stroma, contribute essentially to cancer drug resistance. Zerumbone (ZER) has been used for its anti-tumorigenic feature in various cancers. Also, 5-fluorouracil (5-FU) resistance is widely reported in advanced CRC patients. The possible impacts of combination therapy (or polytherapy is therapy that uses more than one medication or modality) with the use of herbs combined with chemical medicines may lead to improvement in patients' responses. METHODS According to the research design, the influence of 5-FU and ZER on the viability of the CT-26 cell line was examined through the MTT assay, and the size and dimension of the tumors were measured after 21 days of therapy in CRC BALB/c mice, and then the CAF cells were separated from the tissues of the tumors. Genes' mRNA expression and also the level of the proteins of the genes of interest (Survivin, Vimentin, & β -catenin) were evaluated using qRT-PCR and Western blotting on CT-26 and mouse-isolated CAF cells. RESULTS AND CONCLUSIONS We showed that ZER and 5-FU significantly increased our gene expression in the CT-26 cell line and isolated CAFs in the level of protein and mRNA in comparison to the controls (ZER-treated, untreated, & 5-FU-treated). Moreover, evaluating the size of the tumor in ZER and 5-FU-treated groups demonstrated a remarkable decline, which exhibited higher prominence in the group of combination therapy. A combination of natural and chemical drugs can largely enhance the effectiveness of cancer therapy.
Collapse
Affiliation(s)
- Sima Nobari
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Farid Azizi Jalilian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gholamreza Kalvandi
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pediatrics, School of Medicine, Hamadan University of Medical Science, Hamadan, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
41
|
Lin JG, Sun YW, Wu WL, Jiang WP, Zhung FY, Huang GJ. Multi-Target Protective Effects of Sanghuangporus sanghuang Against 5-Fluorouracil-Induced Intestinal Injury Through Suppression of Inflammation, Oxidative Stress, Epitheli-Al-Mesenchymal Transition, and Tight Junction. Int J Mol Sci 2025; 26:3444. [PMID: 40244381 PMCID: PMC11989720 DOI: 10.3390/ijms26073444] [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: 02/18/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Sanghuang (Sanghuangporus sanghuang, SS) is a medicinal fungus with multiple pharmacological effects, including antioxidant, anti-inflammatory, immune-boosting, and anti-cancer activities. 5-fluorouracil (5-FU) is a commonly used chemotherapeutic agent for the treatment of colorectal cancer. It primarily exerts its antitumor effect by inhibiting DNA and RNA synthesis, leading to cell apoptosis. However, it frequently induces adverse effects These issues limit the clinical application of 5-FU. This research aims to determine the potential of SS as a therapeutic agent in reducing 5-FU-induced intestinal mucositis in a mouse model. The results indicated that 5-FU administration significantly increased diarrhea severity, reduced colon length, caused small intestinal villus atrophy, disrupted intestinal architecture, led to insufficient crypt cell proliferation, and resulted in weight loss. It also significantly upregulated inflammatory responses, apoptosis, oxidative stress, and epithelial-mesenchymal transition (EMT) pathways, and disrupted the integrity of intestinal mucosal tight junction, while elevating pro-inflammatory cytokines and reducing antioxidant capacity. However, SS significantly ameliorating alleviating the adverse impacts of the chemotherapeutic agent on the intestinal mucosa. In conclusion, this investigation provides the first evidence of the protective effects of SS on 5-FU-induced mucositis. These findings suggest SS as a potential therapeutic application, offering a promising strategy for reducing the adverse effects of 5-FU chemotherapy and improving the treatment and quality of life for colorectal cancer patients.
Collapse
Affiliation(s)
- Jaung-Geng Lin
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan; (J.-G.L.); (W.-L.W.)
- Chinese Medicine Research Center, China Medical University, Taichung 404, Taiwan
| | - Yu-Wen Sun
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| | - Wen-Liang Wu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan; (J.-G.L.); (W.-L.W.)
| | - Wen-Ping Jiang
- Department of Pharmacy, China Medical University, Taichung 404, Taiwan;
| | - Fang-Yu Zhung
- Department of Nutrition, China Medical University, Taichung 404, Taiwan
| | - Guan-Jhong Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Department of Food Nutrition and Healthy Biotechnology, Asia University, Taichung 413, Taiwan
| |
Collapse
|
42
|
Heckmann M, Sadova N, Sandner G, Neuhauser C, Blank-Landeshammer B, Schwarzinger B, König A, Liang M, Spitzer M, Weghuber J, Stadlbauer V. Herbal extract fermented with inherent microbiota improves intestinal health by exerting antioxidant and anti-inflammatory effects in vitro and in vivo. J Anim Sci Biotechnol 2025; 16:52. [PMID: 40188119 PMCID: PMC11972464 DOI: 10.1186/s40104-025-01178-w] [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: 11/22/2024] [Accepted: 02/13/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Maintaining intestinal health is crucial for the overall well-being and productivity of livestock, as it impacts nutrient absorption, immune function, and disease resistance. Oxidative stress and inflammation are key threats to intestinal integrity. This study explored the antioxidant, anti-inflammatory, and barrier-strengthening properties of a fermented plant macerate (FPM) derived from 45 local herbs, using a specifically developed fermentation process utilizing the plants' inherent microbiota to enhance bioactivity and sustainability. RESULTS In vitro experiments with IPEC-J2 cells showed that FPM significantly reduced intracellular reactive oxygen species (ROS) levels, improved barrier integrity, and enhanced cell migration under stress. Similar antioxidant effects were observed in THP-1 macrophages, where FPM reduced ROS production and modulated inflammatory responses by decreasing pro-inflammatory cytokines [tumor necrosis factor alpha (TNF-α), monokine induced by gamma interferon (MIG), interferon-inducible T cell alpha chemoattractant (I-TAC), macrophage inflammatory proteins (MIP)-1α and -1β] and increasing anti-inflammatory interleukin (IL)-10 levels. Mechanistic studies with HEK-Blue reporter cell lines revealed that FPM inhibited nuclear factor kappa B (NF-κB) activation via a toll-like receptor (TLR)4-independent pathway. In vivo, FPM significantly reduced ROS levels in Drosophila melanogaster and improved activity and LT50 values in Caenorhabditis elegans under oxidative stress, although it did not affect intestinal barrier integrity in these models. CONCLUSION The findings indicate that FPM shows promising application as a functional feed supplement for improving intestinal health in livestock by mitigating oxidative stress and inflammation. Further studies, including livestock feeding trials, are recommended to validate these results.
Collapse
Affiliation(s)
- Mara Heckmann
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
| | - Nadiia Sadova
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Georg Sandner
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Cathrina Neuhauser
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Bernhard Blank-Landeshammer
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Bettina Schwarzinger
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Alice König
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Meizhen Liang
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Michael Spitzer
- TVA Produktions- & Vertriebs-Gesellschaft m.b.H, Dorf 156, 3343, Hollenstein, Austria
| | - Julian Weghuber
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria.
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria.
| | - Verena Stadlbauer
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria.
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria.
| |
Collapse
|
43
|
Lu Y, Mi L, Zhang P, Chen Y, Bai X, Li K, Zhang Y, Li J. Induction of Cuproptosis by Dichloromethane Extract From Patrinia scabiosaefolia Fisch on K562 Cells. ENVIRONMENTAL TOXICOLOGY 2025. [PMID: 40177825 DOI: 10.1002/tox.24516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Cuproptosis is a newly identified form of cell death that relies on copper (Cu) ionophores to transport Cu into cancer cells. As a perennial herb, Patrinia scabiosaefolia Fisch (PS) has garnered significant attention owing to its analgesic, anti-inflammatory, antibacterial, and antitumor properties. Previous research has shown that the extract from PS (DEPS) can inhibit the growth of leukemia cell lines. However, the specific mechanism of its anti-leukemic effect has not been fully clarified. Therefore, this study was conducted to investigate the molecular mechanism of cuproptosis in the treatment of leukemia with DEPS. Our results demonstrated that DEPS up-regulated SLC31A1 and down-regulated ATP7B expression, which increased intracellular copper concentration, down-regulated FDX1, influenced the lipoylation of DLAT and DLD, and subsequently increased the expression of the stress protein HSP70 and the expression of PDHA1, inducing copper death in K562 cells. In addition, we investigated the toxicity of DEPS in vivo and demonstrated its low in vivo toxicity and adequate in vivo safety. In conclusion, our results suggest that DEPS may induce cuproptosis in cells, offering valuable insights for the future application of PS in leukemia treatment.
Collapse
Affiliation(s)
- Yuan Lu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Leyuan Mi
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Clinical Laboratory Center, Gansu Provincial Maternity and Child-Care Hospital/Central Hospital of Gansu Province, Lanzhou, China
| | - Peipei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xinyi Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Kejing Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Ying Zhang
- Clinical Laboratory Division II, the First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Juan Li
- Clinical Laboratory Division II, the First Hospital of Lanzhou University, Lanzhou University, Lanzhou, China
| |
Collapse
|
44
|
Wang G, Wang Y, Jin C, Sun X. Off-label use of anlotinib in malignancies' treatment: efficacy and management of adverse reactions. Pharmacol Rep 2025; 77:392-408. [PMID: 39899257 PMCID: PMC11911265 DOI: 10.1007/s43440-025-00700-1] [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: 08/03/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
Anlotinib is a novel small-molecule multi-target tyrosine kinase inhibitor (TKIs) independently developed in China, it possesses the functions of inhibiting tumor angiogenesis and suppressing tumor growth. Anlotinib has achieved notable therapeutic effects in approved indications for advanced non-small cell lung cancer, soft tissue sarcoma, small cell lung cancer, and medullary thyroid carcinoma. Additionally, with unanimous expert consensus, it has been used off-label in various other tumors, yielding favorable outcomes. This article reviews the efficacy and common adverse reactions, as well as their management, of off-label use of anlotinib in various malignant tumors.
Collapse
Affiliation(s)
- Guangli Wang
- Obstetrics and Gynecology Department, Yanbian University Affiliated Hospital, Yanbian, 133000, China
| | - Yuling Wang
- Obstetrics and Gynecology Department, Yanbian University Affiliated Hospital, Yanbian, 133000, China
| | - Changhao Jin
- Obstetrics and Gynecology Department, Yanbian University Affiliated Hospital, Yanbian, 133000, China
| | - Xiaodan Sun
- The First Department of Gynecological Oncology, Jilin Provincial Cancer Hospital, Changchun, 130012, China.
| |
Collapse
|
45
|
Priyamvada P, Ashok G, Joshi T, Anbarasu S, Anbarasu A, Ramaiah S. Unravelling the molecular mechanistic pathway underlying the anticancer effects of kaempferol in colorectal cancer: a reverse pharmacology network approach. Mol Divers 2025; 29:1049-1067. [PMID: 38795259 DOI: 10.1007/s11030-024-10890-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/30/2024] [Indexed: 05/27/2024]
Abstract
Colorectal cancer (CRC) is the third most diagnosed and highly fatal malignancy, presenting serious health concerns worldwide. The search for an effective cure for CRC is challenging and poses a serious concern. Kaempferol is a potent anti-cancerous bioactive compound often suggested for treating various cancers, including CRC. However, its underlying molecular mechanism against CRC remains unclear. The present study delves into kaempferol's molecular pathways and underlying molecular mechanisms against CRC targets. The target protein-coding genes for kaempferol were retrieved, and the CRC-associated genes were curated. Twelve common targets with a disease specificity index of > 0.6 were validated for their protein expression at different stages of CRC. Over-expressed USP1, SETD7, POLH, TDP1 and RACGAP1 were selected for further studies. The binding affinities of kaempferol to the corresponding proteins were evaluated using molecular docking and Molecular Dynamics (MD) simulations. SETD7 exhibited the highest binding affinity with the lowest binding energy (- 8.06 kcal/mol). Additionally, the MD simulation, and MM-PBSA conferred SETD7-kaempferol complex had the least root-mean-square deviation with lower interaction energy and higher conformational stability. The protein-protein interaction of SETD7 constructed revealed direct interactors, namely, DNMT1, FOXO1, FOXO3, FOXO4, H3-3B, H3-4, H3C12, H3C13, SETD7, SIRT1 and TP53, have a potential role in cancer progression through FOXO signalling. In summary, our study revealed kaempferol's multi-target and synergistic effect on multiple CRC targets and its underlying mechanisms. Finally, the study recommends in-vitro and in-vivo trials for validation of anti-cancerous drugs for CRC.
Collapse
Affiliation(s)
- P Priyamvada
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Gayathri Ashok
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Tushar Joshi
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Suvitha Anbarasu
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India.
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Tamil Nadu, Vellore, 632014, India.
| |
Collapse
|
46
|
Wei S, Ma X, Chen Y, Wang J, Hu L, Liu Z, Mo L, Zhou N, Chen W, Zhu H, Yan S. Alzheimer's Disease-Derived Outer Membrane Vesicles Exacerbate Cognitive Dysfunction, Modulate the Gut Microbiome, and Increase Neuroinflammation and Amyloid-β Production. Mol Neurobiol 2025; 62:5109-5132. [PMID: 39514171 DOI: 10.1007/s12035-024-04579-6] [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: 06/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Although our understanding of the molecular biology of Alzheimer's disease (AD) continues to improve, the etiology of the disease, particularly the involvement of gut microbiota disturbances, remains a challenge. Outer membrane vesicles (OMVs) play a key role in central nervous system diseases, but the impact of OMVs on AD progression remains unclear. In this study, we hypothesized that AD-derived OMVs (OMVsAD) were a risk factor in AD pathology. To test our hypothesis, young APP/PS1 mice (AD mice) were given OMVsAD by gavage. Young AD mice were euthanized 120 days after gavage to assess the intestinal barrier, gut microbiota diversity, mediators of neuroinflammation, glial markers, amyloid burden, and short-chain fatty acid (SCFA) levels. Our results showed that OMVsAD accelerated cognitive dysfunction after 120 days of intragastric administration. Morris water maze experiment and new object recognition test showed that OMVsAD caused significantly poorer spatial ability learning and memory of the AD mice. We observed the OMVsAD-treated APP/PS1 mice display OMVs disrupting the intestinal barrier compared with controls of normal human-derived OMVs. Compared with the OMVsHC group, claudin-5 and ZO-1 related to the intestinal barrier were significantly downregulated in the OMVsAD group. The OMVsAD activate microglia in the cerebral cortex and hippocampus of AD mice, and the levels of IL-1β, IL-6, TNF-α, and NF-Κb were upregulated. We also found that OMVsAD increased Aβ production. 16S rRNA sequencing showed that OMVsAD negatively regulated the α- and β-diversity index of intestinal flora and reduced the levels of SCFA. OMVsAD may change the intestinal flora of young AD, damage the intestinal mucosa and blood-brain barrier, and accelerate AD neuropathological damage.
Collapse
Affiliation(s)
- Shouchao Wei
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Xiaochen Ma
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Yating Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junjun Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Basic Medicine College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Li Hu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Basic Medicine College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhou Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lang Mo
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Ning Zhou
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Wenrong Chen
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - He Zhu
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
| | - Shian Yan
- The Third Department of Neurology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.
| |
Collapse
|
47
|
Huang B, An H, Gui M, Qiu Y, Xu W, Chen L, Li Q, Yao S, Lin S, Khrustaleva TA, Wang R, Lin J. Qingjie Fuzheng Granule prevents colitis-associated colorectal cancer by inhibiting abnormal activation of NOD2/NF-κB signaling pathway mediated by gut microbiota disorder. CHINESE HERBAL MEDICINES 2025. [DOI: 10.1016/j.chmed.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
|
48
|
Yousefi Rad A, Rastegari AA, Shahanipour K, Monajemi R. Moringa oleifera and Its Biochemical Compounds: Potential Multi-targeted Therapeutic Agents Against COVID-19 and Associated Cancer Progression. Biochem Genet 2025; 63:936-959. [PMID: 38583096 DOI: 10.1007/s10528-024-10758-w] [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: 08/22/2023] [Accepted: 02/23/2024] [Indexed: 04/08/2024]
Abstract
The Coronavirus disease-2019 (COVID-19) pandemic is a global concern, with updated pharmacological therapeutic strategies needed. Cancer patients have been found to be more susceptible to severe COVID-19 and death, and COVID-19 can also lead to cancer progression. Traditional medicinal plants have long been used as anti-infection and anti-inflammatory agents, and Moringa oleifera (M. oleifera) is one such plant containing natural products such as kaempferol, quercetin, and hesperetin, which can reduce inflammatory responses and complications associated with viral infections and multiple cancers. This review article explores the cellular and molecular mechanisms of action of M. oleifera as an anti-COVID-19 and anti-inflammatory agent, and its potential role in reducing the risk of cancer progression in cancer patients with COVID-19. The article discusses the ability of M. oleifera to modulate NF-κB, MAPK, mTOR, NLRP3 inflammasome, and other inflammatory pathways, as well as the polyphenols and flavonoids like quercetin and kaempferol, that contribute to its anti-inflammatory properties. Overall, this review highlights the potential therapeutic benefits of M. oleifera in addressing COVID-19 and associated cancer progression. However, further investigations are necessary to fully understand the cellular and molecular mechanisms of action of M. oleifera and its natural products as anti-inflammatory, anti-COVID-19, and anti-cancer strategies.
Collapse
Affiliation(s)
- Ali Yousefi Rad
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Esfahan, Iran
| | - Ali Asghar Rastegari
- Department of Molecular and Cell Biochemistry, Falavarjan Branch, Islamic Azad University, Esfahan, Iran.
| | - Kahin Shahanipour
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Esfahan, Iran
| | - Ramesh Monajemi
- Department of Biology, Falavarjan Branch, Islamic Azad University, Esfahan, Iran
| |
Collapse
|
49
|
Ghorbaninezhad F, Nour MA, Farzam OR, Saeedi H, Vanan AG, Bakhshivand M, Jafarlou M, Hatami-Sadr A, Baradaran B. The tumor microenvironment and dendritic cells: Developers of pioneering strategies in colorectal cancer immunotherapy? Biochim Biophys Acta Rev Cancer 2025; 1880:189281. [PMID: 39929377 DOI: 10.1016/j.bbcan.2025.189281] [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: 05/31/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Colorectal cancer (CRC) is the world's third most frequent cancer, and both its incidence and fatality rates are rising. Despite various therapeutic approaches, neither its mortality rate nor its recurrence frequency has decreased significantly. Additionally, conventional treatment approaches, such as chemotherapy and radiotherapy, have several side effects and risks for patients with CRC. Accordingly, the need for alternative and effective treatments for CRC patients is critical. Immunotherapy that utilizes dendritic cells (DCs) harnesses the patient's immune system to combat cancer cells effectively. DCs are the most potent antigen-presenting cells (APCs), which play a vital role in generating anti-cancer T cell responses. A significant barrier to the immune system's ability to eliminate CRC is the establishment of a potent immunosuppressive tumor milieu by malignant cells. Since DCs are frequently defective in this milieu, the tumor setting significantly reduces the effectiveness of DC-based therapy. Determining central mechanisms contributing to tumor growth by unraveling and comprehending the interaction between CRC tumor milieu and DCs may lead to new therapeutic approaches. This study aims to review DC biology and discuss its role in T-cell-mediated anti-tumor immunity, as well as to highlight the immunosuppressive effects of the CRC tumor milieu on the function of DCs. We will also highlight the tumor microenvironment (TME)-related factors that interfere with DC function as a possible therapeutic target to enhance DC-based cell therapy efficacy.
Collapse
Affiliation(s)
- Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Bakhshivand
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Jafarlou
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Behzad Baradaran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
50
|
Zheng Y, Zheng Y, Chen H, Tan X, Zhang G, Kong M, Jiang R, Yu H, Shan K, Liu J, Zhang R, Liu Z, Wu J. Erianin triggers ferroptosis in colorectal cancer cells by facilitating the ubiquitination and degradation of GPX4. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156465. [PMID: 39938177 DOI: 10.1016/j.phymed.2025.156465] [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: 09/15/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) continues to represent a significant global public health challenge. Ferroptosis, a novel form of cell death dependent on iron and involving lipid peroxidation, has emerged as an effective strategy for treating various cancers with great potential for application. PURPOSE This study aimed to investigate the therapeutic potential of erianin, a novel dibenzyl compound isolated from the well-known herbal medicine Dendrobium chrysotoxum Lindl, in the treatment of CRC through induction of ferroptosis. METHODS Human CRC HCT116 and SW480 cells were employed for in vitro investigations, while an AOM/DSS CRC animal model was established for in vivo experiments. RESULTS The results demonstrated that erianin effectively inhibited the growth of CRC cells and suppressed tumorigenesis in the AOM/DSS CRC animal model. Erianin induced ferroptosis in CRC cells as evidenced by a significant increase in intracellular Fe2+ levels and lipid peroxides, along with a decrease in glutathione. Additionally, ferroptosis inhibitors reversed the cytotoxicity of erianin against CRC cells as well as its induction of ferroptosis. Notably, novel glutathione peroxidase 4 (GPX4), a core regulatory factor of ferroptosis, was found to be overexpressed in human primary colon adenocarcinoma tissues compared with normal tissues. However, erianin significantly reduced GPX4 expression by facilitating its ubiquitination and degradation. Furthermore, the overexpression of GPX4 mitigated erianin-induced ferroptotic cell death; conversely, the silencing of GPX4 amplified these effects. CONCLUSION Erianin demonstrates the potential to inhibit CRC by inducing ferroptosis through accelerating the ubiquitination and degradation of GPX4, indicating its promise as a therapeutic candidate against CRC.
Collapse
Affiliation(s)
- Yuting Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Yinli Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Pathology, Guangzhou 510060, China.
| | - Haipeng Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Xuanjing Tan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Guiyu Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; The Seventh Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518000, China.
| | - Muyan Kong
- The Seventh Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518000, China.
| | - Ruidi Jiang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Hong Yu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Keyao Shan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Jiyao Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| | - Jinjun Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|