1
|
Peng Y, Wu S, Xu Y, Ye X, Huang X, Gao L, Lu J, Liu X. Huangqi-Danshen decoction alleviates renal fibrosis through targeting SCD1 to modulate cGAS/STING signaling. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119364. [PMID: 39832629 DOI: 10.1016/j.jep.2025.119364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/30/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Huangqi-Danshen decoction (HDD) is composed of Huangqi (Astragali Radix) and Danshen (Salviae Miltiorrhizae Radix et Rhizoma) and has been shown to alleviate renal fibrosis. However, the potential therapeutic mechanisms and effective components of HDD remain unclear. AIM OF THE STUDY Both lipid metabolism and cGAS/STING signaling play vital roles in the development and progression of renal fibrosis. However, their relationship in renal fibrosis is largely unknown. The present study aimed to investigate the antifibrotic mechanisms of HDD from the perspective of lipid remodeling and cGAS/STING signaling. MATERIALS AND METHODS In vivo, renal fibrosis was induced by feeding male C57BL/6 mice with 0.2% adenine-diet for 28 consecutive days. The treatment groups were orally administered HDD at low, medium, and high doses of 3.4 g/kg/d, 6.8 g/kg/d, and 13.6 g/kg/d simultaneously with modeling. Renal function was evaluated by the serum levels of urea nitrogen and creatinine, pathological changes of renal tissue were evaluated by Periodic acid-Schiff and Masson's trichrome staining, and renal lipid metabolites were analyzed by lipidomics. Western blotting, immunohistochemistry, and immunofluorescence were used to detect the expressions of fibrosis-related proteins, SCD1, and cGAS/STING signaling-related proteins in renal tissue. In vitro, mouse primary proximal tubular epithelial cells (PTECs) were treated with transforming growth factor-β1 (TGF-β1) or stearoyl-CoA desaturase 1 (SCD1) inhibitor A939572. Additionally, UHPLC-QE-MS analysis and TCMSP database were used to screen the effective components of HDD, and the action mechanisms of these components were verified in mouse primary PTECs. RESULTS HDD dose-dependently improved renal function, pathological injury, and fibrosis in adenine-induced chronic kidney disease (CKD) mice model. Moreover, cGAS/STING signaling was significantly activated in fibrotic kidney and was suppressed by HDD treatment. In renal lipidomics analysis, 521 and 138 differential lipids were identified in control vs. CKD and CKD vs. CKD + HDD, respectively. Of note, lipids increased in fibrotic kidneys were more saturated (fewer double bonds), whereas lipids increased by HDD were less saturated (more double bonds). Further, SCD1 expression was significantly down-regulated in fibrotic kidney and could be restored by HDD treatment. The expression of SCD1 was also down-regulated in Ju CKD patients' dataset and TGF-β1-induced fibrogenic responses in mouse primary PTECs. Mechanistically, specific inhibition of SCD1 expression could activate cGAS/STING signaling in primary PTECs. In addition, three components of HDD (isoimperatorin, baicalin, and miltirone) were screened out. Furthermore, administration of these three components, especially isoimperatorin and miltirone, counteracted the activation of cGAS/STING signaling induced by SCD1 pharmacological inhibition. CONCLUSION HDD could alleviate renal fibrosis, which may be related to the regulation of cGAS/STING signaling through targeting SCD1.
Collapse
Affiliation(s)
- Yu Peng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China; The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| | - Shanshan Wu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China; The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| | - Youcai Xu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China; The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| | - Xiaoqin Ye
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China; The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| | - Xi Huang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China; The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| | - Liwen Gao
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China; Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, 528000, China.
| | - Jiandong Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| | - Xinhui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| |
Collapse
|
2
|
Su W, Xie X, Zhao J, Fan Q, Dong N, Li Q, Du Y, Wu S. Comparative efficacy of Chinese patent medicines in patients with carotid atherosclerotic plaque: a Bayesian network meta- analysis. Chin Med 2023; 18:152. [PMID: 37986011 PMCID: PMC10662928 DOI: 10.1186/s13020-023-00850-5] [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: 07/11/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Traditional Chinese patent medicines (TCPMs) have been widely used to treat carotid atherosclerotic plaque (CAP) in China. However, systematic evaluation of the clinical efficacy of TCPMs for CAP is still unknown, and the comparative efficacy of different TCPMs is unclear. OBJECTIVES This study aims to compare and rank the effectiveness and safety of different TCPMs in treating CAP using a Bayesian network meta- analysis (NMA). METHODS This NMA was performed according to the Preferred Reporting Items for Systematic Reviews and Meta- Analyses (PRISMA) Extension Statement. Eight databases were searched from their inception to August 2023 for randomized controlled trials (RCTs). The articles regarding eligibility and extracted data were screened independently by two authors. The Cochrane Risk of Bias tool was used to evaluate quality and bias. The change of carotid artery intimal- medial thickness (IMT), carotid maximal plaque area, carotid atherosclerotic plaque Course score, serum lipid levels, CRP, and adverse events rate (AER) were used as outcomes. Data from each RCTs were first pooled using random- effect pairwise meta- analyses and illustrated as odds ratios (ORs) or standardized mean differences (SMDs) with 95% confidence interval (CI). NMAs were performed using Stata17.0 software and the GeMTC package of R software to evaluate the comparative effectiveness of TCPMs, and displayed as ORs or SMDs with 95% CI. A Bayesian hierarchical random- effects model was used to conduct NMAs using the Markov Chain Monte Carlo algorithm. The GRADE partially contextualised framework was applied for NMA result interpretation. RESULTS NMA included 27 RCT trials with 4131 patients and nine types of TCPMs. Pairwise meta- analyses indicated that Conventional Western medicine (CWM) + TCPM was superior to CWM in reducing the IMT (SMD: - 1.26; 95% CI - 1.59 to - 0.93), the carotid maximal plaque area (SMD - 1.27; 95% CI - 1.71, - 0.82) and the carotid atherosclerotic plaque Course score (SMD - 0.72; 95% CI 95% CI - 1.20, - 0.25). NMAs demonstrated that CWM + Jiangzhiling pill (JZL) with SUCRA 70.6% exhibited the highest effective intervention for reducing IMT. CWM + SXBX (Shexiang baoxin pill) was superior to other TCPMs in reducing the carotid maximal plaque area (83.0%), the atherosclerotic plaque Course score (92.5%), TC (95.6%) and LDL (92.6%) levels. CWM + NXT (Naoxintong capsule), CWM + XS (Xiaoshuang granules/enteric capsule), and CWM + ZBT (Zhibitai) were superior to other CPMs in improving TG (90.1%), HDL (86.1%), and CRP (92.6%), respectively. No serious adverse events were reported. CONCLUSIONS For CAP patients, CWM + XSBX was among the most effective in reducing carotid maximal plaque area, atherosclerotic plaque Course score, TC and LDL levels, and CWM + JZL was the most effective in reducing IMT. Overall, CWM + XSBX may be considered an effective intervention for the treatment of CAP. This study provides reference and evidence for the clinical optimization of TCPM selection in CAP treatment. More adequately powered, well- designed clinical trials to increase the quality of the available evidence are still needed in the future due to several limitations.
Collapse
Affiliation(s)
- Wenquan Su
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Xiaolong Xie
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jiping Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Qinhua Fan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Naijia Dong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Qingxiao Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yawei Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Shengxian Wu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
3
|
Sun Y, Cai J, Ding S, Bao S. Network Pharmacology Was Used to Predict the Active Components and Prospective Targets of Paeoniae Radix Alba for Treatment in Endometriosis. Reprod Sci 2023; 30:1103-1117. [PMID: 36258089 DOI: 10.1007/s43032-022-01102-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
Abstract
Endometriosis is one of the most common benign gynecologic diseases. Paeoniae Radix Alba (PRA) has been utilized to treat endometriosis. We wished to identify potential targets for PRA in the treatment of endometriosis, as well as to provide a groundwork for future studies into its pharmacological mechanism of action. Network pharmacology was employed to conduct investigations on PRA. Target proteins were chosen from the components of PRA for endometriosis treatment. A protein-protein interaction (PPI) was established using overlapping genes. Analyses of enrichment of function and signaling pathways were undertaken using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes databases to select "hub genes." Finally, the feasibility of analysis based on network pharmacology was determined using real-time reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting. We demonstrated that PRA has 25 bioactive components and 167 putative targets that are therapeutically important. The anti-inflammatory and immune-boosting actions of tumor necrosis factor, albumin, signal transducer and activator of transcription (STAT)3, mitogen-activated protein kinase, Jun, interleukin (IL)-1B, prostaglandin-endoperoxide synthase 2, matrix metalloproteinase-9, vascular endothelial growth factor A, and IL-6 were identified as prospective targets. Seven major compounds in PRA and related to the STAT3 pathway could bind spontaneously to it. RT-qPCR and western blotting showed that expression of STAT3 and phospho-STAT3 was reduced significantly after PRA intervention. Hence, analyses of the active components of traditional Chinese medicine formulations through network pharmacology may open up new ideas for the treatment of diseases.
Collapse
Affiliation(s)
- Yuting Sun
- Department of Gynecology and Obstetrics of Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Junhong Cai
- Medical Laboratory Center, Hainan General Hospital, Hainan Medical University, Haikou, 570102, China
| | - Shun Ding
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital, Hainan Medical University, Haikou, 570102, China
| | - Shan Bao
- Department of Gynecology and Obstetrics of Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China.
| |
Collapse
|
4
|
Li C, Chi C, Li W, Li Z, Wang X, Wang M, Zhang L, Lu J, Liu R. An integrated approach for identifying the efficacy and potential mechanisms of TCM against atherosclerosis-Wu-Zhu-Yu decoction as a case study. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115436. [PMID: 35667584 DOI: 10.1016/j.jep.2022.115436] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis (AS) is a chronic disease that is associated with high morbidity. However, therapeutic approaches are limited. Wu-Zhu-Yu decoction (WZYD) is a well-known traditional Chinese medicine prescription that is traditionally used to treat headaches and vomiting. Modern studies have demonstrated the cardiotonic effects of WZYD. However, whether WZYD can alleviate AS and its underlying mechanisms remain unclear. AIM OF THE STUDY This study aims to investigate the antiatherosclerotic efficacy of WZYD and illustrate its potential mechanisms using an integrated approach combining in vivo and in vitro assessments, including metabolomics, network pharmacology, cell experiments, and molecular docking analyses. MATERIALS AND METHODS In this work, an atherosclerotic mouse model was established by administering a high-fat diet to apolipoprotein-E deficient (ApoE-/-) mice for twelve weeks. Meanwhile, the mice were intragastrically administered WZYD at different dosages. Efficacy evaluation was performed through biochemical and histopathological assessments. The potential active constituents, metabolites, and targets of WZYD in atherosclerosis were predicted by metabolomics combined with network pharmacology analysis, the constituents and targets were further assessed through cell experiments and molecular docking analysis. RESULTS WZYD decreased the lipid levels in serum, reduced the areas of aortic lesions, and attenuated intimal thickening, which had antiatherosclerotic effects in ApoE-/- mice. Metabolomics and network pharmacology approach revealed that the ten constituents (6-shogaol, evodiamine, isorhamnetin, quercetin, beta-carotene, 8-gingerol, kaempferol, 6-paradol, 10-gingerol, and 6-gingerol) of WZYD affected 24 metabolites by acting on the candidate targets, thus resulting in changes in five metabolic pathways (sphingolipid metabolism; glycine, serine and threonine metabolism; arachidonic acid metabolism; tryptophan metabolism; and fatty acid biosynthesis pathway). Cell experiments indicated that the ten key compounds showed antiproliferative effects on the vascular smooth muscle cell. Moreover, the key compounds exhibited direct interactions with the key targets, as assessed by molecular docking analysis. CONCLUSION This study revealed that WZYD exerted therapeutic effects on atherosclerosis, and the potential mechanisms were elucidated. Furthermore, it offered a powerful integrated strategy for studying the efficacy of traditional Chinese medicine and exploring its active components and possible mechanisms.
Collapse
Affiliation(s)
- Caihong Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Chenglin Chi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Wenjing Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Zongchao Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Xinlin Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Minjun Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Leiming Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Rongxia Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| |
Collapse
|
5
|
Chen S, Wu X, Li T, Li Y, Wang B, Cheng W, Teng Y, Yang J, Meng H, Wang L, Lu Z, Jiang Y, Wang Y, Zhao M. Atheroprotective Effects and Mechanisms of Postmarketing Chinese Patent Formulas in Atherosclerosis Models: A Systematic Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:4010607. [PMID: 34873408 PMCID: PMC8643251 DOI: 10.1155/2021/4010607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Some postmarketing Chinese patent formulas have been widely used to treat atherosclerosis (AS) and play critical roles in Chinese healthcare. However, the usage of these herbs is yet controversial due to unclear effects and lack of understanding of the mechanism of action. With the modernization of traditional Chinese formulas, we are to elucidate the atheroprotective properties of these remedies from successful postmarketing experiments in vivo. METHODS In this systematic review, we critically searched the databases, applied stringent criteria, assessed the methodological quality, and examined the current evidence in vivo. RESULTS Consequently, 60 studies were included in the present qualitative synthesis. Data on models, high-fat diet, intervention time, outcome measures, efficacy, and mechanisms were collected. Finally, 23 formulas that could alleviate AS were correlated to the amelioration of plaques, improvement of plaque stability, modification of lipid level and lipid metabolism, and the effects of anti-inflammation and antioxidant stress with multiple components and targets. However, the methodological quality was low and incomplete among the included literature. CONCLUSIONS Thus, taken together, the studies on postmarketing Chinese patent formulas would provide a novel approach to improve the treatment of AS, and rigorously designed studies would provide high-quality evidence.
Collapse
Affiliation(s)
- Shiqi Chen
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaoxiao Wu
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Tong Li
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Li
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Baofu Wang
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Weiting Cheng
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yu Teng
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jingjing Yang
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Hui Meng
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Lei Wang
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Ziwen Lu
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yangyang Jiang
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yahong Wang
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
6
|
Bai Y, Zhang Y, Li S, Zhang W, Wang X, He B, Ju W. Integrated Network Pharmacology Analysis and Experimental Validation to Investigate the Mechanism of Zhi-Zi-Hou-Po Decoction in Depression. Front Pharmacol 2021; 12:711303. [PMID: 34690756 PMCID: PMC8531485 DOI: 10.3389/fphar.2021.711303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/07/2021] [Indexed: 12/16/2022] Open
Abstract
Zhi-Zi-Hou-Po Decoction (ZZHPD) is a well-known traditional Chinese medicine (TCM) that has been widely used in depression. However, the antidepressant mechanism of ZZHPD has not yet been fully elucidated. The purpose of this study was to explore the pharmacological mechanisms of ZZHPD acting on depression by combining ultra flow liquid chromatography with quadrupole time-of-flight mass spectrometry (UFLC-Q-TOF/MS) and network pharmacology strategy. The chemical components of ZZHPD were identified using UFLC-Q-TOF/MS, while the potential drug targets and depression-related targets were collected from databases on the basis of the identified compounds of ZZHPD. Protein-protein interaction (PPI) network, gene ontology (GO), and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses were used to unravel potential antidepressant mechanisms. The predicted antidepressant targets from the pharmacology-based analysis were further verified in vivo. As a result, a total of 31 chemical compounds were identified by UFLC-Q-TOF/MS; 514 promising drug targets were mined by using the Swiss Target Prediction; and 527 depression-related target genes were pinpointed by the GeneCards and OMIM databases. STRING database and Cytoscape's topological analysis revealed 80 potential targets related to the antidepressant mechanism of ZZHPD. The KEGG pathway analysis revealed that the antidepressant targets of ZZHPD were mainly involved in dopaminergic synapse, serotonin synapse, cAMP, and mTOR signaling pathways. Furthermore, based on the animal model of depression induced by chronic corticosterone, the regulatory effects of ZZHPD on the expression of MAOA, MAOB, DRD2, CREBBP, AKT1, MAPK1, HTR1A, and GRIN2B mRNA levels as well as the cAMP signaling pathway and monoaminergic metabolism were experimentally verified in rats. Our study revealed that ZZHPD is expounded to target various genes and pathways to perform its antidepressant effect.
Collapse
Affiliation(s)
- Yongtao Bai
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,Phase I Clinical Research Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yingchun Zhang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Shuolei Li
- Phase I Clinical Research Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenzhou Zhang
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xinhui Wang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Baoxia He
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China.,Phase I Clinical Research Center, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenzheng Ju
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
7
|
Chen X, Zuo J, Hu T, Shi X, Zhu Y, Wu H, Xia Y, Shi W, Wei W. Exploration of the Effect and Mechanism of Fructus Lycii, Rehmanniae Radix Praeparata, and Paeonia lactiflora in the Treatment of AMD Based on Network Pharmacology and in vitro Experimental Verification. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2831-2842. [PMID: 34234414 PMCID: PMC8254409 DOI: 10.2147/dddt.s310481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/25/2021] [Indexed: 12/24/2022]
Abstract
Purpose The aim of this study was to observe the mechanism of Fructus Lycii (FL), Rehmanniae Radix Praeparata (RRP) and Paeonia lactiflora (PL) in treating age-related macular degeneration (AMD) based on network pharmacology and biological experiments. Methods Bioactive compounds, potential targets of FL, RRP and PL, and genes related to AMD, were acquired from public databases. Functional and pathway enrichment analyses of the core targets were conducted by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Subsequently, the finding was further verified with cell experiments. The MTT assay and flow cytometric analysis were used to assess cell viability and apoptosis. The production of reactive oxygen species (ROS) was analyzed by DCFH-DA staining; the activity of antioxidant enzymes was chemically measured with assay kits. The expression of key proteins was evaluated by Western blot analysis. Results Fifty-nine active compounds, 182 potential targets, and 2536 AMD-related human genes were identified. A total of 103 key targets of the three herbs on AMD were identified by protein-protein interaction (PPI) analysis. The abovementioned targets were correlated with nuclear receptor activity, oxidative stress, and apoptosis pathways according to the GO and KEGG analyses. MTT assay and flow cytometry demonstrated that pretreatment of ARPE-19 cells with the three herbs significantly increased cell viability and decreased apoptosis induced by H2O2. The three herbs might reduce the intracellular ROS levels and increase the SOD and CAT activities after H2O2. Furthermore, the three herbs significantly inhibited oxidative stress via increasing the expression of Nrf2, HO-1 and NQO1. Conclusion The combined results of network pharmacology and validation experiments showed that FL, RRP and PL reduce oxidative stress and apoptosis in RPE cells to exert its effect in the treatment of AMD.
Collapse
Affiliation(s)
- Xi Chen
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Jing Zuo
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Tianming Hu
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xiaoqing Shi
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Yujie Zhu
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Hao Wu
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Ying Xia
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Wei Shi
- Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Wei Wei
- First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Ophthalmology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| |
Collapse
|
8
|
Huang Q, Liu R, Liu J, Huang Q, Liu S, Jiang Y. Integrated Network Pharmacology Analysis and Experimental Validation to Reveal the Mechanism of Anti-Insulin Resistance Effects of Moringa oleifera Seeds. Drug Des Devel Ther 2020; 14:4069-4084. [PMID: 33116398 PMCID: PMC7539042 DOI: 10.2147/dddt.s265198] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Insulin resistance (IR) is one of the factors that results in metabolic syndrome, type 2 diabetes mellitus and different aspects of cardiovascular diseases. Moringa oleifera seeds (MOS), traditionally used as an antidiabetic food and traditional medicine in tropical Asia and Africa, have exhibited potential effects in improving IR. To systematically explore the pharmacological mechanism of the anti-IR effects of MOS, we adopted a network pharmacology approach at the molecular level. METHODS By incorporating compound screening and target prediction, a feasible compound-target-pathway network pharmacology model was established to systematically predict the potential active components and mechanisms of the anti-IR effects of MOS. Biological methods were then used to verify the results of the network pharmacology analysis. RESULTS Our comprehensive systematic approach successfully identified 32 bioactive compounds in MOS and 44 potential targets of these compounds related to IR, as well as 37 potential pathways related to IR. Moreover, the network pharmacology analysis revealed that glycosidic isothiocyanates and glycosidic benzylamines were the major active components that improved IR by acting on key targets, such as SRC, PTPN1, and CASP3, which were involved in inflammatory responses and insulin-related pathways. Further biological research demonstrated that the anti-IR effects of MOS were mediated by increasing glucose uptake and modulating the expression of SRC and PTPN1. CONCLUSION Our study successfully predicts the active ingredients and potential targets of MOS for improving IR and helps to illustrate mechanism of action at a systemic level. This study not only provides new insights into the chemical basis and pharmacology of MOS but also demonstrates a feasible method for discovering potential drugs from traditional medicines.
Collapse
Affiliation(s)
- Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Hospital Pharmacy, Central South University, Changsha410008, People’s Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| | - Rong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Hospital Pharmacy, Central South University, Changsha410008, People’s Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| | - Jing Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Hospital Pharmacy, Central South University, Changsha410008, People’s Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| | - Qi Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Hospital Pharmacy, Central South University, Changsha410008, People’s Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Hospital Pharmacy, Central South University, Changsha410008, People’s Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| | - Yueping Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
- Institute of Hospital Pharmacy, Central South University, Changsha410008, People’s Republic of China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| |
Collapse
|
9
|
Zeng Q, Li L, Siu W, Jin Y, Cao M, Li W, Chen J, Cong W, Ma M, Chen K, Wu Z. A combined molecular biology and network pharmacology approach to investigate the multi-target mechanisms of Chaihu Shugan San on Alzheimer's disease. Biomed Pharmacother 2019; 120:109370. [PMID: 31563815 DOI: 10.1016/j.biopha.2019.109370] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/12/2019] [Accepted: 08/21/2019] [Indexed: 12/18/2022] Open
Abstract
Chaihu Shugan San (CSS) is a well-known herbal formula used to nourish liver and blood, promote blood circulation and Qi flow in Traditional Chinese Medicine. Modern pharmacological studies and clinical uses showed that CSS could ameliorate cognitive dysfunction of Alzheimer's disease (AD). The present study aimed to elucidate the multi-target mechanisms of CSS on AD using network pharmacology analysis and verify its effect by biological experiments. Firstly, a total of 152 active compounds in CSS, 520 predicted biological targets and 160 AD-related targets were identified. Subsequently, the networks including herb-compound-target network, AD-target network, and CSS potential target-AD target network were constructed. 60 key targets highly responsible for the beneficial effect of CSS on AD were identified by central network topological analysis. They were significantly characterized as nuclear or cytoplasmic proteins with molecular function of protein binding. They were also enriched in various biological processes through PI3K-Akt signaling pathway, MAPK signaling pathway and HIF signaling pathway by GO function and KEGG pathway enrichment analysis. Pretreatment with CSS ameliorated Aβ-induced neural cell death and reduced the number of apoptotic cells in differentiated PC12 cells. Moreover, increased phosphorylation of Akt accompanied with decreased Bax expression was found after CSS pretreatment, suggesting that Akt signaling pathway was involved in the protective effect of CSS against neural cells death. The present study systematically revealed the multi-target mechanisms of CSS on AD using network pharmacology approach, as well as validated the protective effect of CSS against Aβ-induced neural cells death through Akt signaling pathway. It provided indications for further mechanistic studies and also for the development of CSS as a potential treatment for AD patients.
Collapse
Affiliation(s)
- Qiang Zeng
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Longfei Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wingsum Siu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Jin
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Meiqun Cao
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China
| | - Weifeng Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jian Chen
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Weihong Cong
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Min Ma
- Integrated Chinese and Western Medicine Postdoctoral Research Station, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Keji Chen
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Zhengzhi Wu
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China; Shenzhen Institute of Geriatrics, Shenzhen 518020, China.
| |
Collapse
|
10
|
Pruvot M, Khammavong K, Milavong P, Philavong C, Reinharz D, Mayxay M, Rattanavong S, Horwood P, Dussart P, Douangngeun B, Theppangna W, Fine AE, Olson SH, Robinson M, Newton P. Toward a quantification of risks at the nexus of conservation and health: The case of bushmeat markets in Lao PDR. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 676:732-745. [PMID: 31054417 PMCID: PMC7112076 DOI: 10.1016/j.scitotenv.2019.04.266] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 04/14/2023]
Abstract
Trade of bushmeat and other wildlife for human consumption presents a unique set of challenges to policy-makers who are confronted with multiple trade-offs between conservation, food security, food safety, culture and tradition. In the face of these complex issues, risk assessments supported by quantitative information would facilitate evidence-based decision making. We propose a conceptual model for disease transmission risk analysis, inclusive of these multiple other facets. To quantify several processes included in this conceptual model we conducted questionnaire surveys with wildlife consumers and vendors in semi-urban centers in Lao People's Democratic Republic (Lao PDR, Laos) and direct observations of consumer behaviors. Direct observation of market stalls indicated an estimated average of 10 kg bushmeat biomass per stall per hour. The socio-demographic data suggested that consumption of bushmeat in urban areas was not for subsistence but rather driven by dietary preference and tradition. Consumer behavioral observations indicated that each animal receives an average of 7 contacts per hour. We provide other key parameters to estimate the risk of disease transmission from bushmeat consumption and illustrate their use in assessing the total public health and socio-economic impact of bushmeat consumption. Pursuing integrative approaches to the study of bushmeat consumption is essential to develop effective and balanced policies that support conservation, public health, and rural development goals.
Collapse
Affiliation(s)
- Mathieu Pruvot
- Wildlife Conservation Society, Wildlife Health Program, 2300 Southern Blvd, Bronx, NY 10460, USA.
| | - Kongsy Khammavong
- Wildlife Conservation Society, Lao PDR Program, Vientiane, Lao Democratic People's Republic
| | - Phonesavanh Milavong
- Wildlife Conservation Society, Lao PDR Program, Vientiane, Lao Democratic People's Republic
| | | | - Daniel Reinharz
- Institut de la Francophonie pour la Médecine Tropicale, Vientiane, Lao Democratic People's Republic; Département de Médecine sociale et préventive, Université Laval, Québec, Canada
| | - Mayfong Mayxay
- Lao-Oxford-Mahosot Hospital Wellcome Trust Research Unit, Vientiane, Lao Democratic People's Republic; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sayapeth Rattanavong
- Lao-Oxford-Mahosot Hospital Wellcome Trust Research Unit, Vientiane, Lao Democratic People's Republic
| | - Paul Horwood
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia; Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | | | - Bounlom Douangngeun
- National Animal Health Laboratory, Department of Livestock and Fisheries, Ministry of Agriculture, Ban Khunta, Vientiane, Lao Democratic People's Republic
| | - Watthana Theppangna
- National Animal Health Laboratory, Department of Livestock and Fisheries, Ministry of Agriculture, Ban Khunta, Vientiane, Lao Democratic People's Republic
| | - Amanda E Fine
- Wildlife Conservation Society, Wildlife Health Program, 2300 Southern Blvd, Bronx, NY 10460, USA
| | - Sarah H Olson
- Wildlife Conservation Society, Wildlife Health Program, 2300 Southern Blvd, Bronx, NY 10460, USA
| | - Matthew Robinson
- Lao-Oxford-Mahosot Hospital Wellcome Trust Research Unit, Vientiane, Lao Democratic People's Republic; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Paul Newton
- Lao-Oxford-Mahosot Hospital Wellcome Trust Research Unit, Vientiane, Lao Democratic People's Republic; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|