1
|
Wang SX, Chen BS, Zhang ZJ, Zhu SR, Wang XL, Liu GQ. Isolation, structural elucidation and biosynthetic pathway of bioactive prenyl quinone compounds from Panus lecomtei based on untargeted metabolomics combined with molecular networking. Food Chem 2025; 463:141275. [PMID: 39298853 DOI: 10.1016/j.foodchem.2024.141275] [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: 03/09/2023] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Panus lecomtei is a relatively unfamiliar and undeveloped mushroom. This study generated ethyl acetate extracts of P. lecomtei intracellular (I), extracellular (E) and total fermentation broth (T). Both E and T extracts demonstrated antioxidant and antibacterial activities at 100 to 200 μg/mL. The composition differences of metabolites of these extracts were further studied based on comparative metabolomics by LS/MS and molecular network analysis. The results revealed that there were over 2000 significantly distinct metabolites among the three extracts, with abundant prenyl quinone compounds. Furthermore, the molecular network clarified the conversion relationship of P. lecomtei metabolites. Seven known prenyl quinone derivatives (1-7) were isolated from the E extract. Among them, compound 3 displayed excellent antioxidant activity and modest antibacterial activity. Compound 5 was discovered in fungi for the first time. Finally, a potential biosynthetic route for prenyl quinone in P. lecomtei was suggested.
Collapse
Affiliation(s)
- Si-Xian Wang
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China
| | - Bao-Song Chen
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medicine School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zi-Juan Zhang
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China
| | - Shi-Rong Zhu
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China
| | - Xiao-Ling Wang
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China.
| | - Gao-Qiang Liu
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China.
| |
Collapse
|
2
|
Zhang L, Zhu W, Zhang C. Exploring the Effect and Mechanism of DaYuan Yin Against Acute Lung Injury by Network Pharmacology, Molecular Docking, and Experimental Validation. Drug Des Devel Ther 2024; 18:5541-5561. [PMID: 39650849 PMCID: PMC11625185 DOI: 10.2147/dddt.s491521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/26/2024] [Indexed: 12/11/2024] Open
Abstract
Background DaYuan Yin (DYY), a traditional Chinese medicine for lung diseases, requires further study to understand how it improves acute lung injury (ALI). This study seeks to elucidate the material basis and molecular mechanisms underlying the treatment of ALI with DYY through network pharmacology, molecular docking, and experimental validation. Methods DYY's active components and targets were identified using TCMSP and UHPLC-MS/MS, and a herb-component-target network was created with Cytoscape 3.7.2. ALI target genes were sourced from GeneCards, DisGeNET, and DrugBank. A PPI network was built, with core targets analyzed through GO and KEGG enrichment via Metscape. The therapeutic effects and mechanisms of DYY on LPS-induced ALI in rats were explored, and molecular docking evaluated the interactions between Nrf2, HO-1, TLR4, and the components. Results The study identified 95 active compounds, 234 therapeutic targets, and 2529 ALI-related genes, with 111 shared targets between DYY and ALI. KEGG analysis indicates that the PI3K-AKT, MAPK, and oxidative stress pathways are associated with DYY's anti-ALI effects. Network pharmacology and UHPLC-MS/MS analysis revealed active ingredients like quercetin, Magnolol, and Wogonin. Compared with the model group, DYY reduced the lung dry-wet ratio (W/D) of ALI rats from (5.31 ± 0.51) to (4.47 ± 0.73)(P < 0.05). Meanwhile, the contents of IL-6 and TNF-α in bronchoalveolar lavage fluid (BALF) and MDA, NO and ROS in lung tissue were also significantly decreased. Notably, DYY enhances UCP2 mRNA expression, boosts Nrf2 and HO-1 expression, and inhibits TLR4-mediated pro-inflammatory mediators. Molecular docking analysis showed that the main components of DYY had strong binding ability with HO-1. Conclusion DYY can alleviate inflammation, oxidative stress, and ALI-related changes by targeting the Nrf2/HO-1 mediated TLR4 pathway, providing insights for developing effective ALI treatments.
Collapse
Affiliation(s)
- Lei Zhang
- Pharmacy Department, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, People’s Republic of China
| | - Wei Zhu
- Pharmacy Department, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, People’s Republic of China
| | - Cong Zhang
- Pharmacy Department, Kunshan Rehabilitation Hospital, Kunshan, Jiangsu, People’s Republic of China
| |
Collapse
|
3
|
Cha MH, Choi HJ, Ma JY. Lysophosphatidylcholines Promote Influenza Virus Reproduction through the MAPK/JNK Pathway in PMA-Differentiated THP-1 Macrophages. Int J Mol Sci 2024; 25:6538. [PMID: 38928244 PMCID: PMC11204278 DOI: 10.3390/ijms25126538] [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: 05/13/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Obesity and metabolic syndrome alter serum lipid profiles. They also increase vulnerability to viral infections and worsen the survival rate and symptoms after infection. How serum lipids affect influenza virus proliferation is unclear. Here, we investigated the effects of lysophosphatidylcholines on influenza A virus (IAV) proliferation. IAV particles in the culture medium were titrated using extraction-free quantitative PCR, and viral RNA and protein levels were assessed using real-time PCR and Western blot, respectively. RNA sequencing data were analyzed using PCA and heatmap analysis, and pathway analysis was performed using the KEGG mapper and PathIN tools. Statistical analysis was conducted using SPSS21.0. LPC treatment of THP-1 cells significantly increased IAV proliferation and IAV RNA and protein levels, and saturated LPC was more active in IAV RNA expression than unsaturated LPC was. The functional analysis of genes affected by LPCs showed that the expression of genes involved in IAV signaling, such as suppressor of cytokine signaling 3 (SOCS3), phosphoinositide-3-kinase regulatory subunit 3 (PI3K) and AKT serine/threonine kinase 3 (AKT3), Toll-like receptor 7 (TKR7), and interferon gamma receptor 1 (IFNGR1), was changed by LPC. Altered influenza A pathways were linked with MAPK and PI3K/AKT signaling. Treatment with inhibitors of MAPK or PI3K attenuated viral gene expression changes induced by LPCs. The present study shows that LPCs stimulated virus reproduction by modifying the cellular environment to one in which viruses proliferated better. This was mediated by the MAPK, JNK, and PI3K/AKT pathways. Further animal studies are needed to confirm the link between LPCs from serum or the respiratory system and IAV proliferation.
Collapse
Affiliation(s)
- Min-Ho Cha
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea;
| | | | - Jin-Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea;
| |
Collapse
|
4
|
Wu H, Lou T, Pan M, Wei Z, Yang X, Liu L, Feng M, Shi L, Qu B, Cong S, Chen K, Yang H, Liu J, Li Y, Jia Z, Xiao H. Chaihu Guizhi Ganjiang Decoction attenuates nonalcoholic steatohepatitis by enhancing intestinal barrier integrity and ameliorating PPARα mediated lipotoxicity. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117841. [PMID: 38310988 DOI: 10.1016/j.jep.2024.117841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/28/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is a prominent cause of liver-related death that poses a threat to global health and is characterized by severe hepatic steatosis, lobular inflammation, and ballooning degeneration. To date, no Food and Drug Administration-approved medicine is commercially available. The Chaihu Guizhi Ganjiang Decoction (CGGD) shows potential curative effects on regulation of blood lipids and blood glucose, mitigation of organism inflammation, and amelioration of hepatic function. However, the overall regulatory mechanisms underlying its effects on NASH remain unclear. PURPOSE This study aimed to investigate the efficiency of CGGD on methionine- and choline-deficient (MCD)-induced NASH and unravel its underlying mechanisms. METHODS A NASH model of SD rats was established using an MCD diet for 8 weeks, and the efficacy of CGGD was evaluated based on hepatic lipid accumulation, inflammatory response, and fibrosis. The effects of CGGD on the intestinal barrier, metabolic profile, and differentially expressed genes (DEGs) profile were analyzed by integrating gut microbiota, metabolomics, and transcriptome sequencing to elucidate its mechanisms of action. RESULTS In MCD-induced NASH rats, pathological staining demonstrated that CGGD alleviated lipid accumulation, inflammatory cell infiltration, and fibrosis in the hepatic tissue. After CGGD administration, liver index, liver weight, serum alanine aminotransferase (ALT), and aspartate aminotransferase (AST) contents, liver triglycerides (TG), and free fatty acids (FFAs) were decreased, meanwhile, it down-regulated the level of proinflammatory mediators (TNF-α, IL-6, IL-1β, MCP-1), and up-regulated the level of anti-inflammatory factors (IL-4, IL-10), and the expression of liver fibrosis markers TGFβ, Acta2, Col1a1 and Col1a2 were weakened. Mechanistically, CGGD treatment altered the diversity of intestinal flora, as evidenced by the depletion of Allobaculum, Blautia, norank_f_Erysipelotrichaceae, and enrichment of the probiotic genera Roseburia, Lactobacillus, Lachnoclostridium, etc. The colonic histopathological results indicated that the gut barrier damage recovered in the CGGD treatment group, and the expression levels of colonic short-chain fatty acids (SCFAs)-specific receptors FFAR2, FFAR3, and tight junction (TJs) proteins ZO-1, Occludin, Claudin-1 were increased compared with those in the model group. Further metabolomic and transcriptomic analyses suggested that CGGD mitigated the lipotoxicity caused by glycerophospholipid and eicosanoid metabolism disorders by decreasing the levels of PLA2G4A, LPCAT1, COX2, and LOX5. In addition, CGGD could activate the inhibitory lipotoxic transcription factor PPARα, regulate the proteins of FABP1, APOC2, APOA2, and LPL to promote fatty acid catabolism, and suppress the TLR4/MyD88/NFκB pathway to attenuate NASH. CONCLUSION Our study demonstrated that CGGD improved steatosis, inflammation, and fibrosis on NASH through enhancing intestinal barrier integrity and alleviating PPARα mediated lipotoxicity, which makes it an attractive candidate for potential new strategies for NASH prevention and treatment.
Collapse
Affiliation(s)
- Hao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tianyu Lou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingxia Pan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zuying Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoqin Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lirong Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Menghan Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lixia Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Biqiong Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shiyu Cong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Kui Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Haolan Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jie Liu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yueting Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhixin Jia
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hongbin Xiao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
5
|
Guo H, Cui BD, Gong M, Li QX, Zhang LX, Chen JL, Chi J, Zhu LL, Xu EP, Wang ZM, Dai LP. An ethanolic extract of Arctium lappa L. leaves ameliorates experimental atherosclerosis by modulating lipid metabolism and inflammatory responses through PI3K/Akt and NF-κB singnaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117768. [PMID: 38253275 DOI: 10.1016/j.jep.2024.117768] [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: 10/24/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis (AS), a lipid-induced inflammatory condition of the arteries, is a primary contributor to atherosclerotic cardiovascular diseases including stroke. Arctium lappa L. leaf (ALL), an edible and medicinal herb in China, has been documented and commonly used for treating stroke since the ancient times. However, the elucidations on its anti-AS effects and molecular mechanism remain insufficient. AIM OF THE STUDY To investigate the AS-ameliorating effects and the underlying mechanism of action of an ethanolic extract of leaves of Arctium lappa L. (ALLE). MATERIALS AND METHODS ALLE was reflux extracted using with 70% ethanol. An HPLC method was established to monitor the quality of ALLE. High fat diet (HFD) and vitamin D3-induced experimental AS in rats were used to determine the in vivo effects; and oxidized low-density lipoprotein-induced RAW264.7 macrophage foam cells were used for in vitro assays. Simvatatin was used as positive control. Biochemical assays were implemented to ascertain the secretions of lipids and pro-inflammatory mediators. Haematoxylin-eosin (H&E) and Oil red O stains were employed to assess histopathological alterations and lipid accumulation conditions, respectively. CCK-8 assays were used to measure cytotoxicity. Immunoblotting assay was conducted to measure protein levels. RESULTS ALLE treatment significantly ameliorated lipid deposition and histological abnormalities of aortas and livers in AS rats; improved the imbalances of serum lipids including total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C); notably attenuated serum concentrations of inflammation-associated cytokines/molecules including TNF-α, IL-6, IL-1β, VCAM-1, ICAM-1and MMP-9. Mechanistic studies demonstrated that ALLE suppressed the phosphorylation/activation of PI3K, Akt and NF-κB in AS rat aortas and in cultured foam cells. Additionally, the PI3K agonist 740Y-P notably reversed the in vitro inhibitory effects of ALLE on lipid deposition, productions of TC, TNF-α and IL-6, and protein levels of molecules of PI3K/Akt and NF-κB singnaling pathways. CONCLUSIONS ALLE ameliorates HFD- and vitamin D3-induced experimental AS by modulating lipid metabolism and inflammatory responses, and underlying mechanisms involves inhibition of the PI3K/Akt and NF-κB singnaling pathways. The findings of this study provide scientific justifications for the traditional application of ALL in managing atherosclerotic diseases.
Collapse
Affiliation(s)
- Hui Guo
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Bing-di Cui
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Man Gong
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Qing-Xia Li
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Ling-Xia Zhang
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Jia-Li Chen
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, Guangzhou, 510632, China.
| | - Jun Chi
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Li-Li Zhu
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Er-Ping Xu
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| | - Zhi-Min Wang
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Li-Ping Dai
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China.
| |
Collapse
|
6
|
Yang X, Chi C, Li W, Zhang Y, Yang S, Xu R, Liu R. Metabolomics and lipidomics combined with serum pharmacochemistry uncover the potential mechanism of Huang-Lian-Jie-Du decoction alleviates atherosclerosis in ApoE -/- mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117748. [PMID: 38216103 DOI: 10.1016/j.jep.2024.117748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis (AS) is one of the main cardiovascular diseases (CVDs) leading to an increase in global mortality, and its key pathological features are lipid accumulation and oxidative stress. Huang-Lian-Jie-Du decoction (HLJDD), a representative formula for clearing heat and detoxifying, has been shown to reduce aortic lipid plaque and improve AS. However, multiple components and multiple targets of HLJDD pose a challenge in comprehending its comprehensive mechanism in the treatment of AS. AIM OF THE STUDY This study was designed to illustrate the anti-AS mechanisms of HLJDD in an apolipoprotein E-deficient (ApoE-/-) mouse model from a metabolic perspective. MATERIALS AND METHODS ApoE-/- mice were kept on a high-fat diet (HFD) to induce AS. Serum total cholesterol (TC), total triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) levels were determined to evaluate the influence of HLJDD on dyslipidemia. Oil red O was used to stain mouse aortic lipid plaques, and hematoxylin and eosin (HE) staining was used to assess the pathological changes in the aortic roots. Metabolomics and lipidomics combined with serum pharmacochemistry were performed to research the HLJDD mechanism of alleviating AS. RESULTS In this study, HLJDD treatment improved serum biochemical levels and histopathological conditions in AS mice. A total of 6 metabolic pathways (arginine biosynthesis, glycerophospholipid, sphingolipid, arachidonic acid, linoleic acid, and glycerolipid metabolism) related to 25 metabolic biomarkers and 41 lipid biomarkers were clarified, and 22 prototype components migrating to blood were identified after oral administration of HLJDD. CONCLUSION HLJDD improved AS induced by HFD in ApoE-/- mice. The effects of HLJDD were mainly attributed to regulating lipid metabolism by regulating the metabolic pathways of glycerophospholipids, sphingolipids, arachidonic acid, linoleic acid, and glycerolipids and reducing the levels of oxidative stress by upregulating arginine biosynthesis.
Collapse
Affiliation(s)
- Xiaoli Yang
- 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
| | - Yanyan 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
| | - Shufang Yang
- 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
| | - Ruoxuan Xu
- 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
|
7
|
Fu J, Liang Y, Yu D, Wang Y, Lu F, Liu S. Radix Saposhnikoviae enhancing Huangqi Chifeng Decoction improves lipid metabolism in AS mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117479. [PMID: 37992882 DOI: 10.1016/j.jep.2023.117479] [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: 09/26/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqi Chifeng decoction (HQCF) combined with parsnips is a classic Chinese traditional medicine formula that has certain advantages in the clinical treatment of cardiovascular and cerebrovascular diseases. At present, there is an absence of research on the regulatory effect and mechanism of this formula on atherosclerosis (AS). The synergistic effect of Radix Saposhnikoviae (RS) in HQCF is also unclear. AIM OF THE STUDY This study was designed to investigate the role of RS, which is designed as a guide drug for HQCF, in improving the lipid metabolism of AS. MATERIALS AND METHODS In this study, we studied the effect of HQCF on ApoE-/- mice before and after RS compatibility. Hematoxylin and eosin (HE) staining and oil red staining were used to evaluate atherosclerotic lesions and lipid accumulation in the aorta and liver, respectively. The expression of adenosine monophosphate-activated protein kinase (AMPK) and pAMPK in the aorta was measured by immunofluorescence, and AMPK and sterol regulatory element binding protein-1 (SREBP-1),fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC) in liver tissue were measured by Western blot analysis. Metabolomics was used to compare the changes in serum and liver metabolites of ApoE-/- mice before and after RS combination. RESULTS Compared with the control group, the serum lipid levels of ApoE-/- mice increased, the aortic intima thickened with plaque formation, and liver tissue pathological changes and lipid deposition occurred. Both (HQCFT without RS)HQCS and HQCF can improve the pathological condition of tissue and regulate the blood lipid level. It was noted that HQCF could promote the phosphorylation of AMPK to activate it, inhibit the expression of SREBP-1c and FAS, reduce lipid synthesis, and inhibit ACC to promote the oxidative decomposition of fatty acids. Serum and liver metabolome results showed that HQCS and HQCF treated AS mainly by regulating glycerophospholipid metabolism, sphingolipid metabolism and the arachidonic acid metabolism pathway. Importantly, HQCF showed better efficacy in regulating lipid metabolism than the HQCS group. CONCLUSION HQCF decoction reduces atherosclerotic lesions in the aorta and lipid accumulation in the liver, which may regulate lipid transport and metabolic function by activating the AMPK pathway. These effects can be attributed to the guidance and synergism of RS.
Collapse
Affiliation(s)
- Jiaqi Fu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yuqin Liang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yu Wang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
8
|
Zhang Q, Xu R, Xue R, Mei X, Qin Y, Shen K, Xu J, Su L, Mao C, Xie H, Lu T. Ultra-high-performance liquid chromatography-quadrupole-time of flight-mass spectrometry combined with network pharmacology for analysis of potential quality markers of three processed products of Qingpi. J Sep Sci 2024; 47:e2300281. [PMID: 37994479 DOI: 10.1002/jssc.202300281] [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: 04/10/2023] [Revised: 10/16/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
Qingpi, a well-known traditional Chinese medicine for qi-regulating and commonly processed into three types of pieces, has been widely used in the clinical application of liver disease for thousands of years. In this study, an ultra-high-performance liquid chromatography-quadrupole-time of flight-mass spectrometry approach along with multivariate statistical analysis was developed to assess and characterize the differentiations of three processed products and confirm the potential quality markers of Qingpi. In addition, a systematic analysis combined with network pharmacology and molecular docking was performed to clarify the potential mechanism of Qingpi for the treatment of liver disease. As a result, 18 components were identified and an integrated network of Qingpi-Components-Target-Pathway-Liver Disease was constructed. Eight compounds were finally screened out as the potential quality markers acting on ten main targets and pathways of liver disease. Molecular docking analysis results indicated that the quality markers had a good binding activity with the targets. Overall, this work preliminarily identified the potential quality markers of three processed products of Qingpi, and predicted its targets in the prevention and treatment of liver disease, which can provide supporting information for further study of the pharmacodynamic substances and mechanisms of Qingpi.
Collapse
Affiliation(s)
- Qian Zhang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Ruijie Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Rong Xue
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Xi Mei
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yuwen Qin
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Ke Shen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Jinguo Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Lianlin Su
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Chunqin Mao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Hui Xie
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Tulin Lu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| |
Collapse
|
9
|
Sun X, Xi Y, Yan M, Sun C, Tang J, Dong X, Yang Z, Wu L. Lactiplantibacillus plantarum NKK20 Increases Intestinal Butyrate Production and Inhibits Type 2 Diabetic Kidney Injury through PI3K/Akt Pathway. J Diabetes Res 2023; 2023:8810106. [PMID: 38162631 PMCID: PMC10757665 DOI: 10.1155/2023/8810106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024] Open
Abstract
Nephropathy injury is a prevalent complication observed in individuals with diabetes, serving as a prominent contributor to end-stage renal disease, and the advanced glycation products (AGEs) are important factors that induce kidney injury in patients with diabetes. Addressing this condition remains a challenging aspect in clinical practice. The aim of this study was to explore the effects of Lactiplantibacillus plantarum NKK20 strain (NKK20) which protects against diabetic kidney disease (DKD) based on animal and cell models. The results showed that the NKK20 can significantly reduce renal inflammatory response, serum oxidative stress response, and AGE concentration in diabetic mice. After treatment with NKK20, the kidney damage of diabetic mice was significantly improved, and more importantly, the concentration of butyrate, a specific anti-inflammatory metabolite of intestinal flora in the stool of diabetic mice, was significantly increased. In addition, nontargeted metabolomics analysis showed a significant difference between the metabolites in the mouse serum contents of the NKK20 administration group and those in the nephropathy injury group, in which a total of 24 different metabolites that were significantly affected by NKK20 were observed, and these metabolites were mainly involved in glycerophospholipid metabolism and arachidonic acid metabolism. Also, the administration of butyrate to human kidney- (HK-) 2 cells that were stimulated by AGEs resulted in a significant upregulation of ZO-1, Occludin, and E-cadherin gene expressions and downregulation of α-SMA gene expression. This means that butyrate can maintain the tight junction structure of HK-2 cells and inhibit fibrosis. Butyrate also significantly inhibited the activation of PI3K/Akt pathway. These results indicate that NKK20 can treat kidney injury in diabetic mice by reducing blood glucose and AGE concentration and increasing butyrate production in the intestine. By inhibiting PI3K pathway activation in HK-2 cells, butyrate maintains a tight junction structure of renal tubule epithelial cells and inhibits renal tissue fibrosis. These results suggest that NKK20 is helpful to prevent and treat the occurrence and aggravation of diabetic kidney injury.
Collapse
Affiliation(s)
- Xiaohong Sun
- Department of Clinical Laboratory, Yizheng Hospital, Nanjing Drum Tower Hospital Group, Yizheng 210008, China
| | - Yue Xi
- Medical Laboratory Department, Huai'an Second People's Hospital, Huai'an 223022, China
| | - Man Yan
- Department of Clinical Laboratory, Zhenjiang City Central Blood Station, Zhenjiang 212399, China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Chang Sun
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jianjun Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xueyun Dong
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Zhengnan Yang
- Department of Clinical Laboratory, Yizheng Hospital, Nanjing Drum Tower Hospital Group, Yizheng 210008, China
| | - Liang Wu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
10
|
Chen T, Zhang Y, Chen M, Yang P, Wang Y, Zhang W, Huang W, Zhang W. Tongmai Yangxin pill alleviates myocardial no-reflow by activating GPER to regulate HIF-1α signaling and downstream potassium channels. PHARMACEUTICAL BIOLOGY 2023; 61:499-513. [PMID: 36896463 PMCID: PMC10013430 DOI: 10.1080/13880209.2023.2184481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 12/23/2022] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT The Tongmai Yangxin pill (TMYX) has potential clinical effects on no-reflow (NR); however, the effective substances and mechanisms remain unclear. OBJECTIVE This study evaluates the cardioprotective effects and molecular mechanisms of TMYX against NR. MATERIALS AND METHODS We used a myocardial NR rat model to confirm the effect and mechanism of action of TMYX in alleviating NR. Sprague-Dawley (SD) rats were divided into Control (Con), sham, NR, TMYX (4.0 g/kg), and sodium nitroprusside (SNP, 5.0 mg/kg), and received their treatments once a day for one week. In vitro studies in isolated coronary microvasculature of NR rats and in silico network pharmacology analyses were performed to reveal the underlying mechanisms of TMYX and determine the main components, targets, and pathways of TMYX, respectively. RESULTS TMYX (4.0 g/kg) showed therapeutic effects on NR by improving the cardiac structure and function, reducing NR, ischemic areas, and cardiomyocyte injury, and decreasing the expression of cardiac troponin I (cTnI). Moreover, the mechanism of TMYX predicted by network pharmacology is related to the HIF-1, NF-κB, and TNF signaling pathways. In vivo, TMYX decreased the expression of MPO, NF-κB, and TNF-α and increased the expression of GPER, p-ERK, and HIF-1α. In vitro, TMYX enhanced the diastolic function of coronary microvascular cells; however, this effect was inhibited by G-15, H-89, L-NAME, ODQ and four K+ channel inhibitors. CONCLUSIONS TMYX exerts its pharmacological effects in the treatment of NR via multiple targets. However, the contribution of each pathway was not detected, and the mechanisms should be further investigated.
Collapse
Affiliation(s)
- Ting Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, People's Republic of China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, People's Republic of China
- Institute of Traditional Chinese medicine, Tianjin University of Traditional Chinese medicine, Tianjin, People's Republic of China
| | - Yulong Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, People's Republic of China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, People's Republic of China
| | - Manyun Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, People's Republic of China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, People's Republic of China
| | - Pu Yang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yi Wang
- Institute of Traditional Chinese medicine, Tianjin University of Traditional Chinese medicine, Tianjin, People's Republic of China
| | - Wei Zhang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, People's Republic of China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, People's Republic of China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, People's Republic of China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, People's Republic of China
| |
Collapse
|
11
|
Qian X, Hai W, Chen S, Zhang M, Jiang X, Tang H. Multi-omics data reveals aberrant gut microbiota-host glycerophospholipid metabolism in association with neuroinflammation in APP/PS1 mice. Gut Microbes 2023; 15:2282790. [PMID: 37992400 PMCID: PMC10730179 DOI: 10.1080/19490976.2023.2282790] [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: 04/26/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023] Open
Abstract
Numerous studies have described the notable impact of gut microbiota on the brain in Alzheimer's disease (AD) via the gut - brain axis. However, the molecular mechanisms underlying the involvement of gut microbiota in the development of AD are limited. This study aimed to explore the potential mechanisms of gut microbiota in AD by integrating multi-omics data. In this study, APP/PS1 and WT mice at nine months of age were used as study mouse model. Cognitive function was assessed using the Morris water maze test. The levels of Aβ plaque and neuroinflammation in the brain were detected using immunofluorescence and PET/CT. In addition, we not only used 16S rRNA gene sequencing and metabolomics to explore the variation characteristics of gut microbiota and serum metabolism abundance, but also combined spatial metabolomics and transcriptomics to explore the change in the brain and identify their potential correlation. APP/PS1 mice showed significant cognitive impairment and amyloid-β deposits in the brain. The abundance of gut microbiota was significantly changed in APP/PS1 mice, including decreased Desulfoviobrio, Enterococcus, Turicibacter, and Ruminococcus and increased Pseudomonas. The integration of serum untargeted metabolomics and brain spatial metabolomics showed that glycerophospholipid metabolism was a common alteration pathway in APP/PS1 mice. Significant proliferation and activation of astrocyte and microglia were observed in APP/PS1 mice, accompanied by alterations in immune pathways. Integration analysis and fecal microbiota transplantation (FMT) intervention revealed potential association of gut microbiota, host glycerophospholipid metabolism, and neuroinflammation levels in APP/PS1 mice.
Collapse
Affiliation(s)
- Xiaohang Qian
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyue Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xufeng Jiang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huidong Tang
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Mao T, Xie L, Guo Y, Ji X, Wan J, Cui X, Fan Q, Liu W, Wang S, Han W, Lin Q, Jia W. Mechanistic exploration of Yiqi Liangxue Shengji prescription on restenosis after balloon injury by integrating metabolomics with network pharmacology. PHARMACEUTICAL BIOLOGY 2023; 61:1260-1273. [PMID: 37602438 PMCID: PMC10443980 DOI: 10.1080/13880209.2023.2244533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/28/2023] [Accepted: 07/31/2023] [Indexed: 08/22/2023]
Abstract
CONTEXT Yiqi Liangxue Shengji prescription (YQLXSJ) is a traditional Chinese medicine (TCM) formula that has long been used for treatment after percutaneous coronary intervention (PCI). OBJECTIVE To investigate the putative pharmacological mechanism of YQLXSJ on restenosis through an integrated approach utilizing metabolomics and network pharmacology. MATERIALS AND METHODS Forty male Sprague-Dawley rats were divided into sham, model, YQLXSJ, and positive groups. YQLXSJ group received the treatment of YQLXSJ (6 g/kg/d, i.g.) and the positive group was treated with atorvastatin (2 mg/kg/d, i.g.). After 4 weeks, the improvement in intimal hyperplasia was evaluated by ultrasound, H&E staining, and immunofluorescence. UPLC-MS/MS technology was utilized to screen the differential metabolites. Network pharmacology was conducted using TCMSP, GeneCards, and Metascape, etc., in combination with metabolomics. Eventually, the core targets were acquired and validated. RESULTS Compared to models, YQLXSJ exhibited decreased intima-media thickness on ultrasound (0.23 ± 0.02 mm vs. 0.20 ± 0.01 mm, p < 0.01) and reduced intima thickness by H&E (30.12 ± 6.05 μm vs. 14.32 ± 1.37 μm, p < 0.01). We identified 18 differential metabolites and 5 core targets such as inducible nitric oxide synthase (NOS2), endothelial nitric oxide synthase (NOS3), vascular endothelial growth factor-A (VEGFA), ornithine decarboxylase-1 (ODC1) and group IIA secretory phospholipase A2 (PLA2G2A). These targets were further confirmed by molecular docking and ELISA. DISCUSSION AND CONCLUSIONS This study confirms the effects of YQLXSJ on restenosis and reveals some biomarkers. TCM has great potential in the prevention and treatment of restenosis by improving metabolic disorders.
Collapse
Affiliation(s)
- Tianshi Mao
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Long Xie
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Yanqiong Guo
- Department of Cardiology, Beijing Fengtai District Hospital of Chinese Medicine, Beijing, P.R. China
| | - Xiang Ji
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Jie Wan
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Xiaoyun Cui
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Qian Fan
- Department of Cardiology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, P.R. China
| | - Wei Liu
- Department of Cardiology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, P.R. China
| | - Shuai Wang
- Department of Cardiology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, P.R. China
| | - Wenbo Han
- Department of Cardiology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, P.R. China
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, P.R. China
| | - Wenhao Jia
- Department of Cardiology, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, P.R. China
| |
Collapse
|
13
|
Wang L, Li Z, Lu T, Su L, Mao C, Zhang Y, Zhang X, Jiang X, Xie H, Yu X. The potential mechanism of Choulingdan mixture in improving acute lung injury based on HPLC-Q-TOF-MS, network pharmacology and in vivo experiments. Biomed Chromatogr 2023; 37:e5709. [PMID: 37533317 DOI: 10.1002/bmc.5709] [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: 03/05/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/04/2023]
Abstract
Choulingdan mixture (CLDM) is an empirical clinical prescription for the adjuvant treatment of acute lung injury (ALI). CLDM has been used for almost 30 years in the clinic. However, its mechanism for improving ALI still needs to be investigated. In this study, high-performance liquid chromatography-quadrupole/time-of-flight mass spectrometry (HPLC-Q-TOF-MS/MS) was applied to characterize the overall chemical composition of CLDM. A total of 93 ingredients were characterized, including 25 flavonoids, 20 organic acids, 11 saponins, nine terpenoids, seven tannins and 21 other compounds. Then network pharmacology was applied to predict the potential bioactive components, target genes and signaling pathways of CLDM in improving ALI. Additionally, molecular docking was performed to demonstrate the interaction between the active ingredients and the disease targets. Finally, animal experiments further confirmed that CLDM significantly inhibits pulmonary inflammation, pulmonary edema and oxidative stress in lipopolysaccharide-induced ALI mice by inhibiting the PI3K-AKT signaling pathway. This study enhanced the amount and accuracy of compounds of CLDM and provided new insights into CLDM preventing and treating ALI.
Collapse
Affiliation(s)
- Lili Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengyan Li
- Department of Pharmacy, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, China
| | - Tulin Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lianlin Su
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunqin Mao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiting Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinrui Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaofeng Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Xie
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoling Yu
- Department of Pharmacy, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
14
|
Zhou H, Fang N, Zhou P, Lin B, Wei X, Fu W, Ding Z, Yang J, Wan H. Study on the Mechanism of Naoxintong in the Treatment of Cerebral Ischemia-Reperfusion Injury Based on a Multiomics Method. Rejuvenation Res 2023; 26:159-169. [PMID: 37261991 DOI: 10.1089/rej.2023.0009] [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] [Indexed: 06/03/2023] Open
Abstract
Cerebral ischemia-reperfusion (CIR) injury occurs as a secondary injury during the treatment of ischemic stroke (IS). There is a high death rate and morbidity due to IS throughout the world. Even though Naoxintong Capsule (NXT) is effective in the treatment of CIR, its mechanisms of action are unclear. The study aims to explore the clear mechanism associated with NXT therapy for CIR. We established the model of middle cerebral artery occlusion to evaluate the neurological function and assess the infarct size. Brain tissue metabolomics was used to identify different metabolites, and metabolic profiling systems enriched metabolic pathways. Then, the potential targets of NXT in the treatment of CIR were explored by proteomic, transcriptomic, and metabolomic methods. NXT improves CIR symptoms. We found potential 11 proteins and corresponding metabolites involved in NXT treatment of CIR. Most of these metabolites are regulated to restore after treatment. According to network pharmacology, we found 6 hub genes, including Glb1, Gmps, Pfas, Atic, Gaa, and Acox1, and their associated core metabolites and pathways. This study reveals the complex mechanism of NXT in treating CIR, and provides a new strategy for future researchers to screen related targets and pathways.
Collapse
Affiliation(s)
- Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Ningji Fang
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Peng Zhou
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Bingying Lin
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Xiaoyu Wei
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Wei Fu
- Department of Cardiac-Cerebral Diseases, Yinchuan Cardiac-Cerebral Treatment Internet Hospital, Yinchuan, P.R. China
| | - Zhishan Ding
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| |
Collapse
|
15
|
Liu X, Shao P, Wang Y, Chen Y, Cui S. Anti-inflammatory mechanism of the optimized active ingredients of Sargentodoxa cuneata and Patrinia villosa. Int Immunopharmacol 2023; 120:110337. [PMID: 37244114 DOI: 10.1016/j.intimp.2023.110337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 05/29/2023]
Abstract
Pelvic inflammatory disease (PID) is a common gynecological infection. The combined use of Sargentodoxa cuneata (da xue teng) and Patrinia villosa (bai jiang cao) has been shown to inhibit PID progression. The active components of S. cuneata (emodin, Emo) and P. villosa (acacetin, Aca; oleanolic acid, OA; sinoacutine, Sin) have been identified but the mode of action of this combination of compounds against PID has not been clarified. Therefore, this study aims to investigate the mechanism of these active components against PID through network pharmacological, molecular docking and experimental validation. The results showed the optimal combination of components was 40 µM Emo + 40 µM OA, 40 µM Emo + 40 µM Aca, and 40 µM Emo + 150 µM Sin by cell proliferation and NO release. The potential key targets of this combination in the treatment of PID include SRC, GRB2, PIK3R1, PIK3CA, PTPN11, and SOS1, which act on signaling pathways such as EGFR, PI3K/Akt, TNF, and IL-17. Emo, Aca, OA, and their optimal combination inhibited the expression of IL-6, TNF-α, MCP-1, IL-12p70, IFN-γ, and the M1 phenotype markers CD11c and CD16/32, and promoted the expression of the M2 phenotype markers CD206 and arginase 1 (Arg1). Western blotting confirmed that Emo, Aca, OA, and their optimal combination significantly inhibited the expression of glucose metabolism-related proteins PKM2, PD, HK I, and HK II. This study proved the advantage of combination use of active components from S. cuneata and P. villosa, and clarified that they exert the anti-inflammatory effect by regulation of M1/M2 phenotype transition and regulation of glucose metabolism. The results provide a theoretical basis for the clinical treatment of PID.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, China
| | - Puwei Shao
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, China
| | - Ying Wang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, China
| | - Yuanyuan Chen
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, China
| | - Shuna Cui
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou 225000, China; Department of Gynecology and Obstetrics, Affiliated Hospital of Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
16
|
Law SH, Chan HC, Ke GM, Kamatam S, Marathe GK, Ponnusamy VK, Ke LY. Untargeted Lipidomic Profiling Reveals Lysophosphatidylcholine and Ceramide as Atherosclerotic Risk Factors in apolipoprotein E Knockout Mice. Int J Mol Sci 2023; 24:ijms24086956. [PMID: 37108120 PMCID: PMC10138920 DOI: 10.3390/ijms24086956] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the availability and use of numerous cholesterol-lowering drugs, atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of mortality globally. Many researchers have focused their effort on identifying modified lipoproteins. However, lipid moieties such as lysophosphatidylcholine (LPC) and ceramide (CER) contribute to atherogenic events. LPC and CER both cause endothelial mitochondrial dysfunction, leading to fatty acid and triglyceride (TG) accumulation. In addition, they cause immune cells to differentiate into proinflammatory phenotypes. To uncover alternative therapeutic approaches other than cholesterol- and TG-lowering medications, we conducted untargeted lipidomic investigations to assess the alteration of lipid profiles in apolipoprotein E knockout (apoE-/-) mouse model, with or without feeding a high-fat diet (HFD). Results indicated that, in addition to hypercholesterolemia and hyperlipidemia, LPC levels were two to four times higher in apoE-/- mice compared to wild-type mice in C57BL/6 background, regardless of whether they were 8 or 16 weeks old. Sphingomyelin (SM) and CER were elevated three- to five-fold in apoE-/- mice both at the basal level and after 16 weeks when compared to wild-type mice. After HFD treatment, the difference in CER levels elevated more than ten-fold. Considering the atherogenic properties of LPC and CER, they may also contribute to the early onset of atherosclerosis in apoE-/- mice. In summary, the HFD-fed apoE-/- mouse shows elevated LPC and CER contents and is a suitable model for developing LPC- and CER-lowering therapies.
Collapse
Affiliation(s)
- Shi-Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hua-Chen Chan
- Department of Medical Laboratory Science, College of Medicine, I-Shou University, Kaohsiung 84001, Taiwan
- Center for Lipid Biosciences, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Guan-Ming Ke
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| | - Swetha Kamatam
- Department of Studies in Biochemistry and Molecular Biology, University of Mysore, Manasagangothri, Mysuru 570006, India
| | - Gopal Kedihithlu Marathe
- Department of Studies in Biochemistry and Molecular Biology, University of Mysore, Manasagangothri, Mysuru 570006, India
| | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry, Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Lipid Biosciences, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
17
|
Zhang L, Xiong L, Fan L, Diao H, Tang M, Luo E, Guo W, Yang X, Xing S. Vascular lipidomics analysis reveales increased levels of phosphocholine and lysophosphocholine in atherosclerotic mice. Nutr Metab (Lond) 2023; 20:1. [PMID: 36600244 PMCID: PMC9811766 DOI: 10.1186/s12986-022-00723-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Atherosclerosis (AS) is the major cause of cardiovascular disease, and dyslipidemia is a principal determinant of the initiation and progression of AS. Numerous works have analyzed the lipid signature of blood, but scarce information on the lipidome of vascular tissue is available. This study investigated the lipid profile in the aorta of ApoE-/- mice. METHOD ApoE-/- mice were randomly divided into two groups: (1) the normal diet (ND) group and (2) the high-fat diet (HFD) group. After feeding for 8 weeks, the plasma low-density lipoprotein (LDL), total cholesterol (TC), and triglyceride (TGs) levels were measured. UHPLC-Q Exactive plus MS was used to assess the lipid profile using both positive and negative ionization modes. RESULTS LDL and TC levels were significantly increased in HFD mice, and lipid deposition, plaque area and collagen fiber levels were increased in HFD group. In addition, a total of 131 differential lipids were characterized, including 57 lipids with levels that were increased in the HFD group and 74 with levels that were decreased. Further analysis revealed that the levels of several differentially expressed phosphocholines (PCs) and lysophosphocholines (LPCs) were significantly increased. These PCs included PC (38:3), PC (36:4), PC (36:3), PC (36:2), PC (36:1), PC (34:1e), PC (34:1), PC (32:1), PC (18:0/18:1), and PC (38:5), and the LPCs included LPC (18:1), LPC (18:0) and LPC (16:0). CONCLUSION Our findings indicate the presence of a comprehensive lipid profile in the vascular tissue of atherosclerotic mice, particularly involving PC and LPC, which exhibited significantly increased levels in AS.
Collapse
Affiliation(s)
- Li Zhang
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China ,grid.16821.3c0000 0004 0368 8293Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336 China
| | - Liling Xiong
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Li Fan
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Haoyang Diao
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Mi Tang
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Erdan Luo
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Wenmei Guo
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Xiao Yang
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Shasha Xing
- grid.54549.390000 0004 0369 4060Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731 China
| |
Collapse
|
18
|
Lin CN, Hsu KC, Huang KL, Huang WC, Hung YL, Lee TH. Identification of Metabolomics Biomarkers in Extracranial Carotid Artery Stenosis. Cells 2022; 11:3022. [PMID: 36230983 PMCID: PMC9563778 DOI: 10.3390/cells11193022] [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: 07/16/2022] [Revised: 08/28/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
The biochemical identification of carotid artery stenosis (CAS) is still a challenge. Hence, 349 male subjects (176 normal controls and 173 stroke patients with extracranial CAS ≥ 50% diameter stenosis) were recruited. Blood samples were collected 14 days after stroke onset with no acute illness. Carotid plaque score (≥2, ≥5 and ≥8) was used to define CAS severity. Serum metabolites were analyzed using a targeted Absolute IDQ®p180 kit. Results showed hypertension, diabetes, smoking, and alcohol consumption were more common, but levels of diastolic blood pressure, HDL-C, LDL-C, and cholesterol were lower in CAS patients than controls (p < 0.05), suggesting intensive medical treatment for CAS. PCA and PLS-DA did not demonstrate clear separation between controls and CAS patients. Decision tree and random forest showed that acylcarnitine species (C4, C14:1, C18), amino acids and biogenic amines (SDMA), and glycerophospholipids (PC aa C36:6, PC ae C34:3) contributed to the prediction of CAS. Metabolite panel analysis showed high specificity (0.923 ± 0.081, 0.906 ± 0.086 and 0.881 ± 0.109) but low sensitivity (0.230 ± 0.166, 0.240 ± 0.176 and 0.271 ± 0.169) in the detection of CAS (≥2, ≥5 and ≥8, respectively). The present study suggests that metabolomics profiles could help in differentiating between controls and CAS patients and in monitoring the progression of CAS.
Collapse
Affiliation(s)
- Chia-Ni Lin
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan 333, Taiwan
| | - Kai-Cheng Hsu
- School of Medicine, College of Medicine, Artificial Intelligence Center for Medical Diagnosis, and Department of Neurology, China Medical University Hospital, Taichung 404327, Taiwan
| | - Kuo-Lun Huang
- Stroke Center and Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Wen-Cheng Huang
- Department of Nuclear Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yi-Lun Hung
- Department of Nuclear Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Tsong-Hai Lee
- Stroke Center and Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
19
|
Differential Response of Ileal and Colonic Microbiota in Rats with High-Fat Diet-Induced Atherosclerosis. Int J Mol Sci 2022; 23:ijms231911154. [PMID: 36232451 PMCID: PMC9569969 DOI: 10.3390/ijms231911154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 12/03/2022] Open
Abstract
Growing evidence suggests that gut microbiota are associated with atherosclerosis (AS). However, the functional heterogeneity of each gut segment gives rise to regional differences in gut microbiota. We established a rat model of AS by feeding the rats a high-fat diet for a long period. The pathological and microbiota changes in the ileum and colon of the rats were examined, and correlations between AS and microbiota were analyzed. The aortic mesothelium of the experimental rats was damaged. The intima showed evident calcium salt deposition, indicating that the AS rat model was successfully developed. We noted varying degrees of pathological damage in the ileum and colon of the experimental rats. The 16S rDNA high-throughput sequencing showed significant differences in α-diversity, β-diversity, and microbiota comparisons in the ileum and colon. Furthermore, the ileum and colon of AS rats showed varying degrees of intestinal microbiota disturbance. This article contributes to the study of the relationship between the microbiota in different regions of the gut and AS, and provides new approaches in gut microbiota intervention for the treatment of AS.
Collapse
|
20
|
Song Y, Su D, Yang Y, Zeng Q, Liao L, Chen C, Yang M, Zhu G, Zhang R, Ai Z, Li Y. Two Species Origins Comparison of Herba Patriniae based on Their Ingredients Profile by UPLC-QTOF/MS/MS and Orthogonal Partial Least Squares Discriminant Analysis. Chem Biodivers 2022; 19:e202100961. [PMID: 35979749 DOI: 10.1002/cbdv.202100961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 08/15/2022] [Indexed: 11/10/2022]
Abstract
Herba Patriniae (HP) is widely used as a medicinal and edible material in China. Besides food value, HP attracts more attention due to its medicinal potential. Patrinia villosa Juss. ( PV ) and Patrinia scabiosaefolia Fisch. ( PS ) are the two species origins of HP. These two of HP show different effects on cell proliferation, migration, angiogenesis and anti-diabetic. As we have previously reported, PV and PS show significant differences on their anti-inflammatory ability in the same experimental model. Comparing the ingredient profiles of two different sources will not only facilitate the understanding of their medicinal effects, but also help the development and research of new activities. However, still now, there is no systematic and detailed study to compare the components of PV and PS . In present study, ultra-high performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry was employed to achieve a high-throughput qualitative and thorough analysis of the chemical composition spectrum of HP. A total of 164 compounds were identified, among these compounds, 127 compounds were identified from PV , and 107 compounds were identified from PS . Most of the chemical components was discovered for the first time. Flavonoids, saponins, terpenoids and organic acids, as the main ingredients in PV and PS were 45.45%vs 28.46%, 12.61% vs 32.09%, 14.33% vs 22.38% and 14.58% vs 6.79%, respectively. Flavonoids are the main components of PV , while PS is rich in saponins. PV and PS were classified into two groups by principal component analysis (PCA) and screened out the main molecular differences responsible by orthogonal partial least squares discriminant analysis (OPLS-DA). All the results will be a guide for the quality control, functional activity research, or better clinic use based on the ingredients profile between these two species. Besides, this first study on ingredients profile of two species origins will be beneficial for potential and best resources utilization of both PV and PS .
Collapse
Affiliation(s)
- Yonggui Song
- Jiangxi University of Traditional Chinese Medicine, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, 330000, Nanchang, CHINA
| | - Da Su
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Yanyan Yang
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Qiang Zeng
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Liangliang Liao
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Changlian Chen
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Ming Yang
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Genhua Zhu
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Ruowen Zhang
- Shenzhen Honsan Health Industry Group, Shenzhen Honsan Health Industry Group, 2028 Shenyan Road, Haishan street, Shenzhen, China, Shenzhen, CHINA
| | - Zhifu Ai
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| | - Yanzhen Li
- Jiangxi University of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, Nanchang, CHINA
| |
Collapse
|
21
|
Ji T, Wang J, Xu Z, Cai HD, Su SL, Peng X, Ruan HS. Combination of mulberry leaf active components possessed synergetic effect on SD rats with diabetic nephropathy by mediating metabolism, Wnt/β-catenin and TGF-β/Smads signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115026. [PMID: 35074452 DOI: 10.1016/j.jep.2022.115026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/25/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mulberry leaf has attracted much attention due to its excellent curative effect on diabetes and its complications, whether the combination of its effective components have protective and synergistic effect on diabetic nephropathy (DN) in vivo remain unclear. AIM OF THE STUDY The aim of this study was to investigate the protective and synergistic effect of the combination (MAF1:1 and MAF1:5) of mulberry leaf alkaloids (MA) and flavonoids extract (MF) on DN. MATERIALS AND METHODS A step by step method consisted of network pharmacological prediction, animal in vivo validation and metabolic mechanism research was used to construct the multi-component-target-pathway network of mulberry leaf against DN. Firstly, the potential components and mechanism of mulberry leaf against DN was explored by network pharmacology analysis. Secondly, DN animal model was established to validate the anti-DN activity of these potential compounds. Thirdly, the metabolomics of serum and urine samples from animal experiments was analyzed to explore the anti-DN mechanism of these potential compounds. RESULTS The results of network pharmacology demonstrated that a total of 7 compounds detected in MA and MF exhibited anti-DN activity, their mechanism were strongly in connection with metabolic pathways, arachidonic acid metabolism, sphingolipid signaling pathway, etc. The results of animal experiment indicated that MAF1:1 and MAF1:5 significantly relieved metabolic disorders through regulating Wnt/β-catenin and TGF-β/Smads signaling pathway, just like MF or MA alone. Metabolomics suggested they could regulate 16 serum and 7 urine endogenous metabolites through arachidonic acid metabolism, phenylalanine metabolism and sphingolipid metabolism, thus alleviated DN. Significantly, MAF1:1 and MAF1:5 might possess synergistic effect considering their therapeutic effects on DN rats were superior to the single use of MA or MF. CONCLUSIONS MAF1:1 and MAF1:5 possessed protective and synergistic effect on DN rats through multi-target and multi-pathways. These findings were of great scientific significance and application value to reveal the advantage of mulberry leaf in preventing and treating DN.
Collapse
Affiliation(s)
- Tao Ji
- Zhejiang Pharmaceutical College, Zhejiang Province, Ningbo, 315100, PR China
| | - Juan Wang
- Zhejiang Pharmaceutical College, Zhejiang Province, Ningbo, 315100, PR China
| | - Zhuo Xu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Die Cai
- Zhejiang Pharmaceutical College, Zhejiang Province, Ningbo, 315100, PR China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shu-Lan Su
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xin Peng
- Ningbo Research Institute of Zhejiang University, Zhejiang Province, Ningbo, 315100, PR China.
| | - Hong-Sheng Ruan
- Zhejiang Pharmaceutical College, Zhejiang Province, Ningbo, 315100, PR China
| |
Collapse
|