1
|
Zheng Q, Wang T, Wang S, Chen Z, Jia X, Yang H, Chen H, Sun X, Wang K, Zhang L, Fu F. The anti-inflammatory effects of saponins from natural herbs. Pharmacol Ther 2025; 269:108827. [PMID: 40015518 DOI: 10.1016/j.pharmthera.2025.108827] [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: 04/29/2024] [Revised: 11/20/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Inflammation is a protective mechanism that also starts the healing process. However, inflammatory reaction may cause severe tissue damage. The increased influx of phagocytic leukocytes may produce excessive amount of reactive oxygen species, which leads to additional cell injury. Inflammatory response activates the leukocytes and thus induces tissue damage and prolongs inflammation. The inflammation-induced activation of the complement system may also contribute to cell injury. Non-steroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids are chief agents for treating inflammation associated with the diseases. However, the unwanted side effects of NSAIDs (e.g., gastrointestinal disturbances, skin reactions, adverse renal effects, cardiovascular side effects) and glucocorticoids (e.g., suppression of immune system, Cushing's syndrome, osteoporosis, hyperglycemia) limit their use in patients. Natural herbs are important sources of anti-inflammatory drugs. The ingredients extracted from natural herbs display anti-inflammatory effects to work through multiple pathways with lower risk of adverse reaction. At present, the main anti-inflammatory natural agents include saponins, flavonoids, alkaloids, polysaccharides, and so on. The present article will review the anti-inflammatory effects of saponins including escin, ginsenosides, glycyrrhizin, astragaloside, Panax notoginseng saponins, saikosaponin, platycodin, timosaponin, ophiopogonin D, dioscin, senegenin.
Collapse
Affiliation(s)
- Qinpin Zheng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Sensen Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Zhuoxi Chen
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Xue Jia
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Hui Yang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Huijin Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Xin Sun
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Kejun Wang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Leiming Zhang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China.
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China.
| |
Collapse
|
2
|
Liu HX, Li YC, Su RB, Liu CX, Wen SY. Astragalus injection inhibits the growth of osteosarcoma by activating cytotoxic T lymphocyte and targeting CTSL. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119607. [PMID: 40058477 DOI: 10.1016/j.jep.2025.119607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus membranaceus, commonly known as Huangqi in China, is a traditional herbal medicine that has attracted significant attention for its immunomodulatory effects. It has been widely studied in various clinical contexts, including cancer treatment. Astragalus injection (HQI) is clinically used for treating myocarditis and cardiac insufficiency. However, its potential therapeutic effects on osteosarcoma, a highly aggressive bone tumor, remain largely unexplored. AIM OF THE STUDY The aim of this study was to investigate the potential therapeutic effects of HQI on osteosarcoma and to elucidate its underlying mechanisms of action. Specifically, we aimed to determine whether HQI could inhibit osteosarcoma growth in vivo, identify its key active components and molecular targets, and explore its immunomodulatory effects on the tumor microenvironment. MATERIALS AND METHODS Mice with osteosarcoma were treated with HQI, and tumor growth was monitored. The number of CD8+ T cells in spleen was assessed using flow cytometry. High-performance liquid chromatography coupled with electrospray ionization time-of-flight mass spectrometry (HPLC-ESI-TOF-MS/MS) was used to identify the active ingredients of HQI that entered the peripheral blood of treated mice. Network pharmacology and weighted gene co-expression network analysis (WGCNA) were employed to identify key molecular targets of HQI in osteosarcoma inhibition. In vitro cell assays were conducted to evaluate the effects of HQI and its active components on osteosarcoma cell viability. Molecular docking studies were performed to identify the binding affinity of key active components to the identified molecular targets. The expression of Cathepsin L (CTSL) and the activation of cytotoxic T lymphocytes were assessed in vivo and in vitro to elucidate the primary mechanism of action of HQI and its active component calycosin 7-O-β-D-glucoside (CG). RESULTS Our study found that HQI significantly suppresses osteosarcoma growth in vivo by increasing the number of CD8+ T cells, without causing significant toxic side effects. Eight active ingredients entered the peripheral blood of mice through HPLC-ESI-TOF-MS/MS detection. The network pharmacology and WGCNA revealed that CTSL was a key target of HQI in osteosarcoma inhibition. Cell assays and molecular docking identified CG as the key active component of HQI to inhibit the activity of osteosarcoma cells, capable of binding to CTSL. In vivo, CG activates cytotoxic T lymphocytes and inhibits CTSL expression, thereby exerting its anti-osteosarcoma effects. CONCLUSION Our study demonstrated that HQI, particularly its active component CG, holds potential as a therapeutic agent for osteosarcoma. The primary mechanism underlying its anti-osteosarcoma effects involves modulating the immune response and targeting CTSL. These findings provide a scientific basis for the development of HQI as a novel immunomodulatory therapy for osteosarcoma.
Collapse
Affiliation(s)
- Hai-Xin Liu
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China.
| | - Yu-Chang Li
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China.
| | - Ru-Bin Su
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China.
| | - Cai-Xia Liu
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China.
| | - Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
3
|
Wang H, Huang Z, Chen G, Li Y, Liu Y, Gu H, Cao Y. Astragaloside IV alleviated bone loss in mice with ovariectomy-induced osteoporosis via modulating gut microbiota and fecal metabolism. Front Pharmacol 2025; 16:1548491. [PMID: 40248089 PMCID: PMC12003300 DOI: 10.3389/fphar.2025.1548491] [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: 12/19/2024] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Background Astragaloside IV (AS-IV) is one of the most potent components of Astragalus. It has been reported to promote bone formation and inhibit osteoclastogenesis, suggesting its potential as a candidate for the prevention and treatment of postmenopausal osteoporosis (PMOP). The gut microbiota may play a crucial role in mediating the effects of AS-IV. Objective To investigate the impact of gut microbiota on the efficacy of AS-IV in treating PMOP. Methods Mice were randomly divided into three groups: Sham, ovariectomy (OVX), and AS-IV-treated OVX group (80 mg/kg). Bone loss was evaluated using Micro-CT and histopathology. Immunohistochemistry assessed specific bone markers. Inflammatory levels were measured by enzyme-linked immunosorbent assay (ELISA). Intestinal barrier function was examined via colonic histopathology and immunohistochemistry. Gut microbiota composition was analyzed by 16S rDNA sequencing, while metabolomic profiling identified key metabolites. Correlation analysis was performed to explore relationships between differential bacteria, key metabolites, and bone loss. Results AS-IV improved the femur microarchitecture and modulated bone turnover in OVX mice. AS-IV treatment strengthened the intestinal barrier function and decreased gut permeability. This compound reduced colonic oxidative stress and serum and bone marrow inflammatory cytokine production. 16S rDNA sequencing revealed that AS-IV modulated the gut microbiota composition, while metabolomic analysis showed its effects on pathways related to hormone biosynthesis, D-amino acid metabolism, and galactose metabolism. Conclusion This study provides new insights into the use of AS-IV for treating PMOP, highlighting the gut microbiota and its metabolites as key regulatory factors in AS-IV's therapeutic effects.
Collapse
Affiliation(s)
- Huichao Wang
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Orthopedic Institute of Henan Province, Luoyang, Henan, China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, China
| | - Guangnan Chen
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, China
| | - Yang Li
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Emergency Trauma Center, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Youwen Liu
- Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Orthopedic Institute of Henan Province, Luoyang, Henan, China
| | - Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, China
| | - Yujing Cao
- School of Osteopathy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Emergency Trauma Center, Henan Provincial Hospital of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Zhao D, Ge A, Yan C, Liu X, Yang K, Yan Y, Hao M, Chen J, Daga P, Dai CC, Li C, Cao H. T helper cell 17/regulatory T cell balance regulates ulcerative colitis and the therapeutic role of natural plant components: a review. Front Med (Lausanne) 2025; 11:1502849. [PMID: 40196424 PMCID: PMC11973383 DOI: 10.3389/fmed.2024.1502849] [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: 09/27/2024] [Accepted: 12/23/2024] [Indexed: 04/09/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic relapsing inflammatory disease characterized by progressive mucosal damage. The incidence rate of UC is rising rapidly, which makes the burden of medical resources aggravated. In UC, due to various pathogenic factors such as mucosal immune system disorders, gene mutations and environmental factors disrupting the mucosal barrier function, the midgut pathogenic bacteria and exogenous antigens translocate into the lamina propria, thereby aggravating the inflammatory response and further damages the mucosal barrier. During the progression of UC, Th17 populations that cause inflammation generally increase, while Tregs that suppress Th17 activity decrease. Among them, Th17 mediates immune response, Treg mediates immunosuppression, and the coordinated balance of the two plays a key role in the inflammation and immune process of UC. Natural plant components can regulate biological processes such as immune inflammation from multiple levels of proinflammatory cytokines and signaling pathways. These characteristics have unique advantages and broad prospects in the treatment of UC. In immunomodulation, there is substantial clinical and experimental evidence for the modulatory role of natural plant products in restoring balance between Th17/Treg disturbances in UC. This review summarizes the previous studies on the regulation of Th17/Treg balance in UC by natural plant active ingredients, extracts, and traditional Chinese medicine prescriptions, and provides new evidence for the development and design of lead compounds and natural new drugs for the regulation of Th17/Treg balance in the future, and then provides ideas and evidence for future clinical intervention in the treatment of UC immune disorders and clinical trials.
Collapse
Affiliation(s)
- Da Zhao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Cong Yan
- Department of Urology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan Children’s Hospital), Changsha, China
| | - Xingci Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Kailin Yang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Psychology, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Yexing Yan
- Department of Psychology, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Department of Psychology, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Junpeng Chen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, United States
- Center for Cardiometabolic Science, Division of Environmental Medicine, Christina Lee Brown Envirome Insttitute, University of Louisville, Louisville, KY, United States
| | - Pawan Daga
- Department of Internal Medicine, University of Louisville, Louisville, KY, United States
| | - Charles C. Dai
- Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Maryland Baltimore, Baltimore, MD, United States
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, James Clark Hall, College Park, MD, United States
| | - Changping Li
- School of Mechanical Engineering and Automation, Fuyao University of Science and Technology, Fuzhou, China
| | - Hui Cao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
5
|
Xu P, Qian Y, Xu G, Chu J, He B. Fructosyl-mangiferin ameliorates dextran sulfate sodium-induced colitis in mice via the STAT3/M1/Th17 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156475. [PMID: 39933469 DOI: 10.1016/j.phymed.2025.156475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Inflammatory bowel disease (IBD), a chronic inflammatory condition categorized into ulcerative colitis (UC) and Crohn's disease (CD), affects a growing global patient population. Despite the prevalence, clinically there is a scarcity of effective therapeutic agents. PURPOSE This study investigated the therapeutic effects of fructosyl mangiferin (FM) on UC and elucidated its underlying mechanisms through in vivo and in vitro experiments. METHODS In vivo, a UC model of C57BL/6J mice was established via dextran sulfate sodium (DSS) induction, and the therapeutic effects were assessed through intragastric administration. In vitro, the murine macrophage cell line RAW264.7 was stimulated with lipopolysaccharide (LPS) to establish an M1 polarization model and introduced to explore the role of FM in immune cells. Molecular docking was further employed to investigate the specific molecular mechanisms of FM. RESULTS In vivo experimental findings indicate that FM, like mangiferin (M), preserves mucin secretion and the expression of occludin protein, and both significantly impede the progression of fibrosis associated with colitis. Additionally, FM effectively suppresses M1 macrophage polarization and exerts a pronounced inhibitory effect on the adaptive immune response, outperforming M in mitigating UC. In vitro results corroborate FM's inhibitory action on M1 polarization. Molecular docking studies identified FM as a potential signal transducer and activator of transcription 3 (STAT3) inhibitor, aligning with western blot analyses from both in vivo and in vitro experiments. CONCLUSION In conclusion, following fructosylation, FM exhibits remarkable anti-inflammatory and colonic protective effects. FM's ability to control the progression of UC offers a novel strategy for its potential treatment, warranting further investigation into its clinical application.
Collapse
Affiliation(s)
- Penghong Xu
- School of Pharmaceutical Sciences, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Yuping Qian
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Guo Xu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China
| | - Jianlin Chu
- School of Pharmaceutical Sciences, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China.
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China; College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, 30 Puzhunan Road, Jiangbei New Area, Nanjing 211800, China.
| |
Collapse
|
6
|
Wang F, Zhu Y, Shu H, Zhang X, Duan L, Man D, Wang Y. Astragaloside IV alleviates GDM via regulating gut microbiota and gut microbiota metabolomic. Front Pharmacol 2025; 15:1431240. [PMID: 39885928 PMCID: PMC11780255 DOI: 10.3389/fphar.2024.1431240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/11/2024] [Indexed: 02/01/2025] Open
Abstract
Background Gestational diabetes mellitus (GDM), a severe pregnancy disorder, is a temporary form of diabetes that occurs during gestation. Astragaloside IV (AS IV), a natural and effective composition of Astragalus membranaceus, shows pharmacological effects against diabetes. On the contrary, the effects of AS IV on GDM development are still not clear. This study aims to investigate the role of AS IV in alleviating GDM in rats and determine whether AS IV exerts its anti-GDM properties through the regulation of gut microbiota and metabolite modulation. Methods There were six pregnant SD rats in each of the four groups. First, the GDM model was induced by the streptozotocin (STZ, 45 mg/kg) injection on gestational days (GDs) 1-4, and AS IV intervention (10 mg/kg/d) was administered from 6 days before pregnancy until delivery. The measurements of relevant indicators pertaining to GDM symptoms and reproductive outcomes, along with the 16S rRNA sequencing data and LC-MS-based metabolomic profiles, were assessed across all groups. Results After the 25-day intervention, the GDM model + AS IV group showed significantly decreased fasting blood glucose levels (p = 0.0003), mean insulin levels (p = 0.0001), and insulin resistance index (p = 0.0001). AS IV treatment also decreased the malformation rate (p = 0.0373) and increased the average fetal weight (p = 0.0020) of GDM rats. Compared to the control rats, GDM rats showed a significantly higher abundance of Blautia and Anaerobiospirillum. However, the dramatically elevated abundance of these microorganisms was markedly decreased by AS IV treatment. In contrast, compared to GDM rats without treatment, GDM rats treated with AS IV showed a significantly higher abundance of bacteria (p < 0.05), such as Methanobrevibacter, Dubosiella, and Romboutsia, which are beneficial to the rats. Additionally, we observed dramatically elevated production of metabolites, such as N-acetyl-l-leucine and lithocholic acid, after AS IV treatment through metabolomics analysis (p < 0.05). Furthermore, significant associations between most genera of gut bacteria and the altered levels of the metabolites connected to gut microbiota were also discovered. Conclusion Our study demonstrated that AS IV could be an effective nutritional intervention strategy for targeting gut microbiota and metabolome profiles in GDM and provided experimental evidence supporting the use of AS IV to treat GDM.
Collapse
Affiliation(s)
| | | | | | | | | | - Dongmei Man
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Yanping Wang
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
7
|
Xu Lou I, Yu X, Chen Q. Exploratory review on the effect of Astragalus mongholicus on signaling pathways. Front Pharmacol 2024; 15:1510307. [PMID: 39726784 PMCID: PMC11670317 DOI: 10.3389/fphar.2024.1510307] [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: 10/12/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Background Astragalus mongholicus Bunge [Fabaceae; Astragali radix] (AM), a traditional Chinese medicinal (TCM) botanical drug, has been used for centuries and is gaining growing recognition in medical research for its therapeutic potential. The currently accepted scientific name is Astragalus mongholicus Bunge, with Astragalus membranaceus Fisch. ex Bunge recognized as a taxonomic synonym. This review explores the most relevant scientific studies on AM, focusing on its chemical composition, mechanisms of action, and associated health benefits. Main body AM is commonly used in clinical practice to treat diabetes mellitus, cardiovascular diseases, oncological processes, lipid metabolism disorders, and ulcerative colitis. Recent research has investigated its potential as a product for anti-aging purposes. These therapeutic effects are attributed to the interactions of bioactive metabolites such as Astragaloside IV, Formononetin, and polysaccharides, with various signaling pathways, leading to the activation or inhibition of gene expression. This review aims to map the signaling pathways affected by these metabolites and their effects on different pathologies. Studies suggest that these metabolites act on signaling pathways such as TLR4/MyD88/NF-κB, PI3K/AKT, RNA expression, and tumor receptors. However, further research is necessary to validate the findings in human trials with better methodological quality. Conclusion AM is rich in bioactive metabolites that interact with various signaling pathways, modulating diseases such as diabetes mellitus type 2, cardiovascular diseases, cancer, lipid metabolism disorders, and ulcerative colitis. Although promising, the majority of the studies are conducted in vitro and animal models, and more rigorous human trials are needed to determine the therapeutic potential of AM.
Collapse
Affiliation(s)
| | | | - Qilan Chen
- Department of Cardiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Xiao Q, Luo L, Zhu X, Yan Y, Li S, Chen L, Wang X, Zhang J, Liu D, Liu R, Zhong Y. Formononetin alleviates ulcerative colitis via reshaping the balance of M1/M2 macrophage polarization in a gut microbiota-dependent manner. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156153. [PMID: 39423480 DOI: 10.1016/j.phymed.2024.156153] [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: 08/04/2024] [Revised: 09/18/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Ulcerative colitis (UC), a type of inflammatory bowel disease, presents substantial challenges in clinical treatment due to the limitations of current medications. Formononetin (FN), a naturally compound with widespread availability, exhibits anti-inflammatory, antioxidant, and immunomodulatory properties. PURPOSE This study aimed to investigate the efficacy of FN against UC and its potential regulatory mechanism. METHODS Here, dextran sulfate sodium (DSS) was employed to replicate experimental colitis in mice with concomitant FN treatment. The distribution and localisation of CD68 and F4/80 macrophages in colonic tissues were visualized by immunofluorescence, their chemokine and inflammatory cytokine concentrations were determined by ELISA, and macrophages and M1/M2 subpopulations were determined by flow cytometry. Additionally, 16 s rRNA and LC-MS techniques were used to detect the colonic intestinal microbiota and metabolite profiles, respectively. Correlation analyses was performed to clarify the interactions between differential bacteria, metabolites and M1/M2 macrophages, and pseudo sterile mice were constructed by depletion of gut flora with quadruple antibiotics, followed by faecal microbial transplantation to evaluate its effects on colitis and M1/M2 macrophage polarisation. RESULTS FN dose-dependently alleviated clinical symptoms and inflammatory injury in colonic tissues of colitis mice, with its high-dose efficacy comparable to that of 5-ASA. Concurrently, FN not only inhibited inflammatory infiltration of macrophages and their M1/M2 polarisation balance in colitis mice, but also improved the composition of colonic microbiota and metabolite profiles. However, FN lost its protective effects against DSS-induced colitis and failed to restore the equilibrium of M1/M2 macrophage differentiation following intestinal flora depletion through quadruple antibiotic treatment. Importantly, fecal microbiota transplantation from FN-treated mice restored FN's protective effects against DSS-induced colitis and reestablished its regulatory role in M1/M2 macrophage polarization. CONCLUSION Collectively, FN ameliorated UC through modulating the balance of M1/M2 macrophage polarization in a gut microbiota-dependent manner.
Collapse
Affiliation(s)
- Qiuping Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Lin Luo
- College of Acupuncture and Tuina, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Xiyan Zhu
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Yuhao Yan
- College of Acupuncture and Tuina, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Shanshan Li
- Laboratory Animal Science and Technology Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Liling Chen
- Laboratory Animal Science and Technology Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Xiaomin Wang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Jie Zhang
- Laboratory Animal Science and Technology Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Duanyong Liu
- Jiangxi Provincial Engineering Research Center of Development and Evaluation of TCM classic prescriptions, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; College of Nursing, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Key Laboratory of Prevention and Treatment of Immunological and Metabolic Diseases Related to Prescription and Syndrome, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Ronghua Liu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Youbao Zhong
- College of Acupuncture and Tuina, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Laboratory Animal Science and Technology Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China; Key Laboratory of Prevention and Treatment of Immunological and Metabolic Diseases Related to Prescription and Syndrome, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China.
| |
Collapse
|
9
|
Su Y, Cui Z, Yang X, Jiang Y, Zhang W, Zhang Y, Man C. Lactobacillus paracasei JY062 and its exopolysaccharide enhance the intestinal barrier through macrophage polarization and Th17/Treg cell balance. Food Res Int 2024; 197:115235. [PMID: 39593317 DOI: 10.1016/j.foodres.2024.115235] [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: 07/11/2024] [Revised: 09/18/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024]
Abstract
Ulcerative colitis (UC) is an immune-mediated intestinal disease without a comprehensive cure, and the alleviation of UC has become an urgent problem. The results showed that JY062 with its EPS group (JEC) alleviated the intestinal barrier damage caused by LPS. After JEC intervention on Caco-2 cells, resulted in upregulation of ZO-1, Claudin-1, Occludin and MUC2 transcript levels and decreased mRNA expression of Claudin-2 (p < 0.05). JEC effectively attenuated the inflammatory response in UC mice and restoration of immunoglobulin levels (IgG, IgM and IgA), which resulted in shortening and swelling of the colon, disappearance of goblet cells, infiltration of inflammatory cells and mucosal damage were alleviated in mice. Similarly, changes in the expression of MUC2 and tight junction proteins after JEC intervention also occurred in UC mice. Administration of JEC significantly inhibited the differentiation of pro-inflammatory Th17 cells in the thymus and peripheral blood, promoted the differentiation of CD4+ T cells to Treg cells, and effectively regulated DSS-induced macrophage imbalance, which was manifested by the polarization of pro-inflammatory M1 macrophages to anti-inflammatory M2 macrophages. This study clearly demonstrates that JEC could significantly prevent intestinal barrier on DSS-induced experimental colitis and could be applied as a potential symbiotic strategy to assist in the alleviation of UC.
Collapse
Affiliation(s)
- Yue Su
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science and Engineering, Northeast Agricultural University, Harbin 150030, China
| | - Zhengying Cui
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science and Engineering, Northeast Agricultural University, Harbin 150030, China
| | - Xinyan Yang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science and Engineering, Northeast Agricultural University, Harbin 150030, China
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science and Engineering, Northeast Agricultural University, Harbin 150030, China
| | - Wei Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science and Engineering, Northeast Agricultural University, Harbin 150030, China
| | - Yu Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science and Engineering, Northeast Agricultural University, Harbin 150030, China.
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science and Engineering, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
10
|
Wang J, Pu X, Zhuang H, Guo Z, Wang M, Yang H, Li C, Chang X. Astragaloside IV alleviates septic myocardial injury through DUSP1-Prohibitin 2 mediated mitochondrial quality control and ER-autophagy. J Adv Res 2024:S2090-1232(24)00471-5. [PMID: 39550027 DOI: 10.1016/j.jare.2024.10.030] [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: 08/27/2024] [Revised: 10/06/2024] [Accepted: 10/16/2024] [Indexed: 11/18/2024] Open
Abstract
INTRODUCTION Septic cardiomyopathy (SCM) is a complication of myocardial injury in patients with severe sepsis. OBJECTIVES This study highlights the potential of Astragaloside IV(AS) in the treatment of septic cardiomyopathy and provides a reference for developing cardioprotective drugs targeting DUSP1-PHB2-related mitochondria-ER interaction. METHODS Dual specificity phosphatase-1 (DUSP1)/Prohibitin 2 cardiomyocyte-specific knockout mice (DUSP1/PHB2CKO) /DUSP1 transgenic mice (DUSP1/PHB2TG) were used to generate LPS-induced sepsis models. The pathological mechanism by which AS-IV improves heart injury was detected using cardiac ultrasound, fluorescence staining, transmission electron microscopy, and western blotting. After siRNA treatment of cardiomyocytes with DUSP-1/PHB2, changes in mitochondrial function and morphology were determined using qPCR, western blotting, ELISA, and laser confocal microscopy, and the targeted therapeutic effects of AS-IV were further examined. RESULTS SCM treatment leads to severe mitochondrial dysfunction. However, Astragaloside IV (AS) treatment normalizes mitochondrial homeostasis and ER function. Notably, the protective effect was blocked in DUSP1/Prohibitin 2 cardiomyocyte-specific knockout mice (DUSP1/PHB2CKO) but remained unaffected in DUSP1 transgenic mice (DUSP1/PHB2TG). CONCLUSION This study highlights the potential of AS in the treatment of septic cardiomyopathy and provides a reference for developing cardioprotective drugs targeting DUSP1-PHB2 related mitochondria-ER interaction.
Collapse
Affiliation(s)
- Junyan Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xiangyi Pu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Haowen Zhuang
- School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Zhijiang Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Mengyuan Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Huaihong Yang
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China.
| | - Chun Li
- School of Pharmaceutical Sciences, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin 519000, China.
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
11
|
Shen J, Zhao Y, Cui W. Astragalus mongholicus Bunge extract improves ulcerative colitis by promoting PLCB2 to inhibit colonic epithelial cell pyroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118554. [PMID: 38992398 DOI: 10.1016/j.jep.2024.118554] [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: 05/06/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus mongholicus Bunge (AM) and its active ingredients are mainly used for anti-inflammatory, antiviral, antioxidant, immune regulation, cardiovascular and nervous system protection, anti-cancer, anti-tumor and so on. AIM OF THE STUDY To explore the Astragalus mongholicus Bunge extract pharmacological mechanisms and biology processes which improves ulcerative colitis (UC). MATERIALS AND METHODS Dextran sulfate sodium (DSS)-induced UC models in C57BL/6 mice were established, and the mice were treated with Astragalus mongholicus Bunge extract or salazosulfapyridine (SASP). DSS-induced mice- and human-derived colonic epithelial cell lines were used to reveal the inflammatory environment of UC. After treatment with Astragalus mongholicus Bunge extract, the expression of phospholipase C-β 2 (PLCB2) in the cells was detected by quantitative real-time PCR (qRT-PCR), and cell proliferative activity was detected by cell counting kit 8 (CCK-8) assay. Finally, the levels of pyroptosis-related inflammatory factors in cell culture supernatants was detected by ELISA. RESULTS Treatment of UC mice with Astragalus mongholicus Bunge extract do significantly improved DAI scores and histopathological damage scores, and decreased the levels of Eotaxin, GCSF, KC, MCP-1, TNF-α, and IL-6. Besides, Astragalus mongholicus Bunge extract inhibited the expression of nucleotide-binding oligomerization segment-like receptor family 3 (NLRP3), cleaved Caspase-1, and GSDMD-N in the colonic tissues, and reduced the levels of inflammation-related factors IL-1β and IL-18 in serum and tissues. In vitro, Astragalus mongholicus Bunge extract partially reversed the DSS-induced reduction of PLCB2 expression in CP-M030 and NCM460, promoted cell proliferative activity, and reduced the levels of IL-1β and IL-18. CONCLUSIONS In DDS-induced UC mice, Astragalus mongholicus Bunge extract improves ulcerative colitis by inhibiting colonic epithelial cell pyroptosis through PLCB2 promotion.
Collapse
Affiliation(s)
- Jie Shen
- Department of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital (Affiliated Lihuili Hospital of Ningbo University), China.
| | - Yibin Zhao
- Department of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital (Affiliated Lihuili Hospital of Ningbo University), China.
| | - Wei Cui
- Department of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital (Affiliated Lihuili Hospital of Ningbo University), China.
| |
Collapse
|
12
|
Lu Y, Yu X, Wang Z, Kong L, Jiang Z, Shang R, Zhong X, Lv S, Zhang G, Gao H, Yang N. Microbiota-gut-brain axis: Natural antidepressants molecular mechanism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:156012. [PMID: 39260135 DOI: 10.1016/j.phymed.2024.156012] [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: 05/01/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe mental health condition characterized by persistent depression, impaired cognition, and reduced activity. Increasing evidence suggests that gut microbiota (GM) imbalance is closely linked to the emergence and advancement of MDD, highlighting the potential significance of regulating the "Microbiota-Gut-Brain" (MGB) axis to impact the development of MDD. Natural products (NPs), characterized by broad biological activities, low toxicity, and multi-target characteristics, offer unique advantages in antidepressant treatment by regulating MGB axis. PURPOSE This review was aimed to explore the intricate relationship between the GM and the brain, as well as host responses, and investigated the mechanisms underlying the MGB axis in MDD development. It also explored the pharmacological mechanisms by which NPs modulate MGB axis to exert antidepressant effects and addressed current research limitations. Additionally, it proposed new strategies for future preclinical and clinical applications in the MDD domain. METHODS To study the effects and mechanism by which NPs exert antidepressant effects through mediating the MGB axis, data were collected from Web of Science, PubMed, ScienceDirect from initial establishment to March 2024. NPs were classified and summarized by their mechanisms of action. RESULTS NPs, such as flavonoids,alkaloids,polysaccharides,saponins, terpenoids, can treat MDD by regulating the MGB axis. Its mechanism includes balancing GM, regulating metabolites and neurotransmitters such as SCAFs, 5-HT, BDNF, inhibiting neuroinflammation, improving neural plasticity, and increasing neurogenesis. CONCLUSIONS NPs display good antidepressant effects, and have potential value for clinical application in the prevention and treatment of MDD by regulating the MGB axis. However, in-depth study of the mechanisms by which antidepressant medications affect MGB axis will also require considerable effort in clinical and preclinical research, which is essential for the development of effective antidepressant treatments.
Collapse
Affiliation(s)
- Yitong Lu
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiaowen Yu
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Zhongling Wang
- Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Linghui Kong
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Zhenyuan Jiang
- Department of Neurology, Affiliated Hospital of shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Ruirui Shang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xia Zhong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing 100191, China
| | - Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Haonan Gao
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Ni Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
13
|
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
|
14
|
Zhao M, Cui Y, Wang F, Wu F, Li C, Liu S, Chen B. Ursolic Acid Regulates Immune Balance, Modulates Gut Microbial Metabolism, and Improves Liver Health in Mice. Int J Mol Sci 2024; 25:10623. [PMID: 39408951 PMCID: PMC11477038 DOI: 10.3390/ijms251910623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Ursolic acid (UA) has demonstrated significant immunomodulatory and hepatoprotective effects; however, the underlying mechanisms remain unclear. This study aims to analyze the impact of UA on the gut microbiome, metabolome, and liver transcriptome, investigate UA's role in maintaining gut immune homeostasis and liver health, and evaluate the potential contributions of gut microbes and their metabolites to these beneficial effects. Our findings indicate that UA enhances immune balance in the jejunum, fortifies intestinal barrier function, and promotes overall gut health. UA modulates the intestinal microbiota and its metabolic processes, notably increasing the abundance of beneficial bacteria such as Odoribacter and Parabacteroides, along with their metabolites, including ornithine and lactucin. Additionally, UA inhibits the expression of interleukin-1 receptor 1 (IL1R1) and calcium (Ca2+) voltage-gated channel auxiliary subunit beta 2 (CACNB2) while enhancing the synthesis pathways of retinol and ascorbic acid, thereby exerting a protective influence on liver function. In summary, UA enhances intestinal immune homeostasis and promotes liver health, with these advantageous effects potentially mediated by beneficial bacteria (Odoribacter and Parabacteroides) and their metabolites (ornithine and lactucin).
Collapse
Affiliation(s)
- Man Zhao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071051, China
| | - Yali Cui
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071051, China
| | - Fengxia Wang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071051, China
| | - Fengyang Wu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071051, China
| | - Chong Li
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071051, China
| | - Shudong Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071051, China
| | - Baojiang Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071051, China
| |
Collapse
|
15
|
Liu R, Luo Y, Ma J, Zhang Q, Sheng Y, Li J, Li H, Zhao T. Traditional Chinese medicine for functional gastrointestinal disorders and inflammatory bowel disease: narrative review of the evidence and potential mechanisms involving the brain-gut axis. Front Pharmacol 2024; 15:1444922. [PMID: 39355776 PMCID: PMC11443704 DOI: 10.3389/fphar.2024.1444922] [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: 06/06/2024] [Accepted: 08/23/2024] [Indexed: 10/03/2024] Open
Abstract
Functional gastrointestinal disorders (FGIDs) and inflammatory bowel disease (IBD) are common clinical disorders characterized by recurrent diarrhea and abdominal pain. Although their pathogenesis has not been fully clarified, disruptions in intestinal motility and immune function are widely accepted as contributing factors to both conditions, and the brain-gut axis plays a key role in these processes. Traditional Chinese Medicine (TCM) employs a holistic approach to treatment, considers spleen and stomach impairments and liver abnormality the main pathogenesis of these two diseases, and offers a unique therapeutic strategy that targets these interconnected pathways. Clinical evidence shows the great potential of TCM in treating FGIDs and IBD. This study presents a systematic description of the pathological mechanisms of FGIDs and IBD in the context of the brain-gut axis, discusses clinical and preclinical studies on TCM and acupuncture for the treatment of these diseases, and summarizes TCM targets and pathways for the treatment of FGIDs and IBD, integrating ancient wisdom with contemporary biomedical insights. The alleviating effects of TCM on FGID and IBD symptoms are mainly mediated through the modulation of intestinal immunity and inflammation, sensory transmission, neuroendocrine-immune network, and microbiota and their metabolism through brain-gut axis mechanisms. TCM may be a promising treatment option in controlling FGIDs and IBD; however, further high-quality research is required. This review provides a reference for an in-depth exploration of the interventional effects and mechanisms of TCM in FGIDs and IBD, underscoring TCM's potential to recalibrate the dysregulated brain-gut axis in FGIDs and IBD.
Collapse
Affiliation(s)
- RuiXuan Liu
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - YunTian Luo
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - JinYing Ma
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qi Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yudong Sheng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiashan Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongjiao Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - TianYi Zhao
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
Zhang Y, Kang Q, He L, Chan KI, Gu H, Xue W, Zhong Z, Tan W. Exploring the immunometabolic potential of Danggui Buxue Decoction for the treatment of IBD-related colorectal cancer. Chin Med 2024; 19:117. [PMID: 39210410 PMCID: PMC11360867 DOI: 10.1186/s13020-024-00978-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Danggui Buxue (DGBX) decoction is a classical prescription composed of Astragali Radix (AR) and Angelicae Sinensis Radix (ASR), used to enrich blood, and nourish Qi in Chinese medicine, with the potential to recover energy and stimulate metabolism. Chronic inflammation is a risk factor in the development of inflammatory bowel disease (IBD)-related colorectal cancer (CRC). More importantly, AR and ASR have anti-inflammatory and anti-cancer activities, as well as prefiguring a potential effect on inflammation-cancer transformation. We, therefore, aimed to review the immunometabolism potential of DGBX decoction and its components in this malignant transformation, to provide a helpful complement to manage the risk of IBD-CRC. The present study investigates the multifaceted roles of DGBX decoction and its entire components AR and ASR, including anti-inflammation effects, anti-cancer properties, immune regulation, and metabolic regulation. This assessment is informed by a synthesis of scholarly literature, with more than two hundred articles retrieved from PubMed, Web of Science, and Scopus databases within the past two decades. The search strategy employed utilized keywords such as "Danggui Buxue", "Astragali Radix", "Angelicae Sinensis Radix", "Inflammation", and "Metabolism", alongside the related synonyms, with a particular emphasis on high-quality research and studies yielding significant findings. The potential of DGBX decoction in modulating immunometabolism holds promise for the treatment of IBD-related CRC. It is particularly relevant given the heterogeneity of CRC and the growing trend towards personalized medicine, but the precise and detailed mechanism necessitate further in vivo validation and extensive clinical studies to substantiate the immunometabolic modulation and delineate the pathways involved.
Collapse
Affiliation(s)
- Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Luying He
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, SAR, China
| | - Hui Gu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Wenjing Xue
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, SAR, China.
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
17
|
Huang Y, Wu Q, Li S, Lin X, Yang S, Zhu R, Fu C, Zhang Z. Harnessing nature's pharmacy: investigating natural compounds as novel therapeutics for ulcerative colitis. Front Pharmacol 2024; 15:1394124. [PMID: 39206263 PMCID: PMC11349575 DOI: 10.3389/fphar.2024.1394124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024] Open
Abstract
Backgrounds Ulcerative colitis (UC) is a form of chronic inflammatory bowel disease, and UC diagnosis rates continue to rise throughout the globe. The research and development of new drugs for the treatment of UC are urgent, and natural compounds are an important source. However, there is a lack of systematic summarization of natural compounds and their mechanisms for the treatment of UC. Methods We reviewed the literature in the databases below from their inception until July 2023: Web of Science, PubMed, China National Knowledge Infrastructure, and Wanfang Data, to obtain information on the relationship between natural compounds and UC. Results The results showed that 279 natural compounds treat UC through four main mechanisms, including regulating gut microbiota and metabolites (Mechanism I), protecting the intestinal mucosal barrier (Mechanism II), regulating intestinal mucosal immune response (Mechanism III), as well as regulating other mechanisms (Mechanism Ⅳ) such as cellular autophagy modulation and ferroptosis inhibition. Of these, Mechanism III is regulated by all natural compounds. The 279 natural compounds, including 62 terpenoids, 57 alkaloids, 52 flavonoids, 26 phenols, 19 phenylpropanoids, 9 steroids, 9 saponins, 8 quinonoids, 6 vitamins, and 31 others, can effectively ameliorate UC. Of these, terpenoids, alkaloids, and flavonoids have the greatest potential for treating UC. It is noteworthy to highlight that a total of 54 natural compounds exhibit their therapeutic effects by modulating Mechanisms I, II, and III. Conclusion This review serves as a comprehensive resource for the pharmaceutical industry, researchers, and clinicians seeking novel therapeutic approaches to combat UC. Harnessing the therapeutic potential of these natural compounds may significantly contribute to the improvement of the quality of life of patients with UC and promotion of disease-modifying therapies in the future.
Collapse
Affiliation(s)
- You Huang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuhong Wu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sha Li
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xia Lin
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shasha Yang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Zhu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomei Fu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Zhang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Sun Q, Hu M, Yuan C, Ren B, Zhong M, Zhou S, Wang X, Gao Q, Zeng M, Cai X, Song H. Astragaloside IV ameliorates indomethacin-induced intestinal inflammation in rats through inhibiting the activation of NLRP3 inflammasome. Int Immunopharmacol 2024; 135:112281. [PMID: 38762925 DOI: 10.1016/j.intimp.2024.112281] [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: 02/21/2024] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
The administration of nonsteroidal anti-inflammatory drugs (NSAIDs) may cause significant intestinal alteration and inflammation and lead to the occurrence of inflammatory diseases resembling duodenal ulcers. Astragaloside IV (AS-IV) is a glycoside of cycloartane-type triterpene isolated from the dried root of Astragalus membranaceus (Fisch.) Bge. (family Fabaceae), and has been used for ameliorating the NSAID-induced inflammation in the small intestine. The present study aimed to investigate the effects of AS-IV on indomethacin (IND)-induced inflammation in the small intestine of rats and its underlying mechanisms. Hematoxylin-eosin (H&E) staining, transmission and scanning electron microscopy were carried out to observe the surface morphology and ultrastructure of the small intestinal mucosa. Immunofluorescence and ELISA tests were employed to detect the expressions of NLRP3, ASC, caspase-1, and NF-κB proteins, as well as inflammatory factors IL-1β and IL-18, to uncover potential molecular mechanisms responsible for mitigating small intestinal inflammation. The results demonstrated that AS-IV significantly decreased the ulcer index, improved the surface morphology and microstructure of the small intestinal mucosa, and increased mucosal blood flow. Molecular docking revealed a strong and stable binding capacity of AS-IV to NLRP3, ASC, caspase-1, and NF-κB proteins. Further experimental validation exhibited that AS-IV markedly decreased levels of IL-1β and IL-18, and inhibited the protein expression of NLRP3, ASC, caspase-1, and NF-κB. Our data demonstrate that AS-IV ameliorates IND-induced intestinal inflammation in rats by inhibiting the activation of NLRP3 inflammasome and reducing the release of IL-1β and IL-18, thereby representing a promising therapy for IND-induced intestinal inflammation.
Collapse
Affiliation(s)
- Qifang Sun
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Mingyue Hu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Chengzhi Yuan
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Baoping Ren
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Meiqi Zhong
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Shunhua Zhou
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiaojuan Wang
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Qing Gao
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Meiyan Zeng
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiong Cai
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| | - Houpan Song
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics and School of Chinese Medical Sciences, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China.
| |
Collapse
|
19
|
Wu Q, Yan H, Kang Z. A Review of Traditional Chinese Medicine for Triple Negative Breast Cancer and the Pharmacological Mechanisms. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:987-1011. [PMID: 38879747 DOI: 10.1142/s0192415x2450040x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer. Conventional treatment options for TNBC often have limited efficacy and significant side effects. In recent years, traditional Chinese medicine (TCM) has shown promising results in the treatment of TNBC. TCMs include herb combinations that have synergistic effects to regulate homeostasis in the body, reduce tumor resistance, and improve patient quality of life. At present, three main TCM methods are used to treat TNBC in the clinic: strengthening the body's resistance, dispelling phlegm, and removing cancer toxins. This paper reviews the theories and mechanisms of each in TNBC treatment. The method of strengthening the body's resistance emphasizes enhancing the body's original Qi to fight against pathogenic factors; the method of dispelling phlegm seeks to eliminate phlegm stagnation and alleviate the burden on affected organs; the method of removing cancer toxins focuses on detoxification and detumescence to remove the toxic elements associated with TNBC. Although these methods treat TNBC from different etiologies, they have achieved good therapeutic effects and represent an important academic approach: That is, to cure the disease with a comprehensive view of the body and restore the balance of Yin and Yang. This knowledge lays a foundation for the future development and reasonable application of TCM in the clinic.
Collapse
Affiliation(s)
- Qinhang Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Hongkai Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Ziyi Kang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
20
|
Huang Y, Chen X, Liu X, Lin C, Wang Y. The coumarin component isofraxidin targets the G-protein-coupled receptor S1PR1 to modulate IL-17 signaling and alleviate ulcerative colitis. Int Immunopharmacol 2024; 131:111814. [PMID: 38479159 DOI: 10.1016/j.intimp.2024.111814] [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: 10/07/2023] [Revised: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 04/10/2024]
Abstract
OBJECTIVE The increasing global prevalence of ulcerative colitis (UC) underscores the imperative to explore novel therapeutic approaches. Traditional Chinese medicine has historically shown potential in addressing this ailment. The current study aimed to elucidate the functional attributes and underlying mechanisms of isofraxidin, a coumarin derivative from Acanthopanax, in the context of UC. METHODS A murine model of dextran sodium sulfate (DSS)-induced UC was established, and we conducted a comprehensive assessment of the influence of isofraxidin on UC symptomatology, colonic histopathological manifestations, the inflammatory response, and apoptosis. The potential receptor of isofraxidin was initially identified through the Target database and molecular docking analysis. Subsequent in vivo and in vitro experiments were conducted to determine the effects of isofraxidin on the identified receptor and associated signaling pathways. Transfection was used to examine the receptor's role in the regulatory mechanism of isofraxidin. RESULTS Isofraxidin reduced UC symptoms and colonic histopathological impairments. Furthermore, isofraxidin ameliorated the DSS-induced inflammatory response and apoptosis in tissues. S1PR1 was identified as a target of isofraxidin and effectively suppressed activation of the IL-17 signaling pathway. Intriguingly, cellular experiments indicated that overexpression of S1PR1 counteracted the protective effect of isofraxidin. DISCUSSION In summary, our investigation revealed that isofraxidin could modulate S1PR1 and regulate the IL-17 signaling pathway, thus ameliorating DSS-induced UC. These findings establish a robust foundation for considering isofraxidin as a prospective therapeutic intervention to treat UC.
Collapse
Affiliation(s)
- Yisen Huang
- Department of Gastroenterology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Xiangbo Chen
- Digestive Endoscopy Center, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Xiaoqiang Liu
- Department of Gastroenterology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Chanchan Lin
- Department of Gastroenterology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Yubin Wang
- Department of Gastroenterology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China.
| |
Collapse
|
21
|
Wushouer X, Aximujiang K, Kadeer N, Aihemaiti A, Zhong L, Yunusi K. Effect of huankuile on colon injury in rats with ulcerative colitis by reducing TNF-α and MMP9. Eur J Med Res 2024; 29:102. [PMID: 38321559 PMCID: PMC10845565 DOI: 10.1186/s40001-024-01695-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/24/2024] [Indexed: 02/08/2024] Open
Abstract
OBJECTIVE To explore the mechanism of huankuile (HKL) in colon injury repair in rats with ulcerative colitis (UC). METHODS Fifty SPF Wistar male rats were divided randomly into a normal group, a negative control group, an HKL intervention group ('HKL group') and a 5-aminosalicylic acid intervention group ('5-ASA group'). After 14 days of intervention with corresponding drugs, pathological scores were obtained using the results of immunohistochemical staining; morphological changes were observed by hematoxylin-eosin staining, and the mRNA expression levels of tumour necrosis factor-α (TNF-α), matrix metalloproteinase 9 (MMP9) and interleukin-13 (IL-13) were detected by real-time quantitative PCR. RESULTS After the successful construction of the rat model, it was compared with the rats in the normal group. In the negative group, it was found that the expression of TNF-α and MMP9 was significantly increased in the colonic mucosal epithelia of the rats, the pathological score was significantly increased (P < 0.05), and the mRNA expression levels of TNF-α, MMP9 and IL-13 were increased (P < 0.05). After treatment with HKL, the colonic morphology of the rats returned to normal, the expression of TNF-α and MMP9 in the colonic mucosal epithelium of the rats returned to normal, the pathological score grade was significantly reduced (P < 0.05), and the mRNA expression levels of TNF-α, MMP9 and IL-13 were reduced; these results were largely consistent with those of the normal group, with no statistically significant difference. CONCLUSION HKL effectively improved the general symptoms and tissue injury in UC rats, and the therapeutic effect was better than that of 5-ASA group. Ulcerative colitis in rats increased the expression of TNF-α, MMP9 and IL-13. HKL repaired UC-induced colonic injury in rats by decreasing the expression of TNF-α, MMP9 and IL-13.
Collapse
Affiliation(s)
- Xilinguli Wushouer
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
- Xinjiang key laboratory of Molecular Biology for endemic diseases, Urumqi , 830054, China
| | - Kasimujiang Aximujiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Nafeisha Kadeer
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Abulaiti Aihemaiti
- The Functional Center, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Li Zhong
- The Functional Center, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Kurexi Yunusi
- UygurMedical College, Xinjiang Medical University, Urumqi, 830017, China.
| |
Collapse
|
22
|
Liu X, Wang S, Wu X, Zhao Z, Jian C, Li M, Qin X. Astragaloside IV Alleviates Depression in Rats by Modulating Intestinal Microbiota, T-Immune Balance, and Metabolome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:259-273. [PMID: 38064688 DOI: 10.1021/acs.jafc.3c04063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
This study aims to explore the effects of Astragaloside IV (AS-IV) on abnormal behaviors, intestinal microbiota, intestinal T-immune balance, and fecal metabolism of a model of depression in rats. Herein, we integrally applied 16S rRNA sequencing, molecular biological techniques, and 1H NMR-based fecal metabolomics to demonstrate the antidepression activity of AS-IV. The results suggested that AS-IV regulated the depression-like behaviors of rats, which are presented by an increase of body weight, upregulation of sucrose preference rates, and a decrease of immobility time. Additionally, AS-IV increased the abundances of beneficial bacteria (Lactobacillus and Oscillospira) in a model of depression in rats. Moreover, AS-IV regulated significantly the imbalance of Th17/Treg cells, and the abnormal contents of both anti-inflammatory factors and pro-inflammatory factors. Besides, fecal metabolomics showed that AS-IV improved the abnormal levels of short-chain fatty acids and amino acids. Collectively, our research supplemented new data, supporting the potential of AS-IV as an effective diet or diet composition to improve depression-like behaviors, dysfunctions of microbiota, imbalance of T immune, and the abnormality of fecal metabolome. However, the causality of the other actions was not proven because of the experimental design and the methodology used. The current findings suggest that AS-IV could function as a promising diet or diet composition to alleviate depressed symptoms.
Collapse
Affiliation(s)
- Xiaojie Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
| | - Senyan Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
| | - Xiaoling Wu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
| | - Ziyu Zhao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
| | - Chen Jian
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
| | - Mengyu Li
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
| |
Collapse
|
23
|
Wang M, Sun F, Han X, Wang N, Liu Y, Cai J, Tong S, Wang R, Wang H. Astragaloside IV Inhibits Rotenone-Induced α-syn Presentation and the CD4 T-Cell Immune Response. Mol Neurobiol 2024; 61:252-265. [PMID: 37603153 DOI: 10.1007/s12035-023-03566-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/07/2023] [Indexed: 08/22/2023]
Abstract
The increased α-synuclein (α-syn)-dependent activation of CD4 T cells leads to the progressive loss of dopaminergic (DA) neurons in the substantia nigra (SN) in Parkinson's disease (PD). Astragaloside IV (AS-IV) protects DA neurons against neuroinflammation. The effects of AS-IV on CD4 T-cell-mediated immune responses in PD remain unknown. Rotenone (ROT) injected unilaterally into the substantia nigra pars compacta (SNc) of rats induced PD. AS-IV (20 mg/kg) was intraperitoneally injected once a day for 14 days. The limb hanging test and rotarod test were performed to evaluate the alteration of behavior at 4 and 6 weeks. Total gastrointestinal transit tests were performed at 4 weeks. Western blotting was used to detect the expression of proinflammatory cytokine proteins. Immunofluorescence staining was conducted to test the expression and localization of major histocompatibility complex class II (MHCII), cleaved caspase-1 and α-syn in astrocytes. Flow cytometry analysis, immunohistochemistry and immunofluorescence staining were used to measure the expression of CD4 T-cell subsets in the SN. The application of AS-IV protected against the loss of DA neurons and behavioral deficits in ROT-induced PD rat models. AS-IV administration inhibited the aggregation of α-syn in DA neurons and the expression of proinflammatory cytokines such as TNF-α, IL-18, IL-6 and IL-1β. AS-IV decreased the activation of CD4 T cells and three CD4 T-cell subsets: Tfh, Treg and Th1. AS-IV interrupted the ROT-induced interaction between astrocytes and CD4 T cells and the colocalization of MHCII and α-syn in astrocytes. AS-IV inhibited the expression of α-syn in astrocytes and the colocalization of α-syn and cleaved caspase-1 in astrocytes. AS-IV prevents the loss of DA neurons in PD by inhibiting the activation of α-syn-specific CD4 T cells, which is regulated by MHCII-mediated antigen presentation in astrocytes.
Collapse
Affiliation(s)
- Mengdi Wang
- Department of Neurology, Binzhou Medical University Hospital, No. 661, the 2nd Yellow River Road, Shandong Province, 256603, Binzhou City, China
| | - Fengjiao Sun
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Xiaofeng Han
- Department of Neurology, Binzhou Medical University Hospital, No. 661, the 2nd Yellow River Road, Shandong Province, 256603, Binzhou City, China
| | - Nan Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yalan Liu
- Department of Neurology, Binzhou Medical University Hospital, No. 661, the 2nd Yellow River Road, Shandong Province, 256603, Binzhou City, China
| | - Jinfeng Cai
- Department of Neurology, Binzhou Medical University Hospital, No. 661, the 2nd Yellow River Road, Shandong Province, 256603, Binzhou City, China
| | - Shanshan Tong
- Department of Neurology, Binzhou Medical University Hospital, No. 661, the 2nd Yellow River Road, Shandong Province, 256603, Binzhou City, China
| | - Rui Wang
- Department of Neurology, Binzhou Medical University Hospital, No. 661, the 2nd Yellow River Road, Shandong Province, 256603, Binzhou City, China
| | - Hongcai Wang
- Department of Neurology, Binzhou Medical University Hospital, No. 661, the 2nd Yellow River Road, Shandong Province, 256603, Binzhou City, China.
| |
Collapse
|
24
|
Shen L, Luo H, Fan L, Tian X, Tang A, Wu X, Dong K, Su Z. Potential Immunoregulatory Mechanism of Plant Saponins: A Review. Molecules 2023; 29:113. [PMID: 38202696 PMCID: PMC10780299 DOI: 10.3390/molecules29010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Saponins are extracted from different parts of plants such as seeds, roots, stems, and leaves and have a variety of biological activities including immunomodulatory, anti-inflammatory effects, and hypoglycemic properties. They demonstrate inherent low immunogenicity and possess the capacity to effectively regulate both the innate and adaptive immune responses. Plant saponins can promote the growth and development of the body's immune organs through a variety of signaling pathways, regulate the activity of a variety of immune cells, and increase the secretion of immune-related cytokines and antigen-specific antibodies, thereby exerting the role of immune activity. However, the chemical structure of plant saponins determines its certain hemolytic and cytotoxicity. With the development of science and technology, these disadvantages can be avoided or reduced by certain technical means. In recent years, there has been a significant surge in interest surrounding the investigation of plant saponins as immunomodulators. Consequently, the objective of this review is to thoroughly examine the immunomodulatory properties of plant saponins and elucidate their potential mechanisms, with the intention of offering a valuable point of reference for subsequent research and advancement within this domain.
Collapse
Affiliation(s)
- Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Hao Luo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Fan
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinyu Tian
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Anguo Tang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaofeng Wu
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ke Dong
- Sichuan Yuqiang Herbal Biotechnology Co., Ltd., Chengdu 611130, China
| | - Zhetong Su
- Guangxi Innovates Medical Technology Co., Ltd., Lipu 546600, China
| |
Collapse
|
25
|
Zhong Y, Xiao Q, Huang J, Yu S, Chen L, Wan Q, Zhang Z, Luo L, Song L, Zhao H, Zhou W, Liu D. Ginsenoside Rg1 Alleviates Ulcerative Colitis in Obese Mice by Regulating the Gut Microbiota-Lipid Metabolism-Th1/Th2/Th17 Cells Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:20073-20091. [PMID: 38064669 DOI: 10.1021/acs.jafc.3c04811] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Ginsenoside Rg1 (G-Rg1) has various pharmacological properties including antiobesity, immunomodulatory, and anti-inflammatory effects. This study aimed to explore the therapeutic effects and underlying mechanisms of G-Rg1 on colitis complicated by obesity. The results indicate that G-Rg1 effectively alleviates colitis in obese mice and improves serum lipid levels and liver function. Importantly, G-Rg1 improved the composition of gut microbiota in obese mice with colitis, with increases in alpha diversity indexes Sobs, Ace, and Chao, a significant down-regulation of the relative abundance of Romboutsia, and a significant up-regulation of Rikenellaceae_RC9_gut_group, Lachnospiraceae_NK4A136_group, Enterorhabdus, Desulfovibrio, and Alistipes. Meanwhile, G-Rg1 improved lipid metabolism in the colonic contents of obese mice with colitis. Additionally, G-Rg1 significantly reduced the percentages of helper T (Th)1, Th17, central memory T (TCM), and effector memory T (TEM) cells in obese mice with colitis while significantly increasing Naïve T and Th2 cells. In conclusion, G-Rg1 could be a promising therapeutic option for alleviating obesity complicated by colitis through regulation of the gut microbiota and lipid metabolism as well as Th1/Th2/Th17 cell differentiation.
Collapse
Affiliation(s)
- Youbao Zhong
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
- Institute of Chinese Medicine and Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330004, Jiangxi, China
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qiuping Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Jiaqi Huang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Songren Yu
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Liling Chen
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qi Wan
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Zheyan Zhang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Lin Luo
- College of Acupuncture and Massage, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Lizhao Song
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Haimei Zhao
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
| | - Wen Zhou
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
- Nanchang Medical College, Nanchang, Jiangxi 330004, China
| | - Duanyong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
- Institute of Chinese Medicine and Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330004, Jiangxi, China
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi, China
| |
Collapse
|
26
|
Zhou Z, Yang L, Hu C, Gao R, Zhang X, Shen T. The combination of astragalus injection and ambroxol hydrochloride in the adjuvant treatment of COPD: a systematic review and meta-analysis. Sci Rep 2023; 13:22077. [PMID: 38087032 PMCID: PMC10716149 DOI: 10.1038/s41598-023-49421-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a severe condition that leads to premature mortality and places a significant financial burden on healthcare systems. An adjunctive therapy in COPD includes the simultaneous administration of astragalus injection and ambroxol hydrochloride. Despite its widespread use, the effectiveness of this combined approach in COPD treatment has not been systematically evaluated. Thus, we conducted a systematic review and meta-analysis to assess the efficacy of combining astragalus injection with ambroxol hydrochloride as an adjuvant treatment for COPD. Six electronic databases were used to search for relevant randomized controlled trials, and data analysis was conducted using Review Manager 5.4. A total of 14 randomized controlled trials were included, involving 1070 patients who met the criteria. The results of the meta-analysis indicated that the combination of astragalus injection with ambroxol hydrochloride as an adjuvant treatment can improve various clinical parameters in patients with COPD compared to conventional treatment alone. These parameters include the clinical effective rate (OR = 5.44, 95% CI 3.51-8.43, I2 = 0%), partial pressure of oxygen in artery (MD = 1.12, 95% CI 0.87-1.36, I2 = 5%), partial pressure of carbon dioxide in artery (MD = - 1.43, 95% CI - 1.65 to - 1.21, I2 = 0%), forced expiratory volume in one second (MD = 0.30, 95% CI 0.18-0.42, I2 = 0%), percentage of forced expiratory volume in one second (MD = 16.18, 95% CI 12.60-19.76, I2 = 82%), forced vital capacity (MD = 0.33, 95% CI 0.21-0.45, I2 = 36%), hemoglobin (MD = - 16.17, 95% CI - 20.84 to - 11.51, I2 = 29%), and the ratio of forced expiratory volume in one second to forced vital capacity (MD = 2.51, 95% CI - 0.05 to 5.06, I2 = 0%). The combination of astragalus injection and ambroxol hydrochloride could be a selection of COPD patients as an adjuvant treatment. However, further validation is required to evaluate the effectiveness of combining astragalus injection and ambroxol hydrochloride as an adjunctive treatment for patients with COPD.
Collapse
Affiliation(s)
- Zubing Zhou
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Lele Yang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Chao Hu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Rui Gao
- Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, 610095, China
| | - Xiaobo Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Tao Shen
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
27
|
Wang H, Wei L, Mao D, Che X, Ye X, Liu Y, Chen Y. Combination of oxymatrine (Om) and astragaloside IV (As) enhances the infiltration and function of TILs in triple-negative breast cancer (TNBC). Int Immunopharmacol 2023; 125:111026. [PMID: 37866315 DOI: 10.1016/j.intimp.2023.111026] [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: 09/24/2023] [Accepted: 10/04/2023] [Indexed: 10/24/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype and has a poor response to treatment due to an immunosuppressive microenvironment. Chinese Medicine effective constituents such as oxymatrine (Om) and astragaloside IV (As) have shown promise in cancer treatment by providing anti-fibrosis and immune-enhancing effects. However, the potential combined effect of Om and As on TNBC and its mechanism is still uncertain. This study focuses on exploring the impact of Om and As on enhancing the immunosuppressive microenvironment of TNBC and uncovering the potential mechanism behind it. In this study, a trans-Cancer associated fibroblasts (CAFs) infiltration system of T cells was utilized to investigate the potential benefits of Om, while the impact of As on the morphology and quantity of mitochondria in T cells was examined in a co-culture system with tumor cells. Further to investigate the combined effects of Om and As on tumor suppression and immunosuppressive microenvironment improvement, this study established an in situ TNBC mouse model with 4 T1-luc. In vitro, our findings indicate that Om can effectively suppress the activation of CAFs by downregulating the expression of FAP and α-SMA, and also promoting the infiltration of T cells trans CAFs. It was discovered that the mitochondrial activity of T cells could be improved by increasing the number of mitochondria and cristae. In vivo, the optimal ratio of Om and As (2:1) was found to increase the apoptosis rate of tumor cells in a co-culture system and enhance the infiltration of CD4+ and CD8+ T cells, as confirmed by Flow Cytometry results. Our study suggests that Om and As could enhance the immune system's ability to treat TNBC by improving the infiltration and increasing the anti-tumor function of TILs. This intervention may lead to a promising therapeutic direction for the treatment of TNBC.
Collapse
Affiliation(s)
- Hong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Liangyin Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Dengxuan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xiaoyu Che
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xietao Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
28
|
Tang YZ, Liu Y, Chen YP, Feng TT, Liu YY, Wang Y, Zhang JP, Xu WH. Citropten alleviates acute and recurrent colitis via blockage of NF-κB and JAK/STAT3 pathways. Int Immunopharmacol 2023; 125:111102. [PMID: 37922567 DOI: 10.1016/j.intimp.2023.111102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/18/2023] [Indexed: 11/07/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease, which is characterized by inflammation, with many symptoms including diarrhea, abdominal pain, bloody stool, and weight loss. It is difficult to completely cure and promising therapeutic drug candidates are urgently needed. Citropten, a coumarin-like compound found in traditional Chinese medicine such as Finger Citron Fruit, notopterygium root and citrus peel, has been shown to inhibit the proliferation of tumor cells, protect against depression and suppress the production of inflammatory mediators. In this study, we demonstrated that citropten could alleviate dextran sulfate sodium (DSS)-induced acute and recurrent colitis in mice, with significant improvement in body weight loss, disease activity index, shortened colon length and histological changes. Moreover, citropten dramatically decreased the production of pro-inflammatory mediators in colon tissues and effectively suppressed the proportion of Th17 cells in spleen. Mechanism investigations revealed that citropten significantly inhibited the activation of NF-κB and JAK/STAT3 signaling pathways, thus leading to decreased inflammation, Th17 cells and alleviative colitis. These findings provide novel insights into the anti-colitis effect of citropten, which may be a promising drug candidate for treatment of IBD.
Collapse
Affiliation(s)
- Yu Zhen Tang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Ying Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, PR China
| | - Ya Ping Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Ting Ting Feng
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Ya Yi Liu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Yan Wang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Jun Ping Zhang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China.
| | - Wei Heng Xu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China.
| |
Collapse
|
29
|
Yu S, Guo H, Ji Z, Zheng Y, Wang B, Chen Q, Tang H, Yuan B. Sea Cucumber Peptides Ameliorate DSS-Induced Ulcerative Colitis: The Role of the Gut Microbiota, the Intestinal Barrier, and Macrophage Polarization. Nutrients 2023; 15:4813. [PMID: 38004208 PMCID: PMC10674221 DOI: 10.3390/nu15224813] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
The incidence of ulcerative colitis (UC) is increasing annually. There are few treatments for UC patients, and some drugs have serious side effects. Sea cucumber peptide (SCP) has anti-inflammatory, antioxidant and other biological activities, and various sea cucumber species are in pharmaceutical development. However, relevant studies on the effects of SCP on UC progression are still lacking. In this study, a mouse model of acute colitis was induced by 3% dextran sulfate (DSS), and the effect of 500 mg/kg SCP on colitis was investigated. The results showed that SCP can alleviate DSS-induced colon damage and intestinal barrier damage. SCP significantly inhibited the expression of inflammatory factors and oxidative stress in UC mice. SCP reversed the intestinal microbiota dysregulation induced by DSS, inhibited the growth of Sutterella, Prevotella_9 and Escherichia-Shigella harmful bacteria, and increased the abundance of Lachnospiraceae_NK4A136_group. At the same time, SCP treatment significantly inhibited the LPS-induced polarization of M1 macrophages, which may be mediated by two monopeptides, IPGAPGVP and TGPIGPPGSP, via FPR2. In conclusion, SCP can protect against colitis by modulating the intestinal microbiota composition and the intestinal barrier and inhibiting the polarization of M1 macrophages.
Collapse
Affiliation(s)
- Song Yu
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Haixiang Guo
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Zhonghao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
- Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, China
| | - Yi Zheng
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Bingbing Wang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Qingqing Chen
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Hongyu Tang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (S.Y.); (H.G.); (Z.J.); (Y.Z.); (B.W.); (Q.C.)
| |
Collapse
|
30
|
Zheng LX, Guo KE, Huang JQ, Liu MH, Deng BL, Liu DY, Zhou BG, Zhou W, Zhong YB, Zhao HM. Curcumin alleviated dextran sulfate sodium-induced colitis by recovering memory Th/Tfh subset balance. World J Gastroenterol 2023; 29:5226-5239. [PMID: 37901446 PMCID: PMC10600958 DOI: 10.3748/wjg.v29.i36.5226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/26/2023] [Accepted: 09/07/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Restoration of immune homeostasis by targeting the balance between memory T helper (mTh) cells and memory follicular T helper (mTfh) cells is a potential therapeutic strategy against ulcerative colitis (UC). Because of its anti-inflammatory and immunomodulatory properties, curcumin (Cur) is a promising drug for UC treatment. However, fewer studies have demonstrated whether Cur can modulate the mTh/mTfh subset balance in mice with colitis. AIM To explore the potential mechanism underlying Cur-mediated alleviation of colitis induced by dextran sulfate sodium (DSS) in mice by regulating the mTh and mTfh immune homeostasis. METHODS Balb/c mice were administered 3% and 2% DSS to establish the UC model and treated with Cur (200 mg/kg/d) by gavage on days 11-17. On the 18th d, all mice were anesthetized and euthanized, and the colonic length, colonic weight, and colonic weight index were evaluated. Histomorphological changes in the mouse colon were observed through hematoxylin-eosin staining. Levels of Th/mTh and Tfh/mTfh cell subsets in the spleen were detected through flow cytometry. Western blotting was performed to detect SOCS-1, SOCS-3, STAT3, p-STAT3, JAK1, p-JAK1, and NF-κB p65 protein expression levels in colon tissues. RESULTS Cur effectively mitigates DSS-induced colitis, facilitates the restoration of mouse weight and colonic length, and diminishes the colonic weight and colonic weight index. Simultaneously, it hinders ulcer development and inflammatory cell infiltration in the colonic mucous membrane. While the percentage of Th1, mTh1, Th7, mTh7, Th17, mTh17, Tfh1, mTfh1, Tfh7, mTfh7, Tfh17, and mTfh17 cells decreased after Cur treatment of the mice for 7 d, and the frequency of mTh10, Th10, mTfh10, and Tfh10 cells in the mouse spleen increased. Further studies revealed that Cur administration prominently decreased the SOCS-1, SOCS-3, STAT3, p-STAT3, JAK1, p-JAK1, and NF-κB p65 protein expression levels in the colon tissue. CONCLUSION Cur regulated the mTh/mTfh cell homeostasis to reduce DSS-induced colonic pathological damage, potentially by suppressing the JAK1/STAT3/SOCS signaling pathway.
Collapse
Affiliation(s)
- Lin-Xin Zheng
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Kai-En Guo
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Jia-Qi Huang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Miao-Hua Liu
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Bai-Ling Deng
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Duan-Yong Liu
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Bu-Gao Zhou
- Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Wen Zhou
- Nanchang Medical College, Nanchang 330052, Jiangxi Province, China
| | - You-Bao Zhong
- Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Hai-Mei Zhao
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| |
Collapse
|
31
|
Niu Y, Zhang J, Shi D, Zang W, Niu J. Glycosides as Potential Medicinal Components for Ulcerative Colitis: A Review. Molecules 2023; 28:5210. [PMID: 37446872 DOI: 10.3390/molecules28135210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic, non-specific disease of unknown etiology. The disease develops mainly in the rectum or colon, and the main clinical symptoms include abdominal pain, diarrhea, and purulent bloody stools, with a wide variation in severity. The specific causative factors and pathogenesis of the disease are not yet clear, but most scholars believe that the disease is caused by the interaction of genetic, environmental, infectious, immune, and intestinal flora factors. As for the treatment of UC, medications are commonly used in clinical practice, mainly including aminosalicylates, glucocorticoids, and immunosuppressive drugs. However, due to the many complications associated with conventional drug therapy and the tendency for UC to recur, there is an urgent need to discover new, safer, and more effective drugs. Natural compounds with biodiversity and chemical structure diversity from medicinal plants are the most reliable source for the development of new drug precursors. Evidence suggests that glycosides may reduce the development and progression of UC by modulating anti-inflammatory responses, inhibiting oxidative stress, suppressing abnormal immune responses, and regulating signal transduction. In this manuscript, we provide a review of the epidemiology of UC and the available drugs for disease prevention and treatment. In addition, we demonstrate the protective or therapeutic role of glycosides in UC and describe the possible mechanisms of action to provide a theoretical basis for preclinical studies in drug development.
Collapse
Affiliation(s)
- Yating Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| | - Jun Zhang
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Dianhua Shi
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Weibiao Zang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianguo Niu
- School of Basic Medical Science, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
32
|
Li X, Li Z, Dong X, Wu Y, Li B, Kuang B, Chen G, Zhang L. Astragaloside IV attenuates myocardial dysfunction in diabetic cardiomyopathy rats through downregulation of CD36-mediated ferroptosis. Phytother Res 2023. [PMID: 36882189 DOI: 10.1002/ptr.7798] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/09/2023]
Abstract
Diabetic cardiomyopathy (DCM), one of the major complications of type 2 diabetes, is a leading cause of heart failure and death in advanced diabetes. Although there is an association between DCM and ferroptosis in cardiomyocytes, the internal mechanism of ferroptosis leading to DCM development remains unknown. CD36 is a key molecule in lipid metabolism that mediates ferroptosis. Astragaloside IV (AS-IV) confers various pharmacological effects such as antioxidant, anti-inflammatory, and immunomodulatory. In this study, we demonstrated that AS-IV was able to recover the dysfunction of DCM. In vivo experiments showed that AS-IV ameliorated myocardial injury and improved contractile function, attenuated lipid deposition, and decreased the expression level of CD36 and ferroptosis-related factors in DCM rats. In vitro experiments showed that AS-IV decreased CD36 expression and inhibited lipid accumulation and ferroptosis in PA-induced cardiomyocytes. The results demonstrated that AS-IV decreased cardiomyocyte injury and myocardial dysfunction by inhibiting ferroptosis mediated by CD36 in DCM rats. Therefore, AS-IV regulated the lipid metabolism of cardiomyocytes and inhibited cellular ferroptosis, which may have potential clinical value in DCM treatment.
Collapse
Affiliation(s)
- Xin Li
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Ziwei Li
- Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin Dong
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Yu Wu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Baohua Li
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Bin Kuang
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, China
| | - Gangyi Chen
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Liangyou Zhang
- The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
33
|
Li X, Jiang M, Chen X, Sun W. Etanercept alleviates psoriasis by reducing the Th17/Treg ratio and promoting M2 polarization of macrophages. Immun Inflamm Dis 2022; 10:e734. [PMID: 36444619 PMCID: PMC9639465 DOI: 10.1002/iid3.734] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/16/2022] [Accepted: 10/16/2022] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION This study aimed to investigate the effect of etanercept in psoriasis and its underlying mechanism. METHODS Female mice were treated with imiquimod (IMQ) to induce psoriasis, and intraperitoneally administered etanercept (0.1-0.4 mg/ml). The RAW264.7 cells were treated with LPS and IFN-γ to polarize to M1, and were treated with IL-13 and IL-4 to polarize to M2. RESULTS In our study, Etanercept markedly reduced the psoriasis area and severity index scores, and epidermal thickness of mice induced by IMQ. In addition, etanercept reduced the levels of TNF-α and IL-6/12/23, and enhanced the levels of IL-4/10, reduced Th17/Treg ratio and facilitated the polarization of macrophages to M2 in psoriasis model mice. Furthermore, etanercept inhibited the JAK/STAT3 pathway and increased the protein levels of SOCS1 and SOCS3. CONCLUSIONS In conclusion, our findings indicated that etanercept could inhibit the JAK/STAT3 pathway to reduce Th17/Treg ratio and promote M2 polarization, thereby alleviating psoriasis of mice.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of DermatologyThe Affiliated Huaian No.1 Peoples Hospital of Nanjing Medical UniversityHuai'anJiangsuP.R. China
| | - Ming Jiang
- Department of DermatologyThe Affiliated Huaian No.1 Peoples Hospital of Nanjing Medical UniversityHuai'anJiangsuP.R. China
| | - Xia Chen
- Department of PathologyThe Affiliated Huaian No.1 Peoples Hospital of Nanjing Medical UniversityHuai'anJiangsuP.R. China
| | - Weiguo Sun
- Department of DermatologyThe Affiliated Huaian No.1 Peoples Hospital of Nanjing Medical UniversityHuai'anJiangsuP.R. China
| |
Collapse
|
34
|
Puppala ER, Aochenlar SL, Shantanu PA, Ahmed S, Jannu AK, Jala A, Yalamarthi SS, Borkar RM, Tripathi DM, Naidu VGM. Perillyl alcohol attenuates chronic restraint stress aggravated dextran sulfate sodium-induced ulcerative colitis by modulating TLR4/NF-κB and JAK2/STAT3 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154415. [PMID: 36070663 DOI: 10.1016/j.phymed.2022.154415] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/10/2022] [Accepted: 08/21/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is the most prevalent chronic inflammatory immune bowel disease. The modernization of lifestyle accompanied by the stress to cope with the competition has resulted in a new range of complications where stress became a critical contributing factor for many diseases, including UC. Hence there is an urgent need to develop a dual role in curtailing both systemic and neuroinflammation. Perillyl alcohol (POH) is a natural essential oil found in lavender, peppermint, cherries etc and has been widely studied for its strong anti-inflammatory, antioxidant and anti-stress properties. HYPOTHESIS/PURPOSE POH regulates the various inflammatory signaling cascades involved in chronic inflammation by inhibiting farnesyltransferase enzyme. Several studies reported that POH could inhibit the phosphorylation of NF-κB, STAT3 and promote the endogenous antioxidant enzymes like Nrf2 via farnesyltransferase enzyme inhibition. Also, the effects of POH against UC is not known yet. Thus, this study aims to explore the anti-ulcerative properties of POH on stress aggravated ulcerative colitis in C57BL/6 mice. METHODS Ulcerative colitis was induced by duel exposure of chronic restraint stress (day 1 to day 28) and 2.5% dextran sulphate sodium (day8 to day14) in mice. POH treatment 100 and 200 mg/kg was administred from day14 ti day28 following oral route of administration. Disease activity index, colonoscopy, western blot analysis and histological analysis, neurotransmitter analysis and Gene expression studies were perofomerd to asses the anti-colitis effects of POH. RESULTS The treatment reversed the oxidative stress and inflammatory response by inhibiting TLR4/NF-kB pathway, and IL-6/JAK2/STAT3 pathway in both isolated mice colons and brains. The inhibition of these pathways resulted in a decrease in pro-inflammatory cytokines like IL-6, IL-1β and TNF-α. The treatment improved the physiological and histological changes with decreased ulcerations as examined by colonic endoscopy and Haematoxylin and Eosin staining. The treatment also improved the behavior response as it increased mobility time which was reduced by chronic restrained stress. This was due to increased satiety neurotransmitters like dopamine and serotonin and decreased cortisol in mice brains. CONCLUSION These results infer that POH has significant anti-colitis activity on chronic restraint stress aggravated DSS-induced UC in mice.
Collapse
Affiliation(s)
- Eswara Rao Puppala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - Sunepjungla L Aochenlar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - P A Shantanu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - Sahabuddin Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - Arun Kumar Jannu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - Aishwarya Jala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - Sai Sudha Yalamarthi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101
| | - Dinesh Mani Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary sciences (ILBS), New Delhi, Delhi 110070
| | - V G M Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam, India, 781101.
| |
Collapse
|