1
|
Yang HY, He YL, Chen JJ, Gao K. Naturally Occurring Cleistanthane Diterpenoids and Their Biological Activities. Chem Biodivers 2024:e202402268. [PMID: 39648155 DOI: 10.1002/cbdv.202402268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Cleistanthane diterpenoids, an important class of active natural products, are characterized by a 6/6/6 tricycle system and contain a variety of functional groups. Studies have shown that cleistanthane diterpenoids are widespread in various higher plants and endophytic fungi and exhibit a wide range of biological activities. This review provides extensive coverage of naturally occurring cleistanthane diterpenoids discovered from 1973 to 2024 and sheds light on the sources, structures, biological effects, and primary molecular mechanisms of them and provides a useful reference for the further study and development of these compounds.
Collapse
Affiliation(s)
- Hong-Ying Yang
- Research Institute, Lanzhou Jiaotong University, Lanzhou, People's Republic of China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, People's Republic of China
| | - Yi-Lin He
- Research Institute, Lanzhou Jiaotong University, Lanzhou, People's Republic of China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, People's Republic of China
| | - Jian-Jun Chen
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, People's Republic of China
| | - Kun Gao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, People's Republic of China
| |
Collapse
|
2
|
Wang D, Jin Y, Yang M, Xue Y, Zhang X, Guo Y, Li X, Ma K. Cardioprotective effect of Saussurea involucrata injection against Doxorubicin-induced cardiotoxicity by network pharmacology analysis and experimental verification. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39632659 DOI: 10.3724/abbs.2024170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Doxorubicin (Dox) is widely utilized in the clinical treatment of various cancers. Despite its efficacy, Dox induces numerous adverse effects in humans with significant cardiotoxicity, posing a major limitation to its use. Saussurea involucrata injection (SII), derived from Saussurea involucrata, exhibits notable anti-inflammatory and anti-oxidative stress properties. However, its potential protective effects against Dox-induced cardiotoxicity (DIC) remain unexplored. In this study, we investigate the ability of SII to mitigate DIC and elucidate the underlying mechanisms through experimental research and network pharmacology analysis. Results from both in vitro and in vivo experiments reveal that SII treatment significantly improves Dox-induced cardiac dysfunction, reducing pathological alterations and fibrosis in cardiomyocytes. Moreover, SII has cardioprotective effects by diminishing the inflammation, oxidative stress, and apoptosis triggered by Dox. Network pharmacological analysis further shows that SII downregulates P53 protein expression by activating the AKT/MDM2 signaling pathway, thus attenuating DIC. In conclusion, this study confirms that SII mitigates DIC through downregulation of the AKT/MDM2/P53 signaling pathway, suggesting a promising therapeutic strategy for alleviating DIC.
Collapse
Affiliation(s)
- Ding Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yu Jin
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Mengyu Yang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yajing Xue
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Xiaotong Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yanli Guo
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| |
Collapse
|
3
|
Liu JJ, Yang JB, Wang Y, Hu XR, Wang YD, Nie LX, Wei F, Yu JD, Yao LW, Xu BL, Ma SC, Jin HY. Integrating network pharmacology and experimental validation to investigate the effects and mechanism of Renshen Shouwu decoction for ameliorating Alzheimer's disease. PHARMACEUTICAL BIOLOGY 2024; 62:767-780. [PMID: 39417324 PMCID: PMC11488172 DOI: 10.1080/13880209.2024.2415660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
CONTEXT The mechanism of Renshen Shouwu Decoction (RSSW) in treating Alzheimer's disease (AD) remains unknown. OBJECTIVE This study investigates the effects and mechanism of RSSW for ameliorating AD. MATERIALS AND METHODS Ten SAMR1 mice and 40 SAMP8 mice were divided into five groups: control (SAMR1), model (SAMP8), positive drug (Donepezil, 1.3 mg/kg/d), and RSSW (Low-dose, 117 mg/kg/d; High-dose, 234 mg/kg/d). Starting from 6 months of age, the medications were administered intragastrically for a total of 60 days. Subsequently, memory improvement in rapidly aging mice was assessed using the novel object recognition test and Morris water maze test. Through the identification of absorbed blood components and analysis of network pharmacology, active ingredients and potential targets involved in the treatment of AD were identified. Finally, AD-related biological indicators were detected using western blotting and ELISA. RESULT Our results demonstrated that RSSW effectively ameliorated memory impairments, inhibited tau hyperphosphorylation, and reduced β-amyloid plaque deposition in SAMP8 mice. Thirty absorbed blood components in RSSW were identified, revealing identified 96 major targets that play a key role in alleviating AD. Notably, the obtained main targets were highly enriched in SIRT1-mediated signaling pathways. Subsequent experimental validation confirmed that RSSW activated the SIRT1/NF-κB, SIRT1/AMPK, and SIRT1/p53 signaling cascades. Nine potential active ingredients were predicted through molecular docking. DISCUSSION AND CONCLUSIONS Our research findings suggest the mechanism of RSSW treatment for AD, which ameliorates memory impairments by reducing cortical tissue inflammation and apoptosis.
Collapse
Affiliation(s)
- Jing-jing Liu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Institutes for Food and Drug Control, Beijing, China
| | - Jian-bo Yang
- National Institutes for Food and Drug Control, Beijing, China
| | - Ying Wang
- National Institutes for Food and Drug Control, Beijing, China
| | - Xiao-ru Hu
- National Institutes for Food and Drug Control, Beijing, China
| | - Ya-dan Wang
- National Institutes for Food and Drug Control, Beijing, China
| | - Li-xing Nie
- National Institutes for Food and Drug Control, Beijing, China
| | - Feng Wei
- National Institutes for Food and Drug Control, Beijing, China
| | - Jian-dong Yu
- National Institutes for Food and Drug Control, Beijing, China
| | - Ling-wen Yao
- National Institutes for Food and Drug Control, Beijing, China
| | - Bei-lei Xu
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Shuang-cheng Ma
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Institutes for Food and Drug Control, Beijing, China
| | - Hong-yu Jin
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Lan Y, Lin X, Rao Y, Huang Z. Improving access to domestic innovative medicines: characteristics and trends of approved drugs in China 2010-2024. Drug Discov Today 2024; 29:104240. [PMID: 39542205 DOI: 10.1016/j.drudis.2024.104240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
China has initiated drug regulatory reforms since 2015. Here, we analyze the characteristics and trends of domestic innovative drugs approved for marketing in China from January 2010 to May 2024 to explore the effectiveness of drug regulatory reform. Overall, 219 drugs were approved, with growth in chemicals and therapeutic biologics post-reform. Single-arm trials as an important option for clinical trial design of antineoplastic agents increased. The time for each link from investigational new drug (IND) to new drug application (NDA) has been shortened post-reform. Moreover, the time for access to medical insurance for approved drugs has been shortened and price reductions have been increased. China's drug regulatory reforms have made progress in improving the accessibility of domestic innovative drugs.
Collapse
Affiliation(s)
- Yipeng Lan
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofeng Lin
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanmei Rao
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhe Huang
- School of Business Administration, Shenyang Pharmaceutical University, Shenyang, China; Institute of Drug Regulatory Science, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
5
|
Liu XX, Ma YQ, Kong LY, Su YZ, Robinson N, Liu JP. Unveiling the therapeutic role of Dachaihu decoction in acute cholecystitis: a comprehensive systematic review and meta-analysis of its efficacy and safety. Front Pharmacol 2024; 15:1497072. [PMID: 39664516 PMCID: PMC11631619 DOI: 10.3389/fphar.2024.1497072] [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: 09/16/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Background Dachaihu decoction (Dachaihu tang) plays a crucial role in treating acute illnesses. Recently, a significant number of clinical studies on Dachaihu decoction for acute cholecystitis (AC) have been published. This study was conducted to assess the efficacy and safety of Dachaihu decoction in patients with this condition. Methods To identify relevant randomized controlled trials (RCTs), eight databases and three clinical trial registries were searched from inception to 30 June 2024. Two researchers independently screened and extracted data from eligible studies using EndNote X9 and Microsoft Office Excel 2019. RoB 2.0 was used to assess the risk of bias in the included studies. Stata 17.0 was used for data analysis. Publication bias and its impact on result stability were evaluated using a funnel plot and the "trim-and-fill" method. The quality of evidence was graded using the GRADE assessment system. Results Thirty-three RCTs involving 2,851 participants were included. The treatment group demonstrated improved clinical efficacy (RR = 1.18; 95% CI = 1.13 to 1.24), significantly reduced length of hospital stay (MD = -1.78 days; 95% CI = -2.02 to -1.53), and the incidence of adverse events (RR = 0.31; 95% CI = 0.20 to 0.48). Additionally, there appeared to be reductions in the time for abdominal pain to resolve (MD = -1.92 days; 95% CI = -2.33 to -1.51), fever to disappear (MD = -1.52 days; 95% CI = -1.90 to -1.14), white blood cell count to return to normal (MD = -2.89 days; 95% CI = -3.32 to -2.46), alanine aminotransferase (ALT) levels (MD = -11.88 U/L; 95% CI = -15.29 to -8.47), aspartate aminotransferase (AST) levels (MD = -8.74 U/L; 95% CI = -9.76 to -7.72), neutrophil percentage (MD = -9.68; 95% CI = -11.33 to -8.03), TNF-α levels (SMD = -2.10 pg/L; 95% CI = -2.43 to -2.78), and certainty of evidence (moderate-to-low certainty). Conclusion Dachaihu decoction may be an effective botanical formula for managing AC and a lower incidence of adverse events. However, due to the substantial risk of bias and heterogeneity across the included studies, these findings should be interpreted with caution and require further validation through well-designed, high-quality trials. Systematic Review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=573332.
Collapse
Affiliation(s)
- Xin-xin Liu
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying-qi Ma
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ling-yao Kong
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - You-zhu Su
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nicola Robinson
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- School of Health and Social Care, London South Bank University, London, United Kingdom
| | - Jian-ping Liu
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Department of Community Medicine, The National Research Center in Complementary and Alternative Medicine (NAFKAM), Faculty of Health Science, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
6
|
Tang Y, Luo J, Qin L, Tang C, Qiu C, Li J, Qin L. Network Pharmacology and Molecular Docking-Based Screening of Immunotherapeutic Targets for HuaChanSu Against Breast Cancer. Mol Biotechnol 2024:10.1007/s12033-024-01305-4. [PMID: 39565543 DOI: 10.1007/s12033-024-01305-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/10/2024] [Indexed: 11/21/2024]
Abstract
Breast cancer has emerged as the primary cause of mortality stemming from malignancies among women. HuaChanSu has demonstrated efficacy in suppressing the progression of various malignancies. However, the specific immune targets and pathways influenced by HuaChanSu within mammary tumors remain elusive. This study is designed to uncover potent monomers and pivotal targets associated with HuaChanSu's anti-breast cancer Immunotherapy. The genes pertinent to HuaChanSu and breast cancer were acquired individually from publicly available databases. Interaction analysis using Cytoscape was conducted on common genes to determine the most suitable targets and crucial constituents of HuaChanSu's Immunotherapy against breast cancer. Following this, molecular docking was employed to validate ligand and receptor binding interactions. Lastly, the identified core genes underwent assessment of immune infiltration. The intersection of HuaChanSu and BC targets yielded a total of 49 differentially expressed genes. Bufalin emerged as the most potent constituent in Immunotherapy. Immunoassay data demonstrated significant correlations (r > 0.03, p < 0.05) between S100B, MMP9, FOS, EGFR, KIT, MME, and immune infiltration within BC. Molecular docking further corroborated the effective binding of Bufalin with immune-related genes. Through network pharmacological validation, we propose the extraction of Bufalin, a monomeric constituent of Huachansu, to exert immunomodulatory effects aimed at inhibiting the progression of breast cancer. Most of the target genes (S100B, BIRC5, MMP9, FOS, EGFR, KIT, and MME) are common targets for immunotherapy.
Collapse
Affiliation(s)
- Yujun Tang
- Guangxi Medical University, Nanning, China
| | - Jie Luo
- Guangxi Medical University, Nanning, China
- HengyangMedicaSchool, University of South China, HengYang, China
| | | | | | - Caixin Qiu
- Guangxi Medical University, Nanning, China
| | - Jiehua Li
- Guangxi Medical University, Nanning, China.
| | | |
Collapse
|
7
|
Wu Y, Guo F, Li J, Shi W, Song L, Liu J. Curcumin ameliorates heatstroke-induced lung injury by activating the PI3K/AKT pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03572-z. [PMID: 39521756 DOI: 10.1007/s00210-024-03572-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Heatstroke (HS) poses a significant threat to public health. Curcumin, a polyphenolic compound, has been reported to possess anti-inflammatory and antioxidant properties. This study aimed to investigate the potential therapeutic effects of curcumin on HS-induced lung injury and to elucidate its underlying molecular mechanisms. We utilized network pharmacology to predict the potential targets of curcumin and determine its possible protective effects against HS. Molecular docking was performed to assess the affinity of curcumin to proteins. Forty mice were used for in vivo experiments to evaluate the therapeutic effects of curcumin, divided into four groups (n = 10 per group): normal control (NC), high-temperature control (HTC), low-dose curcumin heatstroke (H100c, 100 mg/kg/day), and high-dose curcumin heatstroke (H200c, 200 mg/kg/day). Furthermore, we evaluated lung pathology, ultrastructural alterations, and protein expression levels of key molecules. Molecular docking indicated a high binding affinity between curcumin and PIK3R1, AKT, and CASP3. In vivo experiments confirm that curcumin pretreatment significantly mitigates HS-induced lung tissue pathology and ultrastructural damage, with the H200c group showing notably greater improvement. Furthermore, curcumin pretreatment markedly enhances the activation of the PI3K/AKT pathway and suppresses the expression of cleaved caspase3, particularly in the H200c group. Our study suggests curcumin may alleviate HS-induced lung injury via the PI3K/AKT pathway, but limitations exist. We did not test key protein knockdown/overexpression, and PI3K/AKT may not be the only pathway. Human and mouse pharmacokinetic differences could affect clinical translation.
Collapse
Affiliation(s)
- Yizhan Wu
- Department of Graduate School, Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Fei Guo
- Department of Emergency Trauma Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang Uygur Autonomous Region, China
| | - Jiajia Li
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command of the PLA, No. 359 Youhao North Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Wenhui Shi
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command of the PLA, No. 359 Youhao North Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Laiyang Song
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command of the PLA, No. 359 Youhao North Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command of the PLA, No. 359 Youhao North Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
8
|
Wang J, Liu Y, Xiu C, Wang X, Liu Y, Hu Y, Yang J, Lei Y. Network Pharmacology-Based Strategy to Explore the Effect and Mechanism of Zhizhu Granule Improving Glucose-Lipid Metabolism in Rats with Metabolic Syndrome. Diabetes Metab Syndr Obes 2024; 17:3833-3846. [PMID: 39440025 PMCID: PMC11495215 DOI: 10.2147/dmso.s477410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Objective To explore the mechanism of the traditional Chinese medicine (TCM), Zhizhu granule (ZZG), in treating metabolic syndrome (MS) based on network pharmacology and pharmacodynamic experiment. Materials and Methods Network pharmacology combined with a pharmacodynamic experiment was used to elucidate the therapeutic mechanism of ZZG in MS. Serum samples were collected from rats with MS, induced by a high-sugar-fat-salt diet (HSFSD) combined with streptozotocin (STZ), to measure the levels of biochemical markers. The glucose (GLU), total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), systolic blood pressure (SBP), and diastolic blood pressure (DBP) were detected. The liver tissue of rats was used for histological examination and Western blot analysis. Results Network pharmacology analysis generated 69 drug-disease common targets and 10 hub genes closely related to ZZG against MS. KEGG pathway analysis revealed that the PI3K/AKT signaling pathway was the most potential pathway, which took part in the therapeutic mechanisms. In the animal experiments section, the therapeutic effect of ZZG on MS and the therapeutic pathway of ZZG on MS were verified. ZZG could significantly decrease the body weight, TC, TG, LDL-C and GLU levels in MS rats (all p<0.01), alleviate hepatocyte steatosis and decrease liver lipid droplet deposition. Western blot analysis indicated that compared with the model group, the expression levels of PI3K, AKT, and IRS-1 protein were significantly increased (all p<0.05), and the FOXO-1 was significantly decreased (all p<0.05) in the ZZG group. Conclusion ZZG can improve glucose-lipid metabolism disorder in rats with metabolic syndrome. The reported results provide experimental evidence for ZZG in the treatment of MS.
Collapse
Affiliation(s)
- Jiali Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People’s Republic of China
| | - Yiqing Liu
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Chengkui Xiu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Xue Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yinan Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yanhong Hu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Jing Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yan Lei
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
9
|
Liu X, Ding H, Chen M, Li X, Xiao Y, Han Y, Zeng H. Shenfu Injection Mediated NLRP3/Caspase 1 Through (R)-Norcoclaurinee Alleviates Sepsis-Induced Cognitive Dysfunction. J Inflamm Res 2024; 17:7295-7310. [PMID: 39429846 PMCID: PMC11488353 DOI: 10.2147/jir.s481171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
Background Shenfu injection (SF) has demonstrated its potential to enhance cellular immunity and induce clinical regression in patients suffering from sepsis or infectious shock. However, the therapeutic effect of SF on sepsis-induced cognitive dysfunction (SAE) and the mechanisms involved are still unclear. We aimed to investigate the mechanism of SF in mice with SAE. Methods Sepsis was constructed by caecal ligation and puncture. Mice were injected intraperitoneally with SF or NLRP3 inhibitor. The hippocampus injury of brain tissues was evaluated, and the levels of inflammatory cytokines (IL-1β, IL-18) and NLRP3 and Caspase 1 were measured. The active ingredients of SF were analyzed using network pharmacology, and molecular docking of the active ingredients of SF with NLRP3 and Caspase 1 was performed. BV-2 cells were treated with LPS or norcoclaurine. CCK-8 detected the cell viability, and the levels of inflammatory cytokines and NLRP3 and Caspase 1 were measured. Results SF and NLRP3 inhibitor increased survival rate and the number of crossing the platform and decreased the escape latency time of sepsis mice. Moreover, SF and NLRP3 inhibitor improved neuronal damage and apoptosis in hippocampus of sepsis mice. In addition, SF and NLRP3 inhibitor reduced the levels of inflammatory cytokines, as well as inflammasomes in sepsis mice. There were 43 active ingredients in SF. Among them, 22 were Renshen and 21 were Fuzi. Renshen and Fuzi, the main active components of SF, form a complex regulatory network with NLRP3 and Caspase 1. (R)-norcoclaurine was most closely bound to NLRP3 with binding energy of -7.2 kJ·mol-1, ignavine was most closely bound to Caspase 1 with binding energy of -8.3 kJ·mol-1. Norcoclaurine increased the cell viability and decreased inflammation and pyroptosis. Conclusion SF regulated NLRP3/Caspase 1 through (R)-norcoclaurinee to prevent SAE.
Collapse
Affiliation(s)
- Xinqiang Liu
- Department of Intensive Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510800, People’s Republic of China
| | - Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510800, People’s Republic of China
| | - Miner Chen
- Department of Intensive Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510800, People’s Republic of China
| | - Xusheng Li
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510800, People’s Republic of China
| | - Yan Xiao
- Department of Intensive Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510800, People’s Republic of China
| | - Yongli Han
- Department of Intensive Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510800, People’s Republic of China
| | - Hongke Zeng
- Department of Intensive Care Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510800, People’s Republic of China
| |
Collapse
|
10
|
Huang X, Peng Y, Lu L, Gao L, Wu S, Lu J, Liu X. Huangqi-Danshen Decoction Against Renal Fibrosis in UUO Mice via TGF-β1 Induced Downstream Signaling Pathway. Drug Des Devel Ther 2024; 18:4119-4134. [PMID: 39296670 PMCID: PMC11410030 DOI: 10.2147/dddt.s457100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
Background Huangqi-Danshen decoction (HDD) is a Chinese medicinal herb pair with good efficacy in treating chronic kidney disease, but its mechanism needs to be clarified. Aim To uncover the underlying mechanism of HDD antagonizing renal fibrosis through network pharmacology (NP) analysis and experimental validation. Materials and Methods The chemical components of water extract of HDD were analyzed by combining the ultra-high performance liquid chromatography coupled with Q-Exactive mass spectrum analysis (UHPLC-QE-MS) and HERB database. NP was used to identify core common targets of HDD components and renal fibrosis. Subsequently, male C57BL/6 mice were divided into Sham, unilateral ureteral obstruction (UUO) and UUO+HDD groups. Renal function, histopathology, Western blotting, and immunohistochemistry analyses were used to evaluate the protective effect of HDD on UUO mice. The effects of HDD on signaling pathways were validated in both UUO mice and transforming growth factor-β1 (TGF-β1)-induced HK-2 cells. Results By combining UHPLC-QE-MS analysis and HERB database, 25 components were screened in HDD extract. There were 270 intersection targets of the 25 components and renal fibrosis. Based on their scores in protein-protein interaction analysis and degree values in component-pathway-target triadic network, 6 core common targets of the 25 components and renal fibrosis were identified, namely phosphoinositide 3-kinase (PI3K), signal transducer and activator of transcription 3 (Stat3), non-receptor tyrosine kinase Src (Src), epidermal growth factor receptor (EGFR), matrix metalloproteinase 9 (MMP9), and MMP2. HDD ameliorated renal tubular damage and collagen deposition and downregulated fibrosis-related proteins expression in UUO mice. Furthermore, HDD was demonstrated to reduce PI3K, Stat3, Src, EGFR, and MMP2 expressions, and enhance MMP9 expression in the kidney of UUO mice and in TGF-β1-induced HK-2 cells. Conclusion HDD can alleviate renal fibrosis which may be related to regulating the expression of essential proteins in the epithelial-mesenchymal transition and extracellular matrix production/degradation signaling pathways.
Collapse
Affiliation(s)
- Xi Huang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Yu Peng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Lingfei Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Liwen Gao
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Shanshan Wu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Jiandong Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
| | - Xinhui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, People's Republic of China
| |
Collapse
|
11
|
Wang P, Gui X, Xu M, Dong F, Li Y, Wang Q, Wang Y, Yao J, Lu L, Liu R. In vivo and in vitro chemical composition and biological activity of traditional vs. dispensing granule decoctions of Coptidis Rhizoma: A comparative study. Biomed Chromatogr 2024; 38:e5960. [PMID: 38992861 DOI: 10.1002/bmc.5960] [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: 09/18/2023] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024]
Abstract
Coptidis Rhizoma (CR) holds significant clinical importance. In this study, we conducted a comparative analysis of CR's dispensing granule decoction (DGD) and traditional decoction (TD) to establish a comprehensive evaluation method for the quality of DGD. We selected nine batches of DGD (three from each of manufacturers A, B and C) and 10 batches of decoction pieces for analysis. We determined the content of representative components using high-performance liquid chromatography and assessed the content of blood components in vivo post-administration using ultra-performance liquid chromatography-mass spectrometry. The antibacterial activity was measured using the drug-sensitive tablet method. To evaluate the overall consistency of DGD and TD, we employed the CRITIC method and Grey relational analysis method. Our CRITIC results indicated no significant difference between the CRITIC scores of DGD-B and TD, with DGD-B exhibiting the highest consistency and overall quality. However, DGD-A and DGD-C showed variations in CRITIC scores compared with TD. After equivalent correction, the quality of DGD-A and DGD-C approached that of TD. Furthermore, our Grey relational analysis results supported the findings of the CRITIC method. This study offers a novel approach to evaluate the consistency between DGD and TD, providing insights into improving the quality of DGD.
Collapse
Affiliation(s)
- Panpan Wang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Henan Traditional Chinese Medicine Clinical Application, Evaluation and Transformation Engineering Research Center, Zhengzhou, Henan, China
- Key Laboratory of Chinese Medicine for Clinical Pharmacology of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Provincial-Ministry Collaborative Innovation Center for TCM Prevention and Treatment of Respiratory Diseases, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Xinjing Gui
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Henan Traditional Chinese Medicine Clinical Application, Evaluation and Transformation Engineering Research Center, Zhengzhou, Henan, China
- Key Laboratory of Chinese Medicine for Clinical Pharmacology of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Provincial-Ministry Collaborative Innovation Center for TCM Prevention and Treatment of Respiratory Diseases, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Manwen Xu
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Fengyu Dong
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yuanyuan Li
- Henan Medical College, Zhengzhou, Henan, China
| | - Qi Wang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yanli Wang
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Henan Traditional Chinese Medicine Clinical Application, Evaluation and Transformation Engineering Research Center, Zhengzhou, Henan, China
- Key Laboratory of Chinese Medicine for Clinical Pharmacology of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Provincial-Ministry Collaborative Innovation Center for TCM Prevention and Treatment of Respiratory Diseases, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Jing Yao
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Henan Traditional Chinese Medicine Clinical Application, Evaluation and Transformation Engineering Research Center, Zhengzhou, Henan, China
- Key Laboratory of Chinese Medicine for Clinical Pharmacology of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Provincial-Ministry Collaborative Innovation Center for TCM Prevention and Treatment of Respiratory Diseases, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Lu Lu
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Ruixin Liu
- Department of Pharmacy, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Henan Traditional Chinese Medicine Clinical Application, Evaluation and Transformation Engineering Research Center, Zhengzhou, Henan, China
- Key Laboratory of Chinese Medicine for Clinical Pharmacology of Traditional Chinese Medicine, Zhengzhou, Henan, China
- Provincial-Ministry Collaborative Innovation Center for TCM Prevention and Treatment of Respiratory Diseases, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Song Y, Long C, Chen W, Li H, Zhao H, Liu L. Cratoxylum formosum ssp. pruniflorum induces gastric cancer cell apoptosis and pyroptosis through the elevation of ROS and cell cycle arrest. Cell Biochem Biophys 2024; 82:2937-2955. [PMID: 39028496 DOI: 10.1007/s12013-024-01408-4] [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] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
Cratoxylum formosum ssp. pruniflorum (CF), a traditional medicinal plant in Southern China, is widely recognized as a popular medicinal and tea plant traditionally utilized by diverse linguistic groups in the region for the treatment of gastrointestinal ailments. The objective of this study was to explore the active components and mechanisms of CF against gastric cancer (GC). The chemical ingredients of CF were obtained by using UPLC-MS/MS-based metabolomics. MGC-803 and HGC-27 cells were employed to investigate the direct anti-GC effect. The potential targets and signaling pathway of CF were identified through network pharmacology and proteomics, followed by subsequent experimental validation. Through UPLC-MS/MS metabolomics analysis, a total of 197 chemical ingredients were identified in CF leaves. Network pharmacology and proteomics techniques revealed 25 potential targets for GC, with a protein-protein interaction (PPI) network highlighting 12 cores targets, including CTNNB1, CDK2, et al. Furthermore, seven key CF ingredients - vismione B, feruloylcholine, α-amyrin, vanillic acid, galangin, cinnamic acid, and caffeic acid - were found to mediate anti-GC effects through pathways such as reactive oxygen species (ROS) and cell cycle signaling pathway. In vitro experiments demonstrated that CF significantly inhibited the proliferation and migration of GC cells, increased intracellular reactive oxygen species (ROS), malondialdehyde (MDA) and lactate dehydrogenase (LDH) levels, arrested the cell cycle at the S-phase, induced apoptosis and pyroptosis, and upregulated expression of apoptosis proteins (Bax, Bax/Bcl-2, cleaved-Caspase-3/Caspase-3), and pyroptosis proteins (GSDMD-N/GSDMD and GSDME-N/GSDME), while downregulating expression of cell cycle proteins (CDK2 and cyclin A1) as well as necroptosis proteins (RIP1 and MLKL). Collectively, these findings reveal CF's therapeutic potential against GC by the augmentation of ROS production, cell cycle arrest, promotion of apoptosis, and pyroptosis, offering valuable evidence for the development and utilization of CF in clinical settings.
Collapse
Affiliation(s)
- Yaya Song
- Key Laboratory of Ecology and Environment in Minority Areas, Minzu University of China, National Ethnic Affairs Commission, Beijing, China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Chunlin Long
- Key Laboratory of Ecology and Environment in Minority Areas, Minzu University of China, National Ethnic Affairs Commission, Beijing, China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine, Minzu University of China, Ministry of Education, Beijing, China
| | - Weizhe Chen
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Hao Li
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Haofeng Zhao
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Liya Liu
- Key Laboratory of Ecology and Environment in Minority Areas, Minzu University of China, National Ethnic Affairs Commission, Beijing, China.
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
13
|
Qi Y, Zhou Q, Zhang Y, Deng J, Li R, Zhang X. Exploring the active components and potential mechanisms of Alpiniae oxyphyllae Fructus in treating diabetes mellitus with depression by UPLC-Q-Exactive Orbitrap/MS, network pharmacology and molecular docking. Metab Brain Dis 2024; 39:1065-1084. [PMID: 38954241 DOI: 10.1007/s11011-024-01374-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
The growing incidence of diabetes mellitus (DM) and depression is a global public health issue. Alpiniae oxyphyllae Fructus (AOF) is a kind of medicinal and edible plant which be found with anti-diabetic property, and could improve depression-like symptoms. This study aimed to screen active targets and potential mechanisms of AOF in treating DM with depression. Injection of streptozotocin (STZ) and exposure to chronic unpredictable mild stress (CUMS) for 4 weeks were used to conduct the DM with depression mice model. Behavioral tests, indexes of glucose metabolism, monoamine neurotransmitters, inflammatory cytokine and oxidative stress were measured. Histopathological change of hippocampus tissue was observing by HE and Nissl staining. UPLC-Q-Exactive Orbitrap/MS, network pharmacology and molecular docking were used to explore the chemical components and mechanisms of AOF on the DM with depression. AOF showed a reversed effect on body weight in DM with depression mice. Glucose metabolism and insulin resistance could be improved by treatment of AOF. In addition, AOF could alleviate depression-like behaviors based on the results of behavior tests and monoamine neurotransmitters. AOF also attenuated STZ-CUMS induced neuron injury in hippocampus. Next, a total of 61 chemical components were identified in the UPLC-Q-Exactive Orbitrap/MS analysis of the extract of AOF. Network pharmacology analysis suggested that 12 active components and 227 targets were screened from AOF, and 1802 target genes were screened from DM with depression, finally 126 intersection target genes were obtained. Drug-disease targets network was constructed and implied that the top five components with a higher degree value includes quercetin, nootkatone, baicalein, (-)-epicatechin and nootkatol. Protein-protein interaction (PPI) network showed that MAPK1, FOS, AKT1, IL6 and TP53 may be the core intersection targets. The mechanism of the effect of AOF on DM with depression was analyzed through gene ontology (GO), and kyoto encyclopedia of genes and genomes (KEGG) enrichment analysis, mainly involved in AGE/RAGE, PI3K/AKT, and MAPK signaling pathways. The results of molecular docking indicated that quercetin, nootkatone, baicalein, (-)-epicatechin and nootkatol all had good binding to the core intersection targets. Overall, our experimental researches have demonstrated that AOF could exert the dual effects of anti-diabetic and anti-depression on DM with depression mice, through multi-targets and multi-pathways.
Collapse
Affiliation(s)
- Yue Qi
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China
| | - Qilun Zhou
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China
| | - Yongping Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China
| | - Jinlan Deng
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China
| | - Ruonan Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China
| | - Xiaofeng Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
14
|
Zhang MY, Zheng SQ. Network pharmacology and molecular dynamics study of the effect of the Astragalus-Coptis drug pair on diabetic kidney disease. World J Diabetes 2024; 15:1562-1588. [PMID: 39099827 PMCID: PMC11292324 DOI: 10.4239/wjd.v15.i7.1562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/13/2024] [Accepted: 05/29/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) is the primary cause of end-stage renal disease. The Astragalus-Coptis drug pair is frequently employed in the management of DKD. However, the precise molecular mechanism underlying its therapeutic effect remains elusive. AIM To investigate the synergistic effects of multiple active ingredients in the Astragalus-Coptis drug pair on DKD through multiple targets and pathways. METHODS The ingredients of the Astragalus-Coptis drug pair were collected and screened using the TCMSP database and the SwissADME platform. The targets were predicted using the SwissTargetPrediction database, while the DKD differential gene expression analysis was obtained from the Gene Expression Omnibus database. DKD targets were acquired from the GeneCards, Online Mendelian Inheritance in Man database, and DisGeNET databases, with common targets identified through the Venny platform. The protein-protein interaction network and the "disease-active ingredient-target" network of the common targets were constructed utilizing the STRING database and Cytoscape software, followed by the analysis of the interaction relationships and further screening of key targets and core active ingredients. Gene Ontology (GO) function and Kyoto Ency-clopedia of Genes and Genomes (KEGG) pathway enrichments were performed using the DAVID database. The tissue and organ distributions of key targets were evaluated. PyMOL and AutoDock software validate the molecular docking between the core ingredients and key targets. Finally, molecular dynamics (MD) simulations were conducted to simulate the optimal complex formed by interactions between core ingredients and key target proteins. RESULTS A total of 27 active ingredients and 512 potential targets of the Astragalus-Coptis drug pair were identified. There were 273 common targets between DKD and the Astragalus-Coptis drug pair. Through protein-protein interaction network topology analysis, we identified 9 core active ingredients and 10 key targets. GO and KEGG pathway enrichment analyses revealed that Astragalus-Coptis drug pair treatment for DKD involves various biological processes, including protein phosphorylation, negative regulation of apoptosis, inflammatory response, and endoplasmic reticulum unfolded protein response. These pathways are mainly associated with the advanced glycation end products (AGE)-receptor for AGE products signaling pathway in diabetic complications, as well as the Lipid and atherosclerosis. Molecular docking and MD simulations demonstrated high affinity and stability between the core active ingredients and key targets. Notably, the quercetin-AKT serine/threonine kinase 1 (AKT1) and quercetin-tumor necrosis factor (TNF) protein complexes exhibited exceptional stability. CONCLUSION This study demonstrated that DKD treatment with the Astragalus-Coptis drug pair involves multiple ingredients, targets, and signaling pathways. We propose a novel approach for investigating the molecular mechanism underlying the therapeutic effects of the Astragalus-Coptis drug pair on DKD. Furthermore, we suggest that quercetin is the most potent active ingredient and specifically targets AKT1 and TNF, providing a theoretical foundation for further exploration of pharmacologically active ingredients and elucidating their molecular mechanisms in DKD treatment.
Collapse
Affiliation(s)
- Mo-Yan Zhang
- Liaoning University of Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang 110847, Liaoning Province, China
| | - Shu-Qin Zheng
- Department of Endocrinology, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning Province, China
| |
Collapse
|
15
|
Meng X, Li C, Gao A, Wang H, Wei L, Sun L. Integrated metabolomics and network pharmacology approach to exploring the anti-inflammatory mechanisms of Chuanwang xiaoyan capsules. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1242:124197. [PMID: 38889492 DOI: 10.1016/j.jchromb.2024.124197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Chuanwang xiaoyan capsules (CWXYC) have anti-inflammatory and detoxification effect, are used in the treatment of acute and chronic tonsillitis, pharyngitis and other inflammation-related diseases clinically. However, the anti-inflammatory mechanisms have not been elucidated. This study aimed to investigate the anti-inflammatory mechanisms of CWXYC using cell metabolomics and network pharmacology strategy. Specifically, CWXYC could efficiently reduce the content of nitric oxide (NO), the cytokines Interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) in LPS-induced RAW264.7 cells. Furthermore, metabolomics was performed to achieve 23 differential metabolites and 9 metabolic pathways containing glutamate metabolism, glutathione metabolism, arginine and proline metabolism, urea cycle, malate-aspartate shuttle, phosphatidylcholine biosynthesis, transfer of acetyl groups into mitochondria, cysteine metabolism and ammonia recycling. The results of network pharmacology showed that CWXYC could treat inflammation through 10 active components, 10 key targets and 55 pathways. Then the results of molecular docking also approved that there existed strong binding energy between the active components and the key targets. Finally, metabolomics and network pharmacology were integrated to get core targets AKT1, SRC and EGFR. Western blot experiments verified that CWXYC could exert anti-inflammatory effect by down-regulating the activated Akt1 and Src proteins. This study demonstrated that CWXYC exerted effects against inflammation, and the potential mechanisms were elucidated. These novel findings will provide an important basis for further mechanism investigations.
Collapse
Affiliation(s)
- Xiangping Meng
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Caihong Li
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Aichun Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Hongjin Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Lan Wei
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China.
| | - Lixin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China.
| |
Collapse
|
16
|
Zhao X, Wu X, Hu Q, Yao J, Yang Y, Wan M, Tang W. Yinchenhao Decoction Protects Against Acute Liver Injury in Mice With Biliary Acute Pancreatitis by Regulating the Gut Microflora-Bile Acids-Liver Axis. Gastroenterol Res Pract 2024; 2024:8882667. [PMID: 38966598 PMCID: PMC11223911 DOI: 10.1155/2024/8882667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 05/22/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Background and Aims: Acute liver injury (ALI) often follows biliary acute pancreatitis (BAP), but the exact cause and effective treatment are unknown. The aim of this study was to investigate the role of the gut microflora-bile acids-liver axis in BAP-ALI in mice and to assess the potential therapeutic effects of Yinchenhao decoction (YCHD), a traditional Chinese herbal medicine formula, on BAP-ALI. Methods: Male C57BL/6 mice were allocated into three groups: negative control (NC), BAP model, and YCHD treatment groups. The severity of BAP-ALI, intrahepatic bile acid levels, and the gut microbiota were assessed 24 h after BAP-ALI induction in mice. Results: Our findings demonstrated that treatment with YCHD significantly ameliorated the severity of BAP-ALI, as evidenced by the mitigation of hepatic histopathological changes and a reduction in liver serum enzyme levels. Moreover, YCHD alleviated intrahepatic cholestasis and modified the composition of bile acids, as indicated by a notable increase in conjugated bile acids. Additionally, 16S rDNA sequencing analysis of the gut microbiome revealed distinct alterations in the richness and composition of the microbiome in BAP-ALI mice compared to those in control mice. YCHD treatment effectively improved the intestinal flora disorders induced by BAP-ALI. Spearman's correlation analysis revealed a significant association between the distinct compositional characteristics of the intestinal microbiota and the intrahepatic bile acid concentration. Conclusions: These findings imply a potential link between gut microbiota dysbiosis and intrahepatic cholestasis in BAP-ALI mice and suggest that YCHD treatment may confer protection against BAP-ALI via the gut microflora-bile acids-liver axis.
Collapse
Affiliation(s)
- Xianlin Zhao
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineWest China HospitalSichuan University, Chengdu 610041, China
- West China School of MedicineSichuan University, Chengdu 610041, China
| | - Xiajia Wu
- West China School of MedicineSichuan University, Chengdu 610041, China
- Institute of Respiratory Health and MultimorbidityWest China HospitalSichuan University, Chengdu 610041, China
| | - Qian Hu
- West China School of MedicineSichuan University, Chengdu 610041, China
| | - Jiaqi Yao
- West China School of MedicineSichuan University, Chengdu 610041, China
| | - Yue Yang
- West China School of MedicineSichuan University, Chengdu 610041, China
| | - Meihua Wan
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineWest China HospitalSichuan University, Chengdu 610041, China
- West China School of MedicineSichuan University, Chengdu 610041, China
| | - Wenfu Tang
- West China Center of Excellence for PancreatitisInstitute of Integrated Traditional Chinese and Western MedicineWest China HospitalSichuan University, Chengdu 610041, China
- West China School of MedicineSichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Zhou Z, Nan Y, Li X, Ma P, Du Y, Chen G, Ning N, Huang S, Gu Q, Li W, Yuan L. Hawthorn with "homology of medicine and food": a review of anticancer effects and mechanisms. Front Pharmacol 2024; 15:1384189. [PMID: 38915462 PMCID: PMC11194443 DOI: 10.3389/fphar.2024.1384189] [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: 02/08/2024] [Accepted: 04/29/2024] [Indexed: 06/26/2024] Open
Abstract
Over the past few years, there has been a gradual increase in the incidence of cancer, affecting individuals at younger ages. With its refractory nature and substantial fatality rate, cancer presents a notable peril to human existence and wellbeing. Hawthorn, a medicinal food homology plant belonging to the Crataegus genus in the Rosaceae family, holds great value in various applications. Due to its long history of medicinal use, notable effects, and high safety profile, hawthorn has garnered considerable attention and plays a crucial role in cancer treatment. Through the integration of modern network pharmacology technology and traditional Chinese medicine (TCM), a range of anticancer active ingredients in hawthorn have been predicted, identified, and analyzed. Studies have shown that ingredients such as vitexin, isoorientin, ursolic acid, and maslinic acid, along with hawthorn extracts, can effectively modulate cancer-related signaling pathways and manifest anticancer properties via diverse mechanisms. This review employs network pharmacology to excavate the potential anticancer properties of hawthorn. By systematically integrating literature across databases such as PubMed and CNKI, the review explores the bioactive ingredients with anticancer effects, underlying mechanisms and pathways, the synergistic effects of drug combinations, advancements in novel drug delivery systems, and ongoing clinical trials concerning hawthorn's anticancer properties. Furthermore, the review highlights the preventive health benefits of hawthorn in cancer prevention, offering valuable insights for clinical cancer treatment and the development of TCM with anticancer properties that can be used for both medicinal and edible purposes.
Collapse
Affiliation(s)
- Ziying Zhou
- Department of Pharmacy, General Hospital of Ningxia Medical University, Yinchuan, China
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Xiangyang Li
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Ping Ma
- Department of Pharmacy, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Guoqing Chen
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Na Ning
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Shicong Huang
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Qian Gu
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Weiqiang Li
- Department of Chinese Medical Gastrointestinal, The Affiliated TCM Hospital of Ningxia Medical University, Wuzhong, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
18
|
Li X, Yang Y, Feng P, Wang H, Zheng M, Zhu Y, Zhong K, Hu J, Ye Y, Lu L, Zhao Q. Quercetin improves the protection of hydroxysafflor yellow a against cerebral ischemic injury by modulating of blood-brain barrier and src-p-gp-mmp-9 signalling. Heliyon 2024; 10:e31002. [PMID: 38803916 PMCID: PMC11128878 DOI: 10.1016/j.heliyon.2024.e31002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Protection of the structural and functional integrity of the blood-brain barrier (BBB) is crucial for treating ischemic stroke (IS). Hydroxysafflor yellow A (HSYA) and quercetin (Quer), two main active components in the edible and medicinal plant Carthamus tinctorius L., have been reported to exhibit neuroprotective effects. We investigated the anti-IS and BBB-protective properties of HSYA and Quer and the underlying mechanisms. They decreased neurological deficits in middle cerebral artery occlusion (MCAO) mice, while their combination showed better effects. Importantly, HSYA and Quer ameliorated BBB permeability. Their effects on reduction of both EB leakage and infarct volume were similar, which may contribute to improved locomotor activities. Moreover, HSYA and Quer showed protective effects for hCMEC/D3 monolayer against oxygen-glucose deprivation. Src, p-Src, MMP-9, and P-gp were associated with ingredients treatments. Furthermore, molecular docking and molecular dynamics simulations revealed stable and tight binding modes of ingredients with Src and P-gp. The current study supports the potential role of HSYA, Quer, and their combination in the treatment of IS by regulating BBB integrity.
Collapse
Affiliation(s)
- Xiang Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yuanxiao Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Pinpin Feng
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Hongwei Wang
- Department of Anesthesiology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Mingzhi Zheng
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yiliang Zhu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Kai Zhong
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Jue Hu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yilu Ye
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Linhuizi Lu
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Qinqin Zhao
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| |
Collapse
|
19
|
Peng Y, Zhou C. Network Pharmacology and Molecular Docking Identify the Potential Mechanism and Therapeutic Role of Scutellaria baicalensis in Alzheimer's Disease. Drug Des Devel Ther 2024; 18:1199-1219. [PMID: 38645989 PMCID: PMC11032720 DOI: 10.2147/dddt.s450739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 04/04/2024] [Indexed: 04/23/2024] Open
Abstract
Aim Scutellaria baicalensis, a traditional Chinese medicinal herb renowned for its anti-inflammatory, antioxidant, and anti-tumor properties, has shown promise in alleviating cognitive impairment associated with Alzheimer's disease. Nonetheless, the exact neuroprotective mechanism of Scutellaria baicalensis against Alzheimer's disease remains unclear. In this study, network pharmacology was employed to explore the possible mechanisms by which Scutellaria baicalensis protects against Alzheimer's disease. Methods The active compounds of Scutellaria baicalensis were retrieved from the TCMSP database, and their corresponding targets were identified. Alzheimer's disease-related targets were obtained through searches in the GeneCards and OMIM databases. Cytoscape 3.6.0 software was utilized to construct a regulatory network illustrating the "active ingredient-target" relationships. Subsequently, the target genes affected by Scutellaria baicalensis in the context of Alzheimer's disease were input into the String database to establish a PPI network. GO analysis and KEGG analysis were conducted using the DAVID database to predict the potential pathways associated with these key targets. Following this, the capacity of these active ingredients to bind to core targets was confirmed through molecular docking. In vitro experiments were then carried out for further validation. Results A total of 36 active ingredients from Scutellaria baicalensis were screened out, which corresponded to 365 targets. Molecular docking results demonstrated the robust binding abilities of Baicalein, Wogonin, and 5,2'-Dihydroxy-6,7,8-trimethoxyflavone to key target proteins (SRC, PIK3R1, and STAT3). In vitro experiments showed that the active components of Scutellaria baicalensis can inhibit STAT3 expression by downregulating the PIK3R1/SRC pathway in Neuro 2A cells. Conclusion In summary, these findings collectively suggest that Scutellaria baicalensis holds promise as a viable treatment option for Alzheimer's disease.
Collapse
Affiliation(s)
- Yutao Peng
- Department of Function, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, People’s Republic of China
| | - Chanjuan Zhou
- Department of Clinical Psychology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, People’s Republic of China
| |
Collapse
|
20
|
Zheng S, Xue C, Li S, Zao X, Li X, Liu Q, Cao X, Wang W, Qi W, Zhang P, Ye Y. Chinese medicine in the treatment of non-alcoholic fatty liver disease based on network pharmacology: a review. Front Pharmacol 2024; 15:1381712. [PMID: 38694920 PMCID: PMC11061375 DOI: 10.3389/fphar.2024.1381712] [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: 02/04/2024] [Accepted: 03/29/2024] [Indexed: 05/04/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a clinicopathological syndrome characterized by abnormalities in hepatic fat deposition, the incidence of which has been increasing year by year in recent years. It has become the largest chronic liver disease globally and one of the important causes of cirrhosis and even primary liver cancer formation. The pathogenesis of NAFLD has not yet been fully clarified. Modern medicine lacks targeted clinical treatment protocols for NAFLD, and most drugs lack efficacy and have high side effects. In contrast, Traditional Chinese Medicine (TCM) has significant advantages in the treatment and prevention of NAFLD, which have been widely recognized by scholars around the world. In recent years, through the establishment of a "medicine-disease-target-pathway" network relationship, network pharmacology can explore the molecular basis of the role of medicines in disease prevention and treatment from various perspectives, predicting the pharmacological mechanism of the corresponding medicines. This approach is compatible with the holistic view and treatment based on pattern differentiation of TCM and has been widely used in TCM research. In this paper, by searching relevant databases such as PubMed, Web of Science, and Embase, we reviewed and analyzed the relevant signaling pathways and specific mechanisms of action of single Chinese medicine, Chinese medicine combinations, and Chinese patent medicine for the treatment of NAFLD in recent years. These related studies fully demonstrated the therapeutic characteristics of TCM with multi-components, multi-targets, and multi-pathways, which provided strong support for the exact efficacy of TCM exerted in the clinic. In conclusion, we believe that network pharmacology is more in line with the TCM mindset of treating diseases, but with some limitations. In the future, we should eliminate the potential risks of false positives and false negatives, clarify the interconnectivity between components, targets, and diseases, and conduct deeper clinical or experimental studies.
Collapse
Affiliation(s)
- Shihao Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Chengyuan Xue
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Size Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoke Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qiyao Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Xu Cao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Wenying Qi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Peng Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yongan Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Zhu L, Liang R, Guo Y, Cai Y, Song F, Hu Y, Liu Y, Ge M, Zheng G. Incorporating Network Pharmacology and Experimental Validation to Identify Bioactive Compounds and Potential Mechanisms of Digitalis in Treating Anaplastic Thyroid Cancer. ACS OMEGA 2024; 9:15590-15602. [PMID: 38585091 PMCID: PMC10993403 DOI: 10.1021/acsomega.4c00373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2024]
Abstract
Anaplastic thyroid cancer (ATC) is one of the most lethal malignant tumors for which there is no effective treatment. There are an increasing number of studies on herbal medicine for treating malignant tumors, and the classic botanical medicine Digitalis and its active ingredients for treating heart failure and arrhythmias have been revealed to have significant antitumor efficacy against a wide range of malignant tumors. However, the main components of Digitalis and the molecular mechanisms of its anti-ATC effects have not been extensively studied. Here, we screened the main components and core targets of Digitalis and verified the relationship between the active components and targets through network pharmacology, molecular docking, and experimental validation. These experiments showed that the active ingredients of Digitalis inhibit ATC cell activity and lead to ATC cell death through the apoptotic pathway.
Collapse
Affiliation(s)
- Lei Zhu
- Suzhou
Medical College of Soochow University, 215123 Suzhou, Jiangsu, China
- Department
of Head and Neck Surgery, the Fifth Hospital Affiliated to Wenzhou
Medical University, Lishui Central Hospital, 323020 Lishui City, Zhejiang Province, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| | - Ruimin Liang
- Otolaryngology
& Head and Neck Center, Cancer Center, Department of Head and
Neck Surgery, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical
College, 310014 Hangzhou, Zhejiang, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| | - Yawen Guo
- Otolaryngology
& Head and Neck Center, Cancer Center, Department of Head and
Neck Surgery, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical
College, 310014 Hangzhou, Zhejiang, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| | - Yefeng Cai
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Department
of Thyroid Surgery, The First Affiliated
Hospital of Wenzhou Medical University, 325015 Wenzhou City, Zhejiang Province, China
| | - Fahuan Song
- Otolaryngology
& Head and Neck Center, Cancer Center, Department of Head and
Neck Surgery, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical
College, 310014 Hangzhou, Zhejiang, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| | - Yiqun Hu
- Otolaryngology
& Head and Neck Center, Cancer Center, Department of Head and
Neck Surgery, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical
College, 310014 Hangzhou, Zhejiang, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| | - Yunye Liu
- Otolaryngology
& Head and Neck Center, Cancer Center, Department of Head and
Neck Surgery, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical
College, 310014 Hangzhou, Zhejiang, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| | - Minghua Ge
- Suzhou
Medical College of Soochow University, 215123 Suzhou, Jiangsu, China
- Otolaryngology
& Head and Neck Center, Cancer Center, Department of Head and
Neck Surgery, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical
College, 310014 Hangzhou, Zhejiang, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| | - Guowan Zheng
- Otolaryngology
& Head and Neck Center, Cancer Center, Department of Head and
Neck Surgery, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical
College, 310014 Hangzhou, Zhejiang, China
- Key
Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
- Clinical
Research Center for Cancer of Zhejiang Province, 310014 Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Zhang J, Luo L, Guo Y, Liu A, Zhang M, Jiang W, Li X, Liu Q, Yu J. Pharmacological effects and target analysis of Guipi wan in the treatment of cerebral ischemia-reperfusion injury. Front Pharmacol 2024; 15:1346226. [PMID: 38515838 PMCID: PMC10955136 DOI: 10.3389/fphar.2024.1346226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
Guipi wan (GPW) is a traditional Chinese medicine commonly used in clinical practice, typically to treat neurological diseases such as neurasthenia and traumatic brain injury. It may have positive effects on cerebral ischemia‒reperfusion injury (cI/R). This study aimed to assess the effects of GPW in a mouse model of cI/R and find its possible targets. C57BL/6J mice were used to establish the cI/R model, and the laser speckle doppler was used to determine the success of the model. GPW was administered intragastrically for 7 days, brain tissue sections were stained with TTC, HE, and TUNEL, Western blot assay was performed to detect the effect of apoptosis-related proteins. Furthermore, we screened active ingredients from the TCM Database and constructed a compound‒target network using the Cytoscape 3.8.0 software. Moreover, we employed protein‒protein interaction and component‒target‒pathway network analyses to determine the potential components of GPW and its target genes, the key target was verified through molecular docking. Finally, we detected the influence of the downstream signaling pathway of the target through Western blot. The results showed that GPW decreased the cerebral infarction area, neurological function scores, and neuronal apoptosis in mice by regulating PI3K/AKT signaling pathway. Network analysis indicated that gamma-aminobutyric acid B receptor 1 (GABBR1) might be a potential target for the treatment of cI/R. Molecular docking indicated that 9 active components in GPW could bind to GABBR1 with desirable binding energy. This study represented the demonstratable effect of GPW in the treatment of cI/R injury and suggested GABBR1 as a potential target using network analysis.
Collapse
Affiliation(s)
- Jianfeng Zhang
- Department of Pharmacy, Eighth Hospital of Xi’an City, Xi’an, China
| | - Li Luo
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Yanyan Guo
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - An Liu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | | | - Wei Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Xi Li
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Qingqing Liu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Jiaoyan Yu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| |
Collapse
|
23
|
Zhu D, Li S, Xu L, Ren X, Wang S, Chen J, Zhao E, Zheng Z. Investigation of the molecular mechanism of Danggui Buxue tang in treating lung cancer using network pharmacology and molecular docking techniques. Nat Prod Res 2024:1-4. [PMID: 38403948 DOI: 10.1080/14786419.2024.2305660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/09/2024] [Indexed: 02/27/2024]
Abstract
This study used network pharmacology and molecular docking techniques to investigate the molecular targets and pathways of Danggui Buxue Tang (DBT) in treating lung cancer. The compound-target network was constructed using the Traditional Chinese Medicine Systems Pharmacology Database (TCMSP), and a lung cancer-specific network was created using the GEO database and Cytoscape software. GO and KEGG pathway analyses were performed to understand the biological processes associated with DBT. The key compounds from Astragalus, kaempferol, and quercetin, and the potential targets are IL-6, IL-1β, FOS, ICAM1, and CCL2. GO enrichment analysis revealed numerous biological process-related entries, while KEGG pathway analysis highlighted the TNF and IL-17 signalling pathways. Molecular docking confirmed the stable binding activity between the main active compounds of DBT and the target proteins. Overall, these findings shed light on the molecular mechanism of DBT in treating lung cancer, providing insights into targets, pathways, and biological processes involved.
Collapse
Affiliation(s)
- Dantong Zhu
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
| | - Shun Li
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, China
| | - Long Xu
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xijing Ren
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, China
| | - Shudong Wang
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
- School of Graduate Studies, Jinzhou Medical University, Jinzhou, China
| | - Jianjun Chen
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
| | - Ershu Zhao
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
- School of Graduate Studies, Dalian Medical University, Dalian, China
| | - Zhendong Zheng
- Oncology Department, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
24
|
Zhou E, Shen Q, Hou Y. Integrating artificial intelligence into the modernization of traditional Chinese medicine industry: a review. Front Pharmacol 2024; 15:1181183. [PMID: 38464717 PMCID: PMC10921893 DOI: 10.3389/fphar.2024.1181183] [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: 03/07/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
Traditional Chinese medicine (TCM) is the practical experience and summary of the Chinese nation for thousands of years. It shows great potential in treating various chronic diseases, complex diseases and major infectious diseases, and has gradually attracted the attention of people all over the world. However, due to the complexity of prescription and action mechanism of TCM, the development of TCM industry is still in a relatively conservative stage. With the rise of artificial intelligence technology in various fields, many scholars began to apply artificial intelligence technology to traditional Chinese medicine industry and made remarkable progress. This paper comprehensively summarizes the important role of artificial intelligence in the development of traditional Chinese medicine industry from various aspects, including new drug discovery, data mining, quality standardization and industry technology of traditional Chinese medicine. The limitations of artificial intelligence in these applications are also emphasized, including the lack of pharmacological research, database quality problems and the challenges brought by human-computer interaction. Nevertheless, the development of artificial intelligence has brought new opportunities and innovations to the modernization of traditional Chinese medicine. Integrating artificial intelligence technology into the comprehensive application of Chinese medicine industry is expected to overcome the major problems faced by traditional Chinese medicine industry and further promote the modernization of the whole traditional Chinese medicine industry.
Collapse
Affiliation(s)
- E. Zhou
- Yuhu District Healthcare Security Administration, Xiangtan, China
| | - Qin Shen
- Department of Respiratory Medicine, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Yang Hou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
25
|
Xia J, Chen J, Xing X, Meng J, Song X, Lou D. Dendrobine regulates STAT3 to attenuate mitochondrial dysfunction and senescence in vascular endothelial cells triggered by oxidized low-density lipoprotein. Drug Dev Res 2024; 85:e22152. [PMID: 38349255 DOI: 10.1002/ddr.22152] [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: 07/11/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/15/2024]
Abstract
Our previous studies have highlighted the potential therapeutic efficacy of dendrobine, an alkaloid, in atherosclerosis (AS), nevertheless, the underlying mechanism remains unclear. This study employs a combination of network pharmacology and in vitro experiments to explore the regulatory pathways involved. Through network pharmacology, the biological function for intersection targets between dendrobine and AS were identified. Molecular docking was conducted to investigate the interaction between the dominant target and dendrobine. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to mimic AS, and the effects of dendrobine on cell viability, lipid deposition, mitochondrial function, and cellular senescence were evaluated. Subsequently, cells were treated with the mitophagy inhibitor Mdivi-1 and the STAT3 agonist colivelin to assess the role of mitophagy and STAT3 signaling in dendrobine regulation. Intersection targets were associated with biological processes, including reactive oxygen species production. Dendrobine attenuated the effects of ox-LDL treatment on HUVECs, mitigating changes in cell activity, lipid deposition, mitochondrial function, and cellular senescence. Both Mdivi-1 and colivelin treatments resulted in decreased cell viability and increased cellular senescence, with colivelin suppressing mitophagy. Cotreatment with Mdivi-1 and colivelin further aggravated cellular senescence and inhibited FoxO signaling. Together, this study indicated that dendrobine regulated the STAT3/FoxO signaling pathway, alleviating mitochondrial dysfunction and cellular senescence. This study contributes valuable insights to the potential clinical application of dendrobine.
Collapse
Affiliation(s)
- Jia Xia
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingyi Chen
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyue Xing
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Meng
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoying Song
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danfei Lou
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Wang T, Gong X, Xia C, Kong W, Geng S, Jiang H, Xiao E, Wang H, Yu Y, Li C, Yuan K. An integrated transcriptomics and network pharmacology approach to explore the mechanism of Wang-Bi tablet against SAPHO syndrome. Int J Rheum Dis 2024; 27:e15077. [PMID: 38402418 DOI: 10.1111/1756-185x.15077] [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: 09/01/2023] [Revised: 01/20/2024] [Accepted: 01/27/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND SAPHO syndrome is recognized as a rare entity with damage to skin and bones due to inflammation. Currently, the treatment for SAPHO syndrome is still a challenge in clinical practice. In this study, an integrated transcriptomics and network pharmacology approach was applied to explore the therapeutic effect and mechanism of Wang-Bi tablet (WBT) on SAPHO syndrome. METHODS The main components of WBT and their targets, as well as the targets of SAPHO syndrome, were collected from databases. Network visualization was performed using Cytoscape software. The GO and KEGG enrichment analysis was executed by David dataset. Then, the molecular mechanism of WBT improving SAPHO syndrome was validated by transcriptomics of peripheral blood neutrophils in SAPHO syndrome. Finally, the above results were validated by molecular docking. RESULTS The Network Pharmacology results showed there are 152 core targets for WBT treatment on SAPHO syndrome. RNA-seq data showed 442 differentially expressed genes (DEGs) in peripheral blood neutrophils of SAPHO patients. Intriguingly, NIK/NF-kappaB-, MyD88-dependent toll-like receptor-, and MAPK pathway were included in the enrichment results of network pharmacology and RNA-seq. Moreover, we verified that the core components of WBT have good affinity with the core targets of NIK/NF-kappaB-, MyD88-dependent toll-like receptor-, and MAPK pathway by molecular docking. CONCLUSIONS This study illustrated that the possible mechanisms of WBT against SAPHO syndrome may be related to NIK/NF-kappaB-, MyD88-dependent toll-like receptor-, and MAPK pathway, and further experiments are needed to prove these predictions.
Collapse
Affiliation(s)
- Ting Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xun Gong
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Congmin Xia
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weijia Kong
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Shaohui Geng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Haixu Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Enfan Xiao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hesong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yingcai Yu
- Department of Biochemistry, College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Chen Li
- Department of Rheumatology, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Yuan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
27
|
Weng J, Wu XF, Shao P, Liu XP, Wang CX. Medicine for chronic atrophic gastritis: a systematic review, meta- and network pharmacology analysis. Ann Med 2024; 55:2299352. [PMID: 38170849 PMCID: PMC10769149 DOI: 10.1080/07853890.2023.2299352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
PURPOSE The aim of this study is to determine the effectiveness and reliability of adding traditional Chinese medicine (TCM) in the clinical intervention and explore mechanisms of action for chronic atrophic gastritis (CAG) through meta- and network pharmacology analysis (NPAs). METHODS A predefined search strategy was used to retrieve literature from PubMed, Embase database, Cochrane Library, China National Knowledge Infrastructure (CNKI), Chinese BioMedical Literature Database (CBM), Wan Fang Data and China Science and Technology Journal Database (VIP). After applying inclusion and exclusion criteria, a total of 12 randomized controlled trials (RCTs) were included for meta-analysis to provide clinical evidence of the intervention effects. A network meta-analysis using Bayesian networks was conducted to observe the relative effects of different intervention measures and possible ranking of effects. The composition of the TCM formulation in the experimental group was analysed, and association rule mining was performed to identify hub herbal medicines. Target genes for CAG were searched in GeneCards, Online Mendelian Inheritance in Man, PharmGKB, Therapeutic Target Database and DrugBank. A regulatory network was constructed to connect the target genes with active ingredients of the hub herbal medicines. Enrichment analyses were performed using the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to examine the central targets from a comprehensive viewpoint. Protein-protein interaction networks (PPINs) were constructed to identify hub genes and conduct molecular docking with differentially expressed genes (DEGs) and corresponding active molecules. RESULTS A total of 1140 participants from 12 RCTs were included in the statistical analysis, confirming that the experimental group receiving the addition of TCM intervention had better clinical efficacy. Seven hub TCMs (Paeonia lactiflora, Atractylodes macrocephala, Pinellia ternata, Citrus reticulata, Codonopsis pilosula, Salvia miltiorrhiza and Coptis chinensis) were identified through association rule analysis of all included TCMs. Thirteen hub genes (CDKN1A, CASP3, STAT1, TP53, JUN, MAPK1, STAT3, MAPK3, MYC, HIF1A, FOS, MAPK14 and AKT1) were obtained from 90 gene PPINs. Differential gene expression analysis between the disease and normal gastric tissue identified MAPK1 and MAPK3 as the significant genes. Molecular docking analysis revealed that naringenin, luteolin and quercetin were the main active compounds with good binding activities to the two hub targets. GO analysis demonstrated the function of the targets in protein binding, while KEGG analysis indicated their involvement in important pathways related to cancer. CONCLUSIONS The results of a meta-analysis of 12 RCTs indicate that TCM intervention can improve the clinical treatment efficacy of CAG. NPAs identified seven hub TCM and 13 target genes associated with their actions, while bioinformatics analysis identified two DEGs between normal and CAG gastric tissues. Finally, molecular docking was employed to reveal the mechanism of action of the active molecules in TCM on the DEGs. These findings not only reveal the mechanisms of action of the active components of the TCMs, but also provide support for the development of new drugs, ultimately blocking the progression from chronic gastritis to gastric cancer.
Collapse
Affiliation(s)
- Jiao Weng
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiu-fang Wu
- The Second Hospital Affiliated with Shenyang Medical University, Shenyang, China
| | - Peng Shao
- The Second Hospital Affiliated with Shenyang Medical University, Shenyang, China
| | - Xing-pu Liu
- The Second Hospital Affiliated with Shenyang Medical University, Shenyang, China
| | - Cai-xia Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
28
|
Gu A, Li J, Wu JA, Li MY, Liu Y. Exploration of Dan-Shen-Yin against pancreatic cancer based on network pharmacology combined with molecular docking and experimental validation. CURRENT RESEARCH IN BIOTECHNOLOGY 2024; 7:100228. [DOI: 10.1016/j.crbiot.2024.100228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
|
29
|
Shi C, Liu X, Han SS, Tang YF, Zeng HL, Du ML, Yang Y, Jia JN, Shi Q, Hou FG. Mechanism of Preventing Recurrence of Stage II-III Colorectal Cancer Metastasis with Immuno-inflammatory and Hypoxic Microenvironment by a Four Ingredients Chinese Herbal Formula: A Bioinformatics and Network Pharmacology Analysis. Curr Pharm Des 2024; 30:2007-2026. [PMID: 38867534 DOI: 10.2174/0113816128294401240523092259] [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: 11/22/2023] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Colorectal Cancer (CRC) is one of the top three malignancies with the highest incidence and mortality. OBJECTIVE The study aimed to identify the effect of Traditional Chinese Medicine (TCM) on postoperative patients with stage II-III CRC and explore the core herb combination and its mechanism. METHODS An observational cohort study was conducted on patients diagnosed with stage II-III CRC from January 2016 to January 2021. The primary outcome was disease-free survival, which was compared between the patients who received TCM or not, and the secondary outcome was the hazard ratio. The relevance principle was used to obtain the candidate herb combinations, and the core combination was evaluated through an assessment of efficacy and representativeness. Then, biological processes and signaling pathways associated with CRC were obtained by Gene Ontology function, Kyoto Encyclopedia of Gene and Genomes pathway, and Wikipathway. Furthermore, hub genes were screened by the Kaplan-Meier estimator, and molecular docking was employed to predict the binding sites of key ingredients to hub genes. The correlation analysis was employed for the correlations between the hub genes and tumor-infiltrating immune cells and hypoxiarelated genes. Ultimately, a quantitative polymerase chain reaction was performed to verify the regulation of hub genes by their major ingredients. RESULTS A total of 707 patients were included. TCM could decrease the metastatic recurrence associated with stage II-III CRC (HR: 0.61, log-rank P < 0.05). Among those patients in the TCM group, the core combination was Baizhu → Yinchen, Chenpi, and Fuling (C combination), and its antitumor mechanism was most likely related to the regulation of BCL2L1, XIAP, and TOP1 by its key ingredients, quercetin and tangeretin. The expression of these genes was significantly correlated with both tumor-infiltrating immune cells and hypoxia- related genes. In addition, quercetin and tangeretin down-regulated the mRNA levels of BCL2L1, XIAP, and TOP1, thereby inhibiting the growth of HCT116 cells. CONCLUSION Overall, a combination of four herbs, Baizhu → Yinchen, Chenpi, and Fuling, could reduce metastatic recurrence in postoperative patients with stage II-III CRC. The mechanism may be related to the regulation of BCL2L1, XIAP, and TOP1 by its key ingredients quercetin and tangeretin.
Collapse
Affiliation(s)
- Chuan Shi
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xing Liu
- Department of Central Laboratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Su-Su Han
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yu-Fei Tang
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Hai-Lun Zeng
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Mei-Lu Du
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yi Yang
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Jia-Ning Jia
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Qi Shi
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Feng-Gang Hou
- Oncology Department III, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| |
Collapse
|
30
|
He L, Shen K, He L, Chen Y, Tang Z. The Mechanism of Plantaginis Semen in the Treatment of Diabetic Nephropathy based on Network Pharmacology and Molecular Docking Technology. Endocr Metab Immune Disord Drug Targets 2024; 24:363-379. [PMID: 37718520 DOI: 10.2174/1871530323666230915100355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 06/28/2023] [Accepted: 07/20/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the common complications of diabetes. Plantaginis Semen (PS) has a variety of therapeutic effects, however its mechanism on DN is unclear. OBJECTIVE This paper aims to find the ingredients, the key targets, and the action pathways of PS on DN from the perspective of network pharmacology. METHODS The databases of network pharmacology, such as Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP), Pharmmapper, OMIM, DrugBank, Gene- Cards, TTD, Disgenet, STRING, and Cytoscape software, were used to find the main ingredients and targets. Gene Ontology (GO) function and Kyoto Encyclopedia of Genome and Genomes (KEGG) pathway enrichment analysis were used to reveal the potential pathways of the PS on DN. The GEO database was used to find the targets of DN based on valid experimental research. The molecular docking technology was used to evaluate the combination between ingredients of PS and the targets. RESULTS A total of 9 active ingredients and 216 potential therapeutic targets were obtained for PS on DN. Hub targets were discovered by the Cytoscape software analysis. CASP3 was screened by Venn diagram by making intersection between GSE30529 and hub genes. Moreover, CASP3 was combined with one of the nine active ingredients, quercetin, by molecular docking analysis. The KEGG pathways were mainly involved in diabetic nephropathy, and were simultaneously associated with CASP3 as followed: AGE-RAGE signaling pathway in diabetic complications, apoptosis, lipid and atherosclerosis, MAPK signaling pathway, TNF signaling pathway, IL-17 signaling pathway, and p53 signaling pathway. CONCLUSION PS can have the treatment on DN through CASP3. Quercetin, as one of the nine active ingredients, can be bounded to CASP3 to inhibit apoptosis in DN. PS can also take action on DN probably through many pathways. The role of PS on DN through other pathways still needs to be further elaborated.
Collapse
Affiliation(s)
- Linlin He
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Kai Shen
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lei He
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yuqing Chen
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
31
|
Chen N, Chu Y, Su S, Zhang Q, Zhang L. Network Pharmacology and Molecular Docking Validation to Explore the Pharmacological Mechanism of Zhuling Decoction against Nephrotic Syndrome. Curr Pharm Des 2024; 30:2244-2256. [PMID: 38910482 DOI: 10.2174/0113816128305808240529115047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/06/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND In recent years, the incidence and prevalence of Nephrotic Syndrome (NS) have been increasing. Zhuling Decoction (ZLD), a classical Chinese medicine, has been clinically proven to be effective for the treatment of NS. However, its underlying mechanism and pharmacodynamic substances remain unclear. OBJECTIVE This study aimed to explore the mechanism of action and chemical components of ZLD against NS using network pharmacology and molecular docking. METHODS Traditional Chinese Medicine Systems Pharmacology (TCMSP), Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicines (BATMAN-TCM), and SwissTargetPrediction databases were used to screen the principal ingredients and the associated targets of ZLD. NS-related targets were obtained from the Online Mendelian Inheritance in Man (OMIM), GeneCards, Therapeutic Target Database (TTD), and Drugbank databases. Shared targets were derived by the intersection of ZLD- and NS-associated targets. Protein-interaction relationships were analyzed using the STRING database and Cytoscape. A visualized drug-active compound-target network of ZLD was established using Cytoscape. Analyses of gene enrichment were performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) methods by the Database for Annotation, Visualization, and Integrated Discovery (DAVID) database. Molecular docking was performed to assess the binding activity between active components and hub targets. RESULTS Polyporusterone E, cerevisterol, alisol B, and alisol B 23-acetate were the primary potential ingredients of ZLD. HMGCR, HSD11B1, NOS2, NR3C1, and NR3C2 were the hub targets of ZLD against NS. Molecular docking showed that polyporusterone E, cerevisterol, and alisol B had high binding activities with targets HMGCR, HSD11B1, and NOS2. CONCLUSION In summary, this study suggests that the main active compounds (polyporusterone E, cerevisterol, alisol B) may have important roles for ZLD acting against NS by binding to hub targets (HMGCR, HSD11B1, and NOS2) and modulating PI3K-Akt, Ras, MAPK, and HIF-1 signaling pathways.
Collapse
Affiliation(s)
- Na Chen
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yanqi Chu
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Su Su
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Qingxia Zhang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|