1
|
Li CX, Xu Q, Jiang ST, Liu D, Tang C, Yang WL. Anticancer effects of salvianolic acid A through multiple signaling pathways (Review). Mol Med Rep 2025; 32:176. [PMID: 40280109 PMCID: PMC12056544 DOI: 10.3892/mmr.2025.13541] [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: 11/04/2024] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Salvia miltiorrhiza Bunge (Salvia miltiorrhiza), commonly referred to as Danshen, is a well‑known herb in traditional Chinese medicine, the active ingredients of which are mostly categorized as water soluble and lipid soluble. Salvianolic acids are the major water‑soluble phenolic acid constituents of Danshen; salvianolic acid B is the most prevalent, with salvianolic acid A (SAA) being the next most predominant form. SAA offers a wide array of pharmacological benefits, including cardiovascular protection, and anti‑inflammatory, antioxidant, antiviral and anticancer activities. SAA is currently undergoing phase III clinical trials for diabetic peripheral neuropathy and has shown protective benefits against cardiovascular illnesses; furthermore, its safety and effectiveness are encouraging. By targeting several signaling pathways, preventing cell cycle progression, tumor cell migration, invasion and metastasis, normalizing the tumor vasculature and encouraging cell apoptosis, SAA can also prevent the growth of malignancies. In addition, it enhances sensitivity to chemotherapeutic drugs, and alleviates their toxicity and side effects. However, the broad therapeutic use of SAA has been somewhat limited by its low content in Salvia miltiorrhiza Bunge and the difficulty of its extraction techniques. Therefore, the present review focuses on the potential mechanisms of SAA in tumor prevention and treatment. With the anticipation that SAA will serve a notable role in clinical applications in the future, these discoveries may offer a scientific basis for the combination of SAA with conventional chemotherapeutic drugs in the treatment of cancer, and could establish a foundation for the development of SAA as an anticancer drug.
Collapse
Affiliation(s)
- Cheng-Xia Li
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qi Xu
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Shi-Ting Jiang
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Dan Liu
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chao Tang
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wen-Li Yang
- Institute for Cancer Medicine, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
2
|
Liang Y, Su T, Zhu S, Sun R, Qin J, Yue Z, Wang X, Liang Z, Tan X, Bian Y, Zhao F, Tang D, Yin G. Astragali Radix-Curcumae Rhizoma normalizes tumor blood vessels by HIF-1α to anti-tumor metastasis in colon cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156562. [PMID: 40023968 DOI: 10.1016/j.phymed.2025.156562] [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: 10/09/2024] [Revised: 02/13/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Abnormal tumor blood vessels can significantly promote the malignant progression of tumors, prompting researchers to focus on drugs that normalize these vessels for clinical treatment. The combination of the Qi-tonifying drug Astragali Radix and the blood-activating drug Curcumae Rhizoma, referred to as AC, exhibited significant anti-tumor metastasis effects. However, the association between the anti-tumor metastasis effect of AC and its potential role in regulating tumor vascular remodeling warrants further exploration. PURPOSE This study aimed to elucidate the mechanism through which AC induces tumor blood vessel normalization in colon cancer (CC). METHODS The potential active components of AC were identified through UPLC-MS/MS. An orthotopic transplantation model of CC was established in BALB/c mice using the CT26-Lucifer cell line, and the effects of AC were evaluated using IVIS imaging, hematoxylin and eosin (H&E) staining, and immunohistochemistry. Network pharmacology and molecular biology analyses were employed to identify the potential direct targets of AC. Subsequently, RT-PCR and Western blotting techniques were utilized to validate the findings obtained from network pharmacology. Furthermore, ELISA and other methodologies were used to investigate glycolysis-related indicators, along with immunofluorescence technology to demonstrate changes in vascular leakage and perfusion characteristics associated with blood vessel normalization. RESULTS We identified HIF-1α as a potential direct target of AC. This interaction influences the glycolytic processes in both tumor cells and tumor-associated endothelial cells (TECs) by directly binding to HIF-1α and modulating its nuclear translocation, thereby determining the integrity of TEC junctions. Mechanistically, AC directly regulates the key enzyme PFKFB3 in glycolysis by modulating HIF-1α expression and inhibiting its nuclear translocation. This action reduces tumor glycolytic flux, decreases the internalization of VE-cad, and influences the expression of downstream matrix metalloproteinases (MMPs), thereby strengthening the adherens and tight junctions between TECs and restoring vascular integrity. CONCLUSION This study presents novel findings that AC can regulate glycolysis through the inhibition of HIF-1α nuclear translocation, thereby promoting the normalization of tumor blood vessels and effectively inhibiting tumor metastasis. These results suggested that AC may serve as an effective therapeutic agent for normalizing tumor blood vessels.
Collapse
Affiliation(s)
- Yan Liang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tingting Su
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shijiao Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ruolan Sun
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiahui Qin
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zengyaran Yue
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu Wang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhongqing Liang
- School of Acupuncture-Moxibustion and Tuina · School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiying Tan
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yong Bian
- Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Fan Zhao
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Decai Tang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Gang Yin
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
3
|
Yuan J, Tao Y, Wang M, Chen Y, Han X, Wu H, Shi H, Huang F, Wu X. Astragaloside II, a natural saponin, facilitates remyelination in demyelination neurological diseases via p75NTR receptor mediated β-catenin/Id2/MBP signaling axis in oligodendrocyte precursor cells. J Adv Res 2025:S2090-1232(25)00273-5. [PMID: 40258474 DOI: 10.1016/j.jare.2025.04.028] [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: 12/18/2024] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Demyelination is a hallmark of neurological disorders such as multiple sclerosis and neuromyelitis optica, leading to neurological deficits. Existing therapies primarily modulate immune responses but lack efficacy in directly promoting myelin repair. Enhancing oligodendrocyte precursor cell (OPC) differentiation and oligodendrocytes (OLs) production is crucial for restoring myelin integrity. OBJECTIVES This study investigated the therapeutic potential of astragaloside II (AS-II), a bioactive saponin with neuroprotective and pro-differentiation properties, derived from Astragalus membranaceus, uniquely in promoting OPC differentiation and myelin endogenous repair, distinguishing it from existing immunomodulatory treatments. AS-II directly targets p75 neurotrophin receptor (p75NTR) signaling, a pathway linked to myelin regeneration but underestimated in current remyelination strategies. METHODS We conducted in vitro OPC differentiation assays and in vivo demyelination models, including cuprizone and experimental autoimmune encephalomyelitis. Drug affinity responsive target stability mass spectrometry, cellular thermal shift assay, and surface plasmon resonance assays identified and validated p75NTR as the direct target of AS-II. p75NTR knockout mice and lentiviral transduction were used to confirm its role. RESULTS AS-II improved neurobehavioral outcomes, increased OLs production, and enhanced myelin integrity by suppressing β-catenin/Id2/MBP signaling. Mechanistically, AS-II bound to p75NTR (Pro253, Ser257), stabilizing its structure and promoting remyelination. In p75NTR knockout mice, AS-II failed to restore myelin or neural function, confirming its p75NTR-dependent mechanism. CONCLUSION AS-II represents a novel therapeutic candidate for demyelinating diseases, offering a targeted approach to myelin regeneration through direct p75NTR modulation and addressing gaps in current treatment strategies.
Collapse
Affiliation(s)
- Jinfeng Yuan
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanlin Tao
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute for Translational Brain Research, Fudan University, Shanghai 200433, China
| | - Mengxue Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yufeng Chen
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinyan Han
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hailin Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
4
|
Wang W, Yuan J, Zhu Y, Li R, Zhang J. Traditional Chinese medicine (TCM) enhances the therapeutic efficiency of a gemcitabine-loaded injectable hydrogel on postoperative breast cancer through modulating the microenvironment. J Mater Chem B 2025; 13:4864-4878. [PMID: 40171620 DOI: 10.1039/d4tb02776k] [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: 04/04/2025]
Abstract
Local injection of the drug-loaded hydrogel at the surgery site is promising for postoperative breast cancer. However, the postoperative changes in the tumor microenvironment, such as inflammation, abnormal angiogenesis and hypoxia, inhibit drug perfusion and contribute to breast cancer recurrence (BCR). Normalizing the abnormal blood vessels can effectively improve perfusion and reduce hypoxia. Here, we encapsulated gemcitabine (GEM) in a PLGA-PEG-PLGA hydrogel (GEM-hydrogel) for local treatment of postoperative breast cancer. The GEM-hydrogel can be injected into the surgery cavity allowing sustained release of the drug. Meanwhile, traditional Chinese medicine (TCM) Shexiang Baoxin Pill (SBP) was given to normalize the blood vessels to enhance drug perfusion. The results suggest that the combination of SBP enhances the therapeutic efficiency of the GEM-hydrogel, inhibiting tumor recurrence. Mechanism studies reveal that SBP works by promoting PDGFB expression in macrophages, subsequently recruiting pericytes, and normalizing blood vessels, finally alleviating hypoxia. This study demonstrates that the combination of TCM and chemotherapeutics is promising for suppressing postoperative tumor recurrence.
Collapse
Affiliation(s)
- Wenxu Wang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jixiang Yuan
- Urology Centre, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200000, China
| | - Yuying Zhu
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ruixiang Li
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jiange Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
5
|
Zhu Q, Zhang R, Zhao Z, Xie T, Sui X. Harnessing phytochemicals: Innovative strategies to enhance cancer immunotherapy. Drug Resist Updat 2025; 79:101206. [PMID: 39933438 DOI: 10.1016/j.drup.2025.101206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025]
Abstract
Cancer immunotherapy has revolutionized cancer treatment, but therapeutic ineffectiveness-driven by the tumor microenvironment and immune evasion mechanisms-continues to limit its clinical efficacy. This challenge underscores the need to explore innovative approaches, such as multimodal immunotherapy. Phytochemicals, bioactive compounds derived from plants, have emerged as promising candidates for overcoming these barriers due to their immunomodulatory and antitumor properties. This review explores the synergistic potential of phytochemicals in enhancing immunotherapy by modulating immune responses, reprogramming the tumor microenvironment, and reducing immunosuppressive factors. Integrating phytochemicals with conventional immunotherapy strategies represents a novel approach to mitigating resistance and enhancing therapeutic outcomes. For instance, nab-paclitaxel has shown the potential in overcoming resistance to immune checkpoint inhibitors, while QS-21 synergistically enhances the efficacy of tumor vaccines. Furthermore, we highlight recent advancements in leveraging nanotechnology to engineer phytochemicals for improved bioavailability and targeted delivery. These innovations hold great promise for optimizing the clinical application of phytochemicals. However, further large-scale clinical studies are crucial to fully integrate these compounds into immunotherapeutic regimens effectively.
Collapse
Affiliation(s)
- Qianru Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao
| | - Ruonan Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ziming Zhao
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao; Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China.
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macao; Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China.
| |
Collapse
|
6
|
Qian C, Huang Y, Zhang S, Yang C, Zheng W, Tang W, Wan G, Wang A, Lu Y, Zhao Y. Integrated identification and mechanism exploration of bioactive ingredients from Salvia miltiorrhiza to induce vascular normalization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156427. [PMID: 39892310 DOI: 10.1016/j.phymed.2025.156427] [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: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND The clinical management of ischemic disease and cancer is complex, with disruptions in local vascular function and tumor angiogenesis contributing to blood stasis, which complicates treatment strategies. Salvia miltiorrhiza, a natural product, is known to restore vascular structure and function. However, its specific roles in concurrently addressing ischemic disease and cancer within the same organism remain poorly understood. PURPOSE This study aimed to explore the material basis, pharmacological effects, and underlying mechanisms of Salvia miltiorrhiza extract (SME) in promoting blood flow recovery in ischemic hindlimbs and inducing tumor vascular normalization. METHODS The pharmacological effects of SME were evaluated in a mouse model combining ischemic hindlimbs and tumors. Mice were administered low (SME-L) or high (SME-H) doses of SME daily, and the gastrocnemius muscle mass and tumor vascular structure were assessed. Laser Doppler perfusion imaging (LDPI) was used to monitor hindlimb blood flow recovery and tumor vascular perfusion. The pharmacokinetics of the key bioactive constituents in SME were characterized by liquid chromatography-mass spectrometry (LC-MS). Interactions between SME's active compounds and predicted targets were investigated using molecular docking, microscale thermophoresis (MST), and luciferase reporter assays. The synergistic effects of the primary components, Tanshinone I (Tan I) and Salvianolic acid A (Sal A), were analyzed through tube formation assays, enzyme-linked immunosorbent assays (ELISA), immunofluorescence staining, and western blot. RESULTS Phytochemical profiling revealed that SME contains several active compounds, including Danshensu, Sal A, Sal B, Tan IIA, and Tan I. SME treatment reduced the frequency of necrotic toes, increased muscle mass, and alleviated hypoxia in the gastrocnemius muscle. SME significantly improved tumor vascular perfusion and notably enhanced pericyte coverage and basement membrane integrity. Pharmacokinetic analysis identified Tan I and Sal A as the key bioactive components that promote vascular normalization. Tan I inhibited FoxO1, preventing endothelial cell activation induced by angiopoietin 2 (Ang2), while Sal A bound to Ang2, facilitating Tie2 activation mediated by Ang1. Both in vitro and in vivo results demonstrated that the combination of Tan I and Sal A exerted a synergistic therapeutic effect on correcting abnormal blood vessels in ischemic hindlimbs and tumors. CONCLUSION Our study innovatively revealed a reliable mouse model wherein the Ang2/Tie2 signaling cascade disrupted the endothelial homeostasis to aggravate the progression of hindlimb ischemia and tumor angiogenesis. This balance can be rescued by the combination therapy of Tan I and Sal A that were both from SME, leading to the occurrence of vascular normalization.
Collapse
Affiliation(s)
- Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Ying Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Shan Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chunmei Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Weiwei Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Weiwei Tang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Guiping Wan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
7
|
Fu Z, Liu H, Kuang Y, Yang J, Luo M, Cao L, Zheng W. β-elemene, a sesquiterpene constituent from Curcuma phaeocaulis inhibits the development of endometriosis by inducing ferroptosis via the MAPK and STAT3 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119344. [PMID: 39800242 DOI: 10.1016/j.jep.2025.119344] [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: 10/01/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rhizome of Curcuma phaeocaulis Valeton, Curcuma wenyujin Y.H. Chen & C. Ling, or Curcuma kwangsiensis S. G. Lee et C. F. Liang, commonly known as Wen-E-Zhu and E'zhu, has been utilized in traditional Chinese medicine for the treatment of cancer and gynecological diseases since antiquity. This traditional medicinal herb is highly esteemed for its efficacy in promoting blood circulation, dissolving blood stasis, reducing swelling, and alleviating pain. β-Elemene (β-ELE), a sesquiterpene compound derived from Curcuma phaeocaulis, has demonstrated potential in inhibiting tumor cell proliferation and inducing ferroptosis, which have been extensively studied in various malignant neoplasms. Previous studies have confirmed that Sparganium stoloniferum-Curcuma phaeocaulis containing β-ELE may possess anti-endometriotic properties. However, the exact mechanism underlying β-ELE's anti-endometriosis activity remains largely unknown and requires further research and investigation. AIM OF THE STUDY To identify the anti-endometriosis target of β-ELE and elucidate the underlying molecular mechanism of β-ELE in endometriosis, focusing on inducing ferroptosis. MATERIALS AND METHODS The target pathway of β-ELE in endometriosis treatment was predicted through network pharmacology and bioinformatics analysis. Surface plasmon resonance-high performance liquid chromatography-protein mass spectrometry (SPR-HPLC-MS) and molecular docking were used to further identify the potential targets of β-ELE in endometriosis. The immortalized endometriosis epithelial cell line 12Z was used for in vitro study. The effect of β-ELE on cell proliferation and migration was detected by CCK-8, EdU and wound healing assay, and ultrastructural changes were examined via transmission electron microscopy. The effect of β-ELE-induced ferroptosis was determined by western blot, immunohistochemistry staining and flow cytometry. SPR affinity analysis was performed to specific the direct interaction between β-ELE and FTH1, FTL, GPX4, STAT3 and MAPK14. To establish a mouse model of endometriosis and to assess the inhibitory effects of β-ELE and ELE injection on endometriosis in vivo as well as safety profile of administration, and investigate the effects and underlying mechanisms of β-ELE and ELE injection on ferroptosis in ectopic lesions. RESULTS SPR-HPLC-MS was employed to identify 76 potential targets of β-ELE for endometriosis treatment, closely linked to ferroptosis. Molecular docking revealed that glutathione peroxidase 4 (GPX4), ferritin heavy chain 1 (FTH1), ferritin light chain (FTL), signal transducer and activator of transcription 3 (STAT3), and mitogen-activated protein kinase 14 (MAPK14) are key action targets of β-ELE in endometriosis. Further investigations revealed that β-ELE inhibited the proliferation and migration of endometriotic cells in vitro while inducing ferroptosis, as evidenced by increased levels of iron, reactive oxygen species (ROS), and lipid peroxidation. In a mouse model, β-ELE inhibited the growth of endometriotic lesions, induced ferroptosis, suppressed fibrosis, and exhibited anti-endometriotic effects. Mechanistically, β-ELE downregulates the expression levels of GPX4, FTH1, and FTL and inhibited the phosphorylation of STAT3 and MAPK14, which may elucidate its underlying molecular mechanisms. CONCLUSION This study demonstrates that the inhibitory effect of β-ELE on endometriosis by inducing ferroptosis in vitro and in vivo. Our results revealed that β-ELE exerts anti-endometriosis effects by inducing ferroptosis via the MAPK and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Zhiyi Fu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Hao Liu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Yanqi Kuang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Jiumei Yang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Meicheng Luo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Lixing Cao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Weilin Zheng
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Peng YC, He ZJ, Yin LC, Pi HF, Jiang Y, Li KY, Tian L, Xie J, Zhang JB, Li CY, Feng GY, Wang K, Zhou DZ, Xie XW, Zhang ZY, Fan TF. Sanguinarine suppresses oral squamous cell carcinoma progression by targeting the PKM2/TFEB aix to inhibit autophagic flux. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156337. [PMID: 39729782 DOI: 10.1016/j.phymed.2024.156337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is one of the most common malignancies. However, there is no effective treatment for OSCC. PURPOSE This study aimed to identify a natural compound with significant efficacy against OSCC and elucidate its primary mechanism of action. METHODS An FDA-approved drug library and an MCE autophagy-related molecular compound library were screened through high-throughput screening to identify an effective natural compound against OSCC. The IC50 value of sanguinarine (Sang) in OSCC cells was determined using a CCK8 assay. Immunoblotting and immunofluorescence staining were used to assess the effect of Sang on autophagic flux in OSCC cells. Changes in the acidic lysosomal environment were evaluated using RFP-GFP-LC3B and LysoSensor Green DND-189. Furthermore, limited proteolysis-coupled mass spectrometry (LiP-MS) and virtual screening techniques were utilized to identify direct binding targets of Sang, which were subsequently validated by surface plasmon resonance (SPR) and microscale thermophoresis (MST). Molecular docking combined with molecular dynamics analysis identified the binding site between the target protein and Sang. In vitro and in vivo investigations with mutant plasmids confirmed this finding. RESULTS Screening led to the identification of the naturally occurring autophagy modulator Sang as a potent inhibitor of OSCC progression. Moreover, Sang impaired lysosomal function through reducing lysosomal-associated membrane proteins, inhibiting lysosomal proteolysis, and altering the lysosomal pH. These effects contributed to defects in autophagic clearance and subsequently suppressed OSCC progression. Notably, Sang bound the phenylalanine 26 (F26) residue in pyruvate kinase M2 (PKM2) and inhibited PKM2 enzymatic activity, subsequently suppressing transcription factor EB (TFEB) expression to inhibit lysosomal function and blocking autophagic flux in OSCC cells. CONCLUSION Our results demonstrate for the first time that Sang can suppress the PKM2/TFEB axis, and influence lysosomal function, thereby blocking autophagy and inhibiting the progression of OSCC, making it a promising therapeutic option for the treatment of OSCC.
Collapse
Affiliation(s)
- Yong-Chun Peng
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhi-Jing He
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lun-Cai Yin
- Department of Oncology, Affiliated Dazu Hospital of Chongqing Medical University, Chongqing 402360, China
| | - Hui-Feng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China. State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400038, China
| | - Yi Jiang
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ke-Yan Li
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China. State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400038, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Army Medical University (Third Military Medical University), Chongqing 400038, China. State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400038, China
| | - Jian-Bo Zhang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chen-Yao Li
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Guan-Ying Feng
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China
| | - Kai Wang
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ding-Zhou Zhou
- Department of Neurosurgery, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, China
| | - Xiao-Wei Xie
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Zhi-Yuan Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China; Department of Oral and Maxillofacial Surgery, Zhang Zhiyuan Academician Workstation, Hainan Western Central Hospital, Shanghai Ninth People's Hospital, Danzhou, Hainan 571700, China.
| | - Teng-Fei Fan
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China; Department of Oral and Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai 200011, China.
| |
Collapse
|
9
|
Cao L, Wang X, Ma X, Xu M, Li J. Potential of natural products and gut microbiome in tumor immunotherapy. Chin Med 2024; 19:161. [PMID: 39567970 PMCID: PMC11580227 DOI: 10.1186/s13020-024-01032-7] [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: 06/22/2024] [Accepted: 11/01/2024] [Indexed: 11/22/2024] Open
Abstract
Immunotherapy is a novel treatment approach for malignant tumors, which has opened a new journey of anti-tumor therapy. Although some patients will show a positive response to immunotherapy, unfortunately, most patients and cancer types do not achieve an ideal response to immunotherapy. Therefore, it is urgent to search for the pathogenesis of sensitized immunotherapy. This review indicates that Fusobacterium nucleatum, Coprobacillus cateniformis, Akkermansia muciniphila, Bifidobacterium, among others, as well as intestinal microbial metabolites are closely associated with resistance to anti-tumor immunotherapy. While natural products of pectin, inulin, jujube, anthocyanins, ginseng polysaccharides, diosgenin, camu-camu, and Inonotus hispidus (Bull).Fr. P. Karst, Icariside I, Safflower yellow, Ganoderma lucidum, and Ginsenoside Rk3, and other Chinese native medicinal compound prescriptions to boost their efficacy of anti-tumor immunotherapy through the regulation of microbiota and microbiota metabolites. However, current research mainly focuses on intestinal, liver, and lung cancer. In the future, natural products could be a viable option for treating malignant tumors, such as pancreatic, esophageal, and gastric malignancies, via sensitizing immunotherapy. Besides, the application characteristics of different types, sources and efficacy of natural products in different immune resistance scenarios also need to be further clarified through the development of future immunotherapy-related studies.
Collapse
Affiliation(s)
- Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Xinmiao Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Xinyi Ma
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Manman Xu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixian'ge Street, Xicheng District, Beijing, China.
| |
Collapse
|
10
|
Cheng Y, Li J, Feng X, Wu Y, Wu X, Lau BWM, Ng SSM, Lee SMY, Seto SW, Leung GPH, Hu Y, Fu C, Zhang S, Zhang J. Taohong Siwu decoction enhances the chemotherapeutic efficacy of doxorubicin by promoting tumor vascular normalization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155995. [PMID: 39270591 DOI: 10.1016/j.phymed.2024.155995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Instead of completely suppressing blood vessels inside tumors, vascular normalization therapy is proposed to normalize and prune the abnormal vasculature in tumor microenvironment (TME) to acquire a normal and stable blood flow and perfusion. The theoretical basis for the use of "blood-activating and stasis-resolving" formulas in Traditional Chinese Medicine to treat cancer is highly consistent with the principle of vascular normalization therapy, suggesting the potential application of these traditional formulas in vascular normalization therapy. PURPOSE To study the underlying mechanisms of a classical "blood-activating and stasis-resolving" formula, Taohong Siwu decoction (TSD), in enhancing the efficacy of chemotherapy for breast cancer treatment. STUDY DESIGN HUVECs and transgenic zebrafish embryos were used as the major model in vitro. A 4T1 mouse breast cancer model was applied to study tumor vasculature normalization of TSD and the combination effects with DOX. RESULTS Our data showed that TSD exhibited anti-angiogenic potential in HUVECs and transgenic zebrafish embryos. After 20 days treatment, TSD significantly normalized the tumor vasculature by remodeling vessel structure, reducing intratumoral hypoxia and vessel leakage, and promoting vessel maturation and blood perfusion in 4T1 breast tumor-bearing mice. Moreover, the anti-tumor efficacy of doxorubicin liposome in 4T1 breast tumors was significantly improved by TSD, including the suppression of tumor cell proliferation, angiogenesis, hypoxia, and the increase of cell apoptosis, which is likely through the vascular normalization induced by TSD. TSD also shifted the macrophage polarization from M2 to M1 phenotype in TME during the combination therapy, as evidenced by the reduced number of CD206+ macrophages and increased number of CD86+ macrophages. Additionally, TSD treatment protected against doxorubicin-induced cardiotoxicity in animals, as evidenced by the reduced cardiomyocytes apoptosis and improved heart function. CONCLUSION This study demonstrated for the first time that TSD as a classical Chinese formula can enhance the drug efficacy and reduce the side effects of doxorubicin. These findings can support that TSD could be used as an adjuvant therapy in combination with conventional chemotherapy for the future breast cancer treatment.
Collapse
Affiliation(s)
- Yanfen Cheng
- School of Food and Biological Engineering, Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), University of Chengdu, Chengdu, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Avenue,Wenjiang District, Chengdu city, Chengdu, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China; The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China.
| | - Xi Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Avenue,Wenjiang District, Chengdu city, Chengdu, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Avenue,Wenjiang District, Chengdu city, Chengdu, China
| | - Xiaoping Wu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Regions of China
| | - Benson Wui Man Lau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China
| | - Shamay Sheung Mei Ng
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China; The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China
| | - Simon Ming-Yuen Lee
- The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China; Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China
| | - Sai-Wang Seto
- The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China; Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong Special Administrative Regions of China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Regions of China
| | - Yichen Hu
- School of Food and Biological Engineering, Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), University of Chengdu, Chengdu, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Avenue,Wenjiang District, Chengdu city, Chengdu, China
| | - Siyuan Zhang
- Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, School of Laboratory Medicine, Chengdu Medical College, Chengdu, China; The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No.1166 Liutai Avenue,Wenjiang District, Chengdu city, Chengdu, China.
| |
Collapse
|