1
|
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
|
2
|
Wei X, Leng X, Liang J, Liu J, Chi L, Deng H, Sun D. Pharmacological potential of natural medicine Astragali Radix in treating intestinal diseases. Biomed Pharmacother 2024; 180:117580. [PMID: 39413615 DOI: 10.1016/j.biopha.2024.117580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/07/2024] [Accepted: 10/14/2024] [Indexed: 10/18/2024] Open
Abstract
Due to changes in diet and lifestyle, the prevalence of intestinal diseases has been increasing annually. Current treatment methods exhibit several limitations, including adverse reactions and drug resistance, necessitating the development of new, safe, and effective therapies. Astragali Radix, a natural medicine utilized for over two millennia, offers unique advantages in treating intestinal ailments due to its multi-component and multi-target properties. This study aims to review the effective components of Astragali Radix that provide intestinal protection and to explore its pharmacological effects and molecular mechanisms across various intestinal diseases. This will provide a comprehensive foundation for using Astragali Radix in treating intestinal diseases and serve as a reference for future research directions. The active components of Astragali Radix with protective effects on the intestines include astragaloside (AS)-IV, AS-III, AS-II, astragalus polysaccharide (APS), cycloastagenol, calycosin, formononetin, and ononin. Astragali Radix and its active components primarily address intestinal diseases such as colorectal cancer (CRC), inflammatory bowel disease (IBD), and enterocolitis through mechanisms including anti-inflammatory actions, antioxidative stress responses, anti-proliferation and invasion activities, regulation of programmed cell death, immunoregulation, restoration of the intestinal epithelial barrier, and modulation of the intestinal microbiota and its metabolites. Consequently, Astragali Radix demonstrates significant intestinal protective activity and represents a promising natural treatment for intestinal diseases. However, the pharmacological actions and mechanisms of some active components in Astragali Radix remain unexplored. Moreover, further comprehensive toxicological and clinical studies are required to ascertain its safety and clinical effectiveness.
Collapse
Affiliation(s)
- Xiunan Wei
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Xiaohui Leng
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Junwei Liang
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Jiahui Liu
- Department of Gastroenterology, Shandong Provincial Third Hospital, Jinan 250014, China.
| | - Lili Chi
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Hualiang Deng
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Dajuan Sun
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
3
|
Wubuli A, Chai J, Liu H, Nijat D, Li J, Xia G, Cao Q, Zhang S, Huang W, Aipire A, Li J. In vivo pharmacokinetics of Glycyrrhiza uralensis polysaccharides. Front Pharmacol 2024; 15:1431221. [PMID: 39101144 PMCID: PMC11294697 DOI: 10.3389/fphar.2024.1431221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/24/2024] [Indexed: 08/06/2024] Open
Abstract
Glycyrrhiza uralensis polysaccharides (GUPS) are widely applied in biomedicine and functional food due to their multiple pharmacological activities and low toxicity. Despite their widespread use, the in vivo metabolic profile of GUPS remains poorly understood. To address this gap, we developed a quantitative analysis method that involves labeling GUPS with visible fluorescein (5-DTAF) and near-infrared (NIR) fluorescein (Cy7), resulting in stable conjugates with substitution degrees of 0.81% for 5-DTAF and 0.39% for Cy7. The pharmacokinetic studies showed a biphasic elimination pattern in the blood concentration-time curve following both intravenous and oral administration, consistent with a two-compartment model. Using fluorescence quantification and NIR imaging, we observed that GUPS was distributed to various tissues, exhibiting higher concentrations particularly in liver, kidney and lung. Excretion studies indicated that feces were the major excretion pathway of GUPS after oral administration (60.98%), whereas urine was the main pathway after intravenous administration (31.16%). Notably, GUPS could be absorbed rapidly by gut (Tmax 1 ± 0.61 h) and showed a biological half-time t1/2 26.4 ± 7.72 h after oral administration. Furthermore, the Caco-2 cells uptake studies illustrated that macropinocytosis and clathrin-mediated endocytosis were participated in the transport of GUPS in intestine epithelium. This comprehensive analysis of the in vivo pharmacokinetics of GUPS not only enhances our understanding of its metabolic pathways but also establishes a foundational basis for its clinical application, optimizing its therapeutic potential and safety profile.
Collapse
Affiliation(s)
- Abudukahaer Wubuli
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Junwei Chai
- Urumqi Xinze Ziqi Biotechnology Company, Limited, Urumqi, China
| | - Haoqiang Liu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Dilaram Nijat
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jianmin Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Guoyu Xia
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Qi Cao
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Saidan Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | | | - Adila Aipire
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| |
Collapse
|
4
|
Chen X, Wei DD, Lin M, Wang XS, Kang HJ, Ni L, Qian DW, Guo S, Duan JA. Comparative evaluation of four Lycium barbarum cultivars on NaIO 3-induced retinal degeneration mice via multivariate statistical analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117889. [PMID: 38336183 DOI: 10.1016/j.jep.2024.117889] [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: 11/01/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The fruit of Lycium barbarum L. (goji berry) is a traditional Chinese medicine and is often used to improve vision. While various goji cultivars may differentially treat retinal degeneration, however their comparative effectiveness remains unclear. AIM OF THE STUDY To evaluate the protective effects of four goji cultivars on NaIO3-induced retinal degeneration mouse model and identify the most therapeutically potent cultivar. MATERIALS AND METHODS The principal compounds in the extracts of four goji cultivars were characterized by UPLC-Q-TOF/MS. A retinal degeneration mouse model was established via NaIO3 injection. Dark-light transition and TUNEL assays were used to assess visual function and retinal apoptosis. The levels of antioxidative, inflammatory, and angiogenic markers in serums and eyeballs were measured. Hierarchical cluster analysis, principal component analysis and partial least squares-discriminant analysis were used to objectively compare the treatment responses. RESULTS Sixteen compounds were identified in goji berry extracts. All goji berry extracts could reverse NaIO3-induced visual impairment, retinal damage and apoptosis. The samples from the cultivar of Ningqi No.1 significantly modulated oxidative stress, inflammation, and vascular endothelial growth factor levels, which are more effectively than the other cultivars based on integrated multivariate profiling. CONCLUSION Ningqi No.1 demonstrated a stronger protective effect on mouse retina than other goji cultivars, and is a potential variety for further research on the treatment of retinal degeneration.
Collapse
Affiliation(s)
- Xin Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization/ National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Dan-Dan Wei
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization/ National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ming Lin
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization/ National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xue-Sen Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization/ National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Jie Kang
- Ningxia Innovation Center of Goji R & D, Yinchuan, 750002, China
| | - Liang Ni
- Guizhou Tongde Pharmaceutical Co., Ltd, Tongren, 554300, China
| | - Da-Wei Qian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization/ National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Sheng Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization/ National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization/ National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
5
|
Wang J, Zhu K, Zhang M, Zhou Q, Ji W, Yao Z, Li D. Pharmacokinetics, tissue distribution, and subacute toxicity of oral carrageenan in mice. Int J Biol Macromol 2024; 266:130725. [PMID: 38490394 DOI: 10.1016/j.ijbiomac.2024.130725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
Carrageenan (CGN) is a typical sulfated polysaccharide widely applied in the food and pharmaceutical industries. Its in vivo behavior plays vital roles in understanding structural and biological functional relationships. The lack of UV chromophores in highly sulfated polysaccharides presents a challenge for their in vivo behavior studies. Therefore, this study aimed to develop a fast and effective quantitative fluorescence method for investigating the pharmacokinetics and tissue distribution of CGN. Fluorescence isothiocyanate labeling of CGN (FCGN) and microplate reader-based measurements were developed and validated to study its pharmacokinetics. These results showed that the FCGN concentration peaked at 3 h, the mean residence time was 36.6 h, and the clearance rate was 0.1 L/h/kg. Most of the FCGN was excreted in the feces, while 9.2 % was excreted in the urine, suggesting absorption and metabolism. The pharmacokinetic parameters indicated that the FCGN was absorbed quickly, eliminated slowly, and could remain in the body for a sustained profile. Moreover, ex vivo imaging and quantification of FCGN in tissues revealed that FCGN accumulated in the liver and kidney. Furthermore, oral administration of CGN or KOs for 14 days led to changes in liver and kidney indices. Histological analysis of significant organs revealed hepatocyte necrosis in the liver, renal tubular vacuolization in the kidney, and incomplete colonic epithelial cells. The KOs had a more significant effect on inflammatory cell infiltration than did CGNs. These in vivo findings laid the foundation for the study and application of CGN in food and pharmaceutical applications.
Collapse
Affiliation(s)
- Jiahui Wang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Kehan Zhu
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Miaomiao Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Qian Zhou
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Wen Ji
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Zhen Yao
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China
| | - Duxin Li
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
6
|
Zhang Y, Zhang XY, Shi SR, Ma CN, Lin YP, Song WG, Guo SD. Natural products in atherosclerosis therapy by targeting PPARs: a review focusing on lipid metabolism and inflammation. Front Cardiovasc Med 2024; 11:1372055. [PMID: 38699583 PMCID: PMC11064802 DOI: 10.3389/fcvm.2024.1372055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Inflammation and dyslipidemia are critical inducing factors of atherosclerosis. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors and control the expression of multiple genes that are involved in lipid metabolism and inflammatory responses. However, synthesized PPAR agonists exhibit contrary therapeutic effects and various side effects in atherosclerosis therapy. Natural products are structural diversity and have a good safety. Recent studies find that natural herbs and compounds exhibit attractive therapeutic effects on atherosclerosis by alleviating hyperlipidemia and inflammation through modulation of PPARs. Importantly, the preparation of natural products generally causes significantly lower environmental pollution compared to that of synthesized chemical compounds. Therefore, it is interesting to discover novel PPAR modulator and develop alternative strategies for atherosclerosis therapy based on natural herbs and compounds. This article reviews recent findings, mainly from the year of 2020 to present, about the roles of natural herbs and compounds in regulation of PPARs and their therapeutic effects on atherosclerosis. This article provides alternative strategies and theoretical basis for atherosclerosis therapy using natural herbs and compounds by targeting PPARs, and offers valuable information for researchers that are interested in developing novel PPAR modulators.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Endocrinology and Metabolism, Guiqian International General Hospital, Guiyang, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Chao-Nan Ma
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Yun-Peng Lin
- Department of General Surgery, Qixia Traditional Chinese Medicine Hospital in Shandong Province, Yantai, China
| | - Wen-Gang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| |
Collapse
|
7
|
Zhang Y, Wang H, Zheng Y, Wu Z, Liu J, Cheng F, Wang K. Degradation of Angelica sinensis polysaccharide: Structures and protective activities against ethanol-induced acute liver injury. Carbohydr Polym 2024; 328:121745. [PMID: 38220331 DOI: 10.1016/j.carbpol.2023.121745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/16/2024]
Abstract
Angelica sinensis polysaccharide (ASP) possesses diverse bioactivities; however, its metabolic fate following oral administration remains poorly understood. To intuitively determine its intestinal digestion behavior after oral administration, ASP was labeled with fluorescein, and it was found to accumulate and be degraded in the cecum and colon. Therefore, we investigated the in vitro enzymatic degradation behavior and identified the products. The results showed that ASP could be degraded into fragments with molecular weights similar to those of the fragments observed in vivo. Structural characterization revealed that ASP is a highly branched acid heteropolysaccharide with AG type II domains, and its backbone is predominantly composed of 1,3-Galp, →3,6)-Galp-(1→6)-Galp-(1→, 1,4-Manp, 1,4-Rhap, 1,3-Glcp, 1,2,3,4-Galp, 1,3,4,6-Galp, 1,3,4-GalAp and 1,4-GlcAp, with branches of Araf, Glcp and Galp. In addition, the high molecular weight enzymatic degradation products (ASP H) maintained a backbone structure almost identical to that of ASP, but exhibited only partial branch changes. Then, the results of ethanol-induced acute liver injury experiments revealed that ASP and ASP H reduced the expression of aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase (LDH), and malondialdehyde (MDA) and increased the superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) levels, thereby relieving ethanol-induced acute liver injury.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China
| | - Haoyu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China
| | - Yuheng Zheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China
| | - Zhijing Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China
| | - Junxi Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China
| | - Fang Cheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, PR China.
| | - Kaiping Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, PR China.
| |
Collapse
|
8
|
Wu Y, Yin W, Hao P, Chen Y, Yu L, Yu X, Wu Y, Li X, Wang W, Zhou H, Yuan Y, Quan X, Yu Y, Hu B, Chen S, Zhou Z, Sun W. Polysaccharide from Panax japonicus C.A. Mey prevents non-alcoholic fatty liver disease development based on regulating liver metabolism and gut microbiota in mice. Int J Biol Macromol 2024; 260:129430. [PMID: 38228199 DOI: 10.1016/j.ijbiomac.2024.129430] [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: 10/23/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024]
Abstract
In this study, a new polysaccharide (PSPJ) with specific molecular weight and monosaccharide compositions was isolated and purified from the water extract of Panacis Japonici Rhizoma (PJR). 16S rRNA analysis and untargeted metabolomic analysis were used to assess PSPJ's efficacy in averting non-alcoholic fatty liver disease (NAFLD). This study indicated that PSPJ significantly reduced liver fat accumulation, the increase in blood lipids and ALT caused by HFD, indicating that PSPJ can prevent NAFLD. We demonstrated through cell experiments that PSPJ does not directly affect liver cells. The gut microbiota disorder and alterations in short-chain fatty acids (SCFAs) induced by the high-fat diet (HFD) were ameliorated by PSPJ, as evidenced by the analysis of 16S rRNA. In particular, supplementing PSPJ reduced the abundance of Turicibacter, Dubosiella, and Staphylococcus, and increased the abundance of Bacteroides, Blautia, and Lactobacillus. Untargeted metabolomic analysis shows that PSPJ improves liver metabolic disorders by regulating arachidonic acid metabolism, carbohydrate digestion and absorption, fatty acid biosynthesis, fatty acid metabolism and retinol metabolism. The findings of our investigation indicate that PSPJ has the potential to modulate liver metabolism through alterations in the composition of intestinal bacteria, hence preventing NAFLD.
Collapse
Affiliation(s)
- Yi Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Wen Yin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ping Hao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yueru Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lingyun Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xingjian Yu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento 95817, CA, United States of America
| | - Yu Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaocong Li
- College of Medicine, Hubei Three Gorges Polytechnic, No.31 Stadium Road, Yichang 443000, China
| | - Wenjia Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; College of Animal Science and Technology, Ningxia University, China
| | - Hui Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuan Yuan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoyu Quan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yue Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bing Hu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shouhai Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhenlei Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Wenjing Sun
- Guangxi Key Laboratory of Agricultural Resources Chemistry and Biotechnology, College of Biology & Pharmacy, Yulin Normal University, No. 1303 Jiaoyu East Road, Yulin 537000, Guangxi, China.
| |
Collapse
|
9
|
Lu Y, Lin M, Ou S, Sun L, Qian K, Kuang H, Wu Y. Astragalus polysaccharides ameliorate epileptogenesis, cognitive impairment, and neuroinflammation in a pentylenetetrazole-induced kindling mouse model. Front Pharmacol 2024; 15:1336122. [PMID: 38405667 PMCID: PMC10884767 DOI: 10.3389/fphar.2024.1336122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/31/2024] [Indexed: 02/27/2024] Open
Abstract
Background: Epilepsy is a prevalent neurological disease where neuroinflammation plays a significant role in epileptogenesis. Recent studies have suggested that Astragalus polysaccharides (APS) have anti-inflammatory properties, which make them a potential candidate for neuroprotection against central nervous system disease. Nevertheless, the extent of their effectiveness in treating epilepsy remains enigmatic. Therefore, our study aims to investigate the potential of APS to mitigate epileptogenesis and its comorbidities by exploring its underlying mechanism. Methods: Initially, we employed pentylenetetrazol-induced seizure mice to validate APS' effectiveness. Subsequently, we employed network pharmacology analysis to probe the possible targets and signaling pathways of APS in treating epilepsy. Ultimately, we verified the key targets and signaling pathways experimentally, predicting their mechanisms of action. Results: APS have been observed to disturb the acquisition process of kindling, leading to reduced seizure scores and a lower incidence of complete kindling. Moreover, APS has been found to improve cognitive impairments and prevent hippocampal neuronal damage during the pentylenetetrazole (PTZ)-kindling process. Subsequent network pharmacology analysis revealed that APS potentially exerted their anti-epileptic effects by targeting cytokine and toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB) signaling pathways. Finally, experimental findings showed that APS efficiently inhibited the activation of astrocytes and reduced the release of pro-inflammatory mediators, such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). In addition, APS impeded the activation of the TLR4/NF-κB signaling cascade in a PTZ-induced kindling mouse model. Conclusion: The outcomes of our study suggest that APS exerts an impact on epileptogenesis and mitigates cognitive impairment by impeding neuroinflammatory processes. The mechanism underlying these observations may be attributed to the modulation of the TLR4/NF-κB signaling pathway, resulting in a reduction of the release of inflammatory mediators. These findings partially agree with the predictions derived from network pharmacology analyses. As such, APS represents a potentially innovative and encouraging adjunct therapeutic option for epileptogenesis and cognitive deficit.
Collapse
Affiliation(s)
- Yuling Lu
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Minglin Lin
- Department of Colorectal and Anal Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Sijie Ou
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Lanfeng Sun
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Kai Qian
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Huimin Kuang
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yuan Wu
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
10
|
Xu J, Yu Y, Chen K, Wang Y, Zhu Y, Zou X, Xu X, Jiang Y. Astragalus polysaccharides ameliorate osteoarthritis via inhibiting apoptosis by regulating ROS-mediated ASK1/p38 MAPK signaling pathway targeting on TXN. Int J Biol Macromol 2024; 258:129004. [PMID: 38151083 DOI: 10.1016/j.ijbiomac.2023.129004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
This research aims to explore the potential of astragalus polysaccharides (APS) in treating osteoarthritis. The primary component of APS extracted in this study was glucose, and noticeably it had a relatively high content of glucuronic acids. In vitro, APS reduced ROS levels, protected chondrocytes from apoptosis, and promoted collagen II expression by regulating ASK1 (apoptosis-signal-regulating kinase1)/p38 cell apoptosis pathway. Further co-immunoprecipitation and immunofluorescence localization experiments demonstrated that the thioredoxin (TXN) antioxidant system was responsible for its bioactivity. Moreover, TXN silencing remarkably blocked the protective effects of APS, indicating that APS inhibited chondrocyte apoptosis by targeting TXN. In vivo, APS effectively mitigated cartilage loss and chondrocyte apoptosis and decreased expressions of p-ASK1 and p-p38. Collectively, this research first demonstrated that APS could ameliorate osteoarthritis by ASK1/p38 signaling pathway through regulating thioredoxin. In conclusion, APS holds promise as a nutraceutical supplement for osteoarthritis in future drug development.
Collapse
Affiliation(s)
- Jintao Xu
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yaohui Yu
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kai Chen
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yishu Wang
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Zhu
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiangjie Zou
- Jiangsu Province Hospital, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xianghong Xu
- Department of Endocrinology, Nanjing First Hospital, Nanjing, China
| | - Yiqiu Jiang
- Department of Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|