1
|
Zhang Y, Zhang Y, Zhang J, Lai W, He G, Shi J, Zhang C, Xiong L, Wang T, Ye F, Jiang X. Integrated transcriptomics and metabolomics unravel the key metabolic pathways involved in the therapeutic mechanism of Salvianic acid A against hepatic fibrosis. Toxicol Appl Pharmacol 2025; 500:117398. [PMID: 40389097 DOI: 10.1016/j.taap.2025.117398] [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: 02/07/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/21/2025]
Abstract
Effective drugs for the clinical treatment of hepatic fibrosis have not yet been identified. Salvianic acid A (SAA) protective mechanisms primarily include anti-inflammation, anti-oxidative stress, and modulation of immune system function. Metabolic dysfunction is well recognized as the driver for hepatic fibrosis. However, the precise action mode and underlying mechanism of SAA in modulating hepatic metabolism to combat hepatic fibrosis remain incompletely understood. This study aimed to investigate the metabolic mechanism by which SAA improves hepatic fibrosis based on metabolomics and transcriptomics profiling. A mouse model of carbon tetrachloride (CCl4)-induced hepatic fibrosis mouse model was established, and protective effects of SAA were evaluated through pathological characteristics. Integrated metabolomics and transcriptomics analysis revealed three key altered metabolic pathways: bile secretion, carbohydrate digestion and absorption, and regulation of lipolysis in adipocytes. SAA modulated the bile secretion pathway, dependent on reducing water channel protein Aqp1, cholesterol synthesis enzyme Hmgcr and Na+/K+-ATPase enzyme Atp1a3, accompanied by up-regulating metabolites glutathione and glucose levels. SAA also regulated carbohydrate digestion and absorption by decreasing the glucose homeostasis-related Akt3, essential enzyme G6pc for gluconeogenesis/glycogenolysis and glucose transporter Atp1a3 with a concomitant increase of metabolites D-galactose, maltose, and sucrose levels. Moreover, SAA improved lipolysis in adipocytes in liver fibrosis through inhibiting lipolysis related Prkg1, lipid transporter Fabp4, lipolysis-associated Akt3 and increasing lipolysis mediator Adrb3, along with upregulated levels of metabolites adenosine monophosphate and norepinephrine. In conclusion, SAA alleviates hepatic fibrosis through modulating metabolic disorders, mainly relying on the metabolic improvements of bile secretion, carbohydrate digestion and absorption and adipocyte lipolysis.
Collapse
Affiliation(s)
- Yunan Zhang
- School of Medicine Shanghai University, Shanghai 200444, China; Department of Infectious Disease, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Second Affiliated Hospital, Shanghai University, Wenzhou Sixth People's Hospital, Wenzhou 325000, China
| | - Yan Zhang
- Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jianan Zhang
- School of Medicine Shanghai University, Shanghai 200444, China
| | - Wenfang Lai
- Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Guiqing He
- School of Medicine Shanghai University, Shanghai 200444, China; Department of Infectious Disease, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Second Affiliated Hospital, Shanghai University, Wenzhou Sixth People's Hospital, Wenzhou 325000, China
| | - Jichan Shi
- School of Medicine Shanghai University, Shanghai 200444, China; Department of Infectious Disease, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Second Affiliated Hospital, Shanghai University, Wenzhou Sixth People's Hospital, Wenzhou 325000, China
| | - Chuan Zhang
- School of Medicine Shanghai University, Shanghai 200444, China; Shanghai Zhongshi Pharmaceutical Co., Ltd., Shanghai 201908, China
| | - Liyan Xiong
- School of Medicine Shanghai University, Shanghai 200444, China; Shanghai Zhongshi Pharmaceutical Co., Ltd., Shanghai 201908, China.
| | - Tingfang Wang
- School of Medicine Shanghai University, Shanghai 200444, China; Shanghai Zhongshi Pharmaceutical Co., Ltd., Shanghai 201908, China.
| | - Fei Ye
- School of Medicine Shanghai University, Shanghai 200444, China; Shanghai Zhongshi Pharmaceutical Co., Ltd., Shanghai 201908, China.
| | - Xiangao Jiang
- School of Medicine Shanghai University, Shanghai 200444, China; Department of Infectious Disease, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, Second Affiliated Hospital, Shanghai University, Wenzhou Sixth People's Hospital, Wenzhou 325000, China.
| |
Collapse
|
2
|
Zhao J, Shi L, Yang Y, Zhu J, Zhou Z, Dong P, Liu S, Yang Z, Gong W. Wu-Mei-Wan promotes ferroptosis in gallbladder cancer through STAT3 negative regulation: An integrated HPLC, proteomics, network pharmacology, and experimental validation study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119671. [PMID: 40188897 DOI: 10.1016/j.jep.2025.119671] [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: 12/23/2024] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a traditional Chinese medicine, Wu-Mei-Wan (WMW) has shown promise as a second-line treatment for gallbladder cancer, but its mechanism remains to be explored. AIM OF THE STUDY To determine the specific mechanism of WMW active ingredients (Palmatine et al.) inducing ferroptosis in gallbladder cancer (GBC) and its synergistic potential with gemcitabine. MATERIALS AND METHODS Subcutaneous tumor in nude mice was used to analyze the combined effect of gemcitabine. The effective components into blood were identified by HPLC. Combined with proteomics, network pharmacology and bioinformatics analysis, the effective components and targets of WMW promoting ferroptosis in GBC were identified in vitro and in vivo. The anticancer effects of WMW on different GBC cell lines were evaluated by CCK-8 assay, colony formation and EdU staining. A variety of molecular biology experiments were used to explore the mechanism. RESULTS WMW treatment enhanced the sensitivity of GBC to gemcitabine, which induced ferroptosis and effectively inhibited the malignant phenotype of GBC. Network pharmacology and blood component identification identified the key components of WMW inhibiting GBC. Palmitine and other components were identified as active components into the blood. Proteomics and molecular docking validation further revealed the STAT3-centered regulatory network in GBC cells. Molecular experiments have shown that WMW induced ferroptosis by negatively regulating downstream molecules of p-STAT3 transcription of GPX4, ACSL4, HIF1α, and FTH1. CONCLUSIONS WMW induces ferroptosis in GBC through the p-STAT3 axis and enhances sensitivity to gemcitabine, suggesting the potential of WMW as a second-line therapeutic for GBC.
Collapse
Affiliation(s)
- Jingwei Zhao
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Liuqing Shi
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Yue Yang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Jiayun Zhu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Zhe Zhou
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Ping Dong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Shilei Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Ziyi Yang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Wei Gong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai, 200092, China; Research Institute of Biliary Tract Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
3
|
Gui J, Lin W, Meng C, Cui Y, Lan W, He J, Azad MAK, Kong X. Network pharmacology and molecular docking reveal the mechanism of Chinese herb ultrafine powder improving meat nutritional value in aged laying hens. Poult Sci 2025; 104:105047. [PMID: 40138971 PMCID: PMC11985111 DOI: 10.1016/j.psj.2025.105047] [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: 12/26/2024] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
This study investigated the effects of dietary Chinese herb ultrafine powder (CHUP) supplementation on meat quality, plasma biochemical parameters, and fatty acid and amino acid composition in pectoral muscles of aged laying hens. A total of 576 Xinyang black-feather laying hens (300-d-old) were randomly allocated to eight groups, including the control group (fed a basal diet) and different CHUP groups (details in 'Materials and methods' section). The trial lasted 120 d. The findings showed that L-LF and L-LF-T supplementation increased the contents of polyunsaturated fatty acids and unsaturated fatty acids (P < 0.05), while CHUP supplementation increased (P < 0.05) the total essential amino acid content in pectoral muscles. Network pharmacology analysis predicted that L-LF-T supplementation mainly influenced the PPAR signaling pathway, which is associated with meat quality. These findings suggest that CHUP supplementation can enhance the nutritional value of pectoral muscles, potentially through its association with the PPAR signaling pathway in aged laying hens.
Collapse
Affiliation(s)
- Jue Gui
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China
| | - Wenchao Lin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - Chengwen Meng
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China
| | - Yadong Cui
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Wei Lan
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
| | - M A K Azad
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China.
| | - Xiangfeng Kong
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100008, China; College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| |
Collapse
|
4
|
Hai Q, Wang Y, Li H, Pei H, Wang N, Zhang X, Fan M, Liao J, Wen W, Zhao J, Yang L, Cui H. Plantamajoside Promotes NGF/TrkA Pathway to Inhibit Neuronal Apoptosis and Improve Diabetic Peripheral Neuropathy. J Cell Mol Med 2025; 29:e70571. [PMID: 40289624 PMCID: PMC12034938 DOI: 10.1111/jcmm.70571] [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: 12/10/2024] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
The symptoms caused by diabetic peripheral neuropathy (DPN) have severely impacted patients' quality of life. While plantamajoside (PMS) exhibits neuroprotective properties, its efficacy and molecular mechanisms against DPN are unexplored. This study first established a high glucose (HG)-induced in vitro model of DPN and investigated the neuroprotective effects of PMS on RSC96 cells. We next demonstrated the anti-apoptotic effects of PMS and NGF/TrkA pathway mediated neurotrophic effects. Finally, we established a DPN mouse model and confirmed the therapeutic effects of PMS on DPN mice through behavioural tests and pathological staining, while also assessing the impact of PMS on the NGF/TrkA pathway and apoptosis. Our results showed that, in HG-induced DPN models, PMS enhanced cell viability while reducing LDH activity. Transcriptomics results indicated that the Apoptosis and Neurotrophins signalling pathways were key pathways for PMS on DPN. PMS treatment reduced HG-induced RSC96 cell apoptosis while enhancing NGF levels and upregulating NGF/TrkA-related protein expression. However, this protection was abolished by TrkA inhibitor or NGF neutralising antibodies. In vivo experimental results showed that PMS improved the mechanical pain threshold, thermal pain reaction time, and nerve conduction velocity of DPN mice. PMS improved pathological damage to the sciatic nerve, enhanced the number of Nissl bodies, reduced TUNEL-positive expression, and upregulated NGF levels. Furthermore, PMS reduced apoptosis and elevated NGF/TrkA-related protein expression in the sciatic nerve of DPN mice. In conclusion, PMS alleviates DPN through activating the NGF/TrkA pathway and inhibiting apoptosis.
Collapse
Affiliation(s)
- Qingshan Hai
- Nanjing University of Chinese MedicineNanjinChina
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Yuming Wang
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Hanzhou Li
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Huan Pei
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Ning Wang
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Xiaoxia Zhang
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Mingyao Fan
- Basic Medical SchoolYunnan University of Chinese MedicineKunmingChina
| | - Jiabao Liao
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Weibo Wen
- Nanjing University of Chinese MedicineNanjinChina
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Jie Zhao
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Ling Yang
- School of NursingYunnan University of Chinese MedicineKunmingChina
| | - Huantian Cui
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| |
Collapse
|
5
|
Ding XR, Zheng RF, Kaderyea K, Han YL, Wang SB, Xu L, Zeng X, Su WL, Wu LH, Xing JG. Chinese herbal formula Regan Saibisitan alleviates airway inflammation of chronic bronchitis via inhibiting the JAK2/STAT3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119336. [PMID: 39788165 DOI: 10.1016/j.jep.2025.119336] [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: 11/01/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Regan Saibisitan (RGS) is a classic prescription used to treat cough, pneumonia, and other respiratory infections in Uygur medicine. It is a granule composed of 12 kinds of medicinal materials. However, the mechanism by which RGS regulates lung disease remains unclear. AIM OF THE STUDY Chronic bronchitis (CB) is characterized by persistent, non-specific inflammation in the trachea, bronchial mucosa, and surrounding tissues mainly resulting from infectious or non-infectious factors. This study aimed to explore the function of RGS in alleviating airway inflammation associated with chronic bronchitis, and to examine the mechanisms by which RGS exerts its effects via the JAK 2/STAT 3 signaling pathway. MATERIALS AND METHODS The CB mouse model was established by cigarette smoking (CS) and intranasal administration of lipopolysaccharide (LPS, 20 μg), histological changes of bronchial epithelium, collagen deposition, mucus secretion in lung tissue and inflammatory factors were assayed. Transcriptomics analysis was performed to detect the differentially regulated genes in lung tissue of CB mice treated with RGS. The effect of RGS on JAK 2/STAT 3 pathway was investigated in CB mice and NCI-H292 cells treated with PMA using western blotting, ELISA, and immunohistochemical analysis. RESULTS RGS treatment significantly improved the thickening of bronchial epithelium, decreased collagen deposition and secretion of mucus, and the levels of inflammatory factors in CB mice. Transcriptomics analysis showed that most of 402 differentially expressed genes in RGS-treated CB mice were related to inflammatory response. The results in CB mice and NCI-H292 cells showed that RGS reduced the phosphorylation level of JAK 2 and STAT 3. In addition, the use of JAK 2 inhibitor AG490 confirmed that JAK 2/STAT 3 pathway played a key role in the effects of RGS on CB. CONCLUSIONS RGS suppresses inflammation and improves chronic bronchitis in NCI-H292 cells and CB mice, at least in part, via inhibiting the JAK 2/STAT 3 pathway. This study demonstrated that RGS could be a potential drug in treating CB disease.
Collapse
Affiliation(s)
- Xiao-Rui Ding
- Pharmacy School, Shihezi University, Xinjiang, 832000, China; Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China
| | - Rui-Fang Zheng
- Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China
| | - Kader Kaderyea
- Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China
| | - Yu-Lin Han
- Hetian Uygur Pharmaceutical Co., Ltd, 142 Hanggui Road, Hetian District, 848200, China
| | - Shou-Bao Wang
- Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China; Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lei Xu
- Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China
| | - Xin Zeng
- Pharmacy School, Shihezi University, Xinjiang, 832000, China; Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China
| | - Wen-Ling Su
- Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China.
| | - Le-He Wu
- Hetian Uygur Pharmaceutical Co., Ltd, 142 Hanggui Road, Hetian District, 848200, China.
| | - Jian-Guo Xing
- Pharmacy School, Shihezi University, Xinjiang, 832000, China; Xinjiang Key Laboratory of Uygur Medicine, Xinjiang Institute of Materia Medica, Xinjiang, 830000, China.
| |
Collapse
|
6
|
Zhou M, Niu H, Lu D, Zhang H, Luo D, Yu Z, Huang G, Li J, Xiong C, Tang Q, Zhang H, Liang F, Chen R. Wu Mei Wan suppresses colorectal cancer stemness by regulating Sox9 expression via JAK2/STAT3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:118998. [PMID: 39489363 DOI: 10.1016/j.jep.2024.118998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/22/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wu Mei Wan (WMW) is a traditional Chinese herbal formula with a long-standing history in Chinese medicine, valued for its therapeutic properties. However, its potential anti-cancer effects, especially against colorectal cancer (CRC), have not been fully elucidated. AIM OF THE STUDY This study aims to investigate the effects of WMW on colorectal cancer stemness and to elucidate the underlying molecular mechanisms, focusing on the modulation of Sox9 expression via the JAK2/STAT3 signaling pathway. MATERIALS AND METHODS WMW was prepared and analyzed using UPLC-MS to identify their main components. To study the therapeutic effects of WMW, AOM/DSS-induced CRC mouse models were established. A comprehensive suite of experimental techniques, including in vivo imaging, cell culture, transfection, CCK-8 assays, colony formation assays, wound healing assays, cell migration assays, Western blotting, dot blot analysis, RT-qPCR, immunohistochemistry, cell transcriptome sequencing, and gene set enrichment analysis, were utilized to explore the pharmacological effects and mechanisms of WMW. RESULTS WMW significantly inhibited CRC cell viability, proliferation, invasion, and migration in vitro. Mechanistically, WMW suppressed CRC stemness by downregulating Sox9 expression through the JAK2/STAT3 signaling pathway. Additionally, the regulation of methylation and demethylation mediated by TET1 and DNMT3a expression was directly associated with the JAK2/STAT3 pathway's modulation of Sox9 expression. In vivo, WMW treatment attenuated CRC progression and metastasis with minimal toxicity. CONCLUSION These findings suggest that WMW exerts potent anti-CRC stemness effects by regulating Sox9 via the JAK2/STAT3 signaling pathway, underscoring its potential as a promising therapeutic agent for CRC treatment.
Collapse
Affiliation(s)
- Minfeng Zhou
- Department of Integrative Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China
| | - Huifang Niu
- Jianghan University School of Medicine, 8 Triangle Lake Road, 430056, Wuhan City, China; Jianghan University Institute of Acupuncture and Moxibustion, 8 Triangle Lake Road, 430056, Wuhan City, China
| | - Damin Lu
- Hubei University of Chinese Medicine.Tanhualin one, Wuhan City, 430065, China
| | - Haiming Zhang
- Wuhan Central Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Nanjing Road. Wuhan City, 430014, China
| | - Dan Luo
- Wuhan Hospital of Integrated Chinese and Western Medicine Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Zhongshan Avenue. Wuhan City, 430033, China
| | - Zhaomin Yu
- Hubei Hospital of Integrated Traditional Chinese and Western Medicine, Diamond Horn Lake Road. Wuhan City, 430000, China
| | - Guichen Huang
- Department of Integrative Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China
| | - Jinxiao Li
- Department of Integrative Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China
| | - Chutong Xiong
- Department of Integrative Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China
| | - Qian Tang
- Hubei University of Chinese Medicine.Tanhualin one, Wuhan City, 430065, China
| | - Hongxing Zhang
- Jianghan University School of Medicine, 8 Triangle Lake Road, 430056, Wuhan City, China; Jianghan University Institute of Acupuncture and Moxibustion, 8 Triangle Lake Road, 430056, Wuhan City, China.
| | - Fengxia Liang
- Hubei University of Chinese Medicine.Tanhualin one, Wuhan City, 430065, China.
| | - Rui Chen
- Department of Integrative Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China.
| |
Collapse
|
7
|
Wang Y, Ji H, Chen G, Zhou J, Zhang D, Wang X. GNLY as A Novel Cis-eQTL and Cis-pQTL Mediated Susceptibility Gene in Suppressing Prostatitis. Mendelian Randomization Study. Arch Med Res 2025; 56:103098. [PMID: 39437618 DOI: 10.1016/j.arcmed.2024.103098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/24/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Prostatitis is characterized by high prevalence, low cure rates, and frequent recurrences, and remains one of the most clinically challenging problems. Hence, in this article, we first integrated Mendelian randomization (MR) with expression quantitative trait loci (eQTL) and protein quantitative trait loci (pQTL) data to identify novel therapeutic targets and their potential metabolic mechanisms for prostatitis. METHODS Prostatitis-related genetic data, eQTLs, pQTLs, and 1400 metabolites were downloaded from online databases. MR, or summary data-based MR (SMR) analyses were applied to assess the potential causal relationships between exposures and predicted outcomes. Sensitivity analysis was conducted using pleiotropy, heterogeneity, and leave-one-out analysis to evaluate the robustness of our results. RESULTS Based on our results, we first identified and validated GNLY as a novel cis-eQTL and cis-pQTL-mediated susceptibility gene for reducing prostatitis risk in five independent datasets (one discovery dataset and four validation datasets) (all p <0.05). Meanwhile, we also found that the GNLY eQTL could increase the metabolite of sphingomyelin level (d18:0/20:0, d16:0/22:0) risks (p <0.05), and the metabolite of sphingomyelin level (d18:0/20:0, d16:0/22:0) could reduce the risk of prostatitis (p <0.05). According to the above-mentioned relationships, we finally revealed the potential metabolic mechanism of GNLY eQTL in suppressing prostatitis via regulating the metabolite of sphingomyelin level (d18:0/20:0, d16:0/22:0). CONCLUSIONS We successfully identified GNLY as a novel cis-eQTL and cis-pQTL-mediated susceptibility gene in suppressing prostatitis and its potential metabolic mechanism via regulating sphingomyelin (d18:0/20:0, d16:0/22:0) levels, providing a novel therapeutic target and paving the way for future GNLY-related studies in prostatitis.
Collapse
Affiliation(s)
- Yi Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hao Ji
- Department of Urology, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu Province, China
| | - Guihua Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianhua Zhou
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Dongliang Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Tao S, Wang H, Ji Q, Yang Y, Wei G, Li R, Zhou B. Integration of Metabolomics and Transcriptomics to Reveal the Antitumor Mechanism of Dendrobium officinale Polysaccharide-Based Nanocarriers in Enhancing Photodynamic Immunotherapy in Colorectal Cancer. Pharmaceutics 2025; 17:97. [PMID: 39861745 PMCID: PMC11769008 DOI: 10.3390/pharmaceutics17010097] [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: 11/02/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Background: The mechanism of Dendrobium officinale polysaccharide-based nanocarriers in enhancing photodynamic immunotherapy in colorectal cancer (CRC) remains poorly understood. Methods: The effects of TPA-3BCP-loaded cholesteryl hemisuccinate-Dendrobium officinale polysaccharide nanoparticles (DOP@3BCP NPs) and their potential molecular mechanism of action in a tumor-bearing mouse model of CRC were investigated using non-targeted metabolomics and transcriptomics. Meanwhile, a histopathological analysis (H&E staining, Ki67 staining, and TUNEL assay) and a qRT-PCR analysis revealed the antitumor effects of DOP@3BCP NPs with and without light activation. Results: Through metabolomics and transcriptomics analysis, we found an alteration in the metabolome and functional pathways in the examined tumor tissues. The metabolic analysis showed 69 and 60 differentially expressed metabolites (DEMs) in positive- and negative-ion modes, respectively, in the treated samples compared to the Control samples. The transcriptomics analysis showed that 1352 genes were differentially expressed among the three groups. The differentially regulated functional pathways were primally related to the antitumor immune response. The results of the pathological histology assay and qRT-PCR analysis verified the findings of the integrated metabolomics and transcriptomics analysis. Conclusions: Overall, our findings elucidate the potential antitumor mechanisms of the D. officinale polysaccharide-based nanocarrier in enhancing photodynamic immunotherapy in CRC.
Collapse
Affiliation(s)
- Shengchang Tao
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Huan Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (H.W.); (G.W.)
| | - Qiufeng Ji
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Yushan Yang
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Gang Wei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; (H.W.); (G.W.)
| | - Ruiming Li
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Benjie Zhou
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China; (S.T.); (Q.J.); (Y.Y.)
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|