1
|
Chen J, Ye W. Molecular mechanisms underlying Tao-Hong-Si-Wu decoction treating hyperpigmentation based on network pharmacology, Mendelian randomization analysis, and experimental verification. PHARMACEUTICAL BIOLOGY 2024; 62:296-313. [PMID: 38555860 DOI: 10.1080/13880209.2024.2330609] [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: 10/23/2023] [Accepted: 03/02/2024] [Indexed: 04/02/2024]
Abstract
CONTEXT Hyperpigmentation, a common skin condition marked by excessive melanin production, currently has limited effective treatment options. OBJECTIVE This study explores the effects of Tao-Hong-Si-Wu decoction (THSWD) on hyperpigmentation and to elucidate the underlying mechanisms. MATERIALS AND METHODS We employed network pharmacology, Mendelian randomization, and molecular docking to identify THSWD's hub targets and mechanisms against hyperpigmentation. The Cell Counting Kit-8 (CCK-8) assay determined suitable THSWD treatment concentrations for PIG1 cells. These cells were exposed to graded concentrations of THSWD-containing serum (2.5%, 5%, 10%, 15%, 20%, 30%, 40%, and 50%) and treated with α-MSH (100 nM) to induce an in vitro hyperpigmentation model. Assessments included melanin content, tyrosinase activity, and Western blotting. RESULTS ALB, IL6, and MAPK3 emerged as primary targets, while quercetin, apigenin, and luteolin were the core active ingredients. The CCK-8 assay indicated that concentrations between 2.5% and 20% were suitable for PIG1 cells, with a 50% cytotoxicity concentration (CC50) of 32.14%. THSWD treatment significantly reduced melanin content and tyrosinase activity in α-MSH-induced PIG1 cells, along with downregulating MC1R and MITF expression. THSWD increased ALB and p-MAPK3/MAPK3 levels and decreased IL6 expression in the model cells. DISCUSSION AND CONCLUSION THSWD mitigates hyperpigmentation by targeting ALB, IL6, and MAPK3. This study paves the way for clinical applications of THSWD as a novel treatment for hyperpigmentation and offers new targeted therapeutic strategies.
Collapse
Affiliation(s)
- Jun Chen
- Department of Geriatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Wenyi Ye
- Department of Traditional Chinese Internal Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
2
|
Velázquez-Enríquez JM, Santos-Álvarez JC, Ramírez-Hernández AA, Reyes-Jiménez E, Pérez-Campos Mayoral L, Romero-Tlalolini MDLÁ, Jiménez-Martínez C, Arellanes-Robledo J, Villa-Treviño S, Vásquez-Garzón VR, Baltiérrez-Hoyos R. Chlorogenic acid attenuates idiopathic pulmonary fibrosis: An integrated analysis of network pharmacology, molecular docking, and experimental validation. Biochem Biophys Res Commun 2024; 734:150672. [PMID: 39260206 DOI: 10.1016/j.bbrc.2024.150672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
AIMS Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung condition, the cause of which remains unknown and for which no effective therapeutic treatment is currently available. Chlorogenic acid (CGA), a natural polyphenolic compound found in different plants and foods, has emerged as a promising agent due to its anti-inflammatory, antioxidant, and antifibrotic properties. However, the molecular mechanisms underlying the therapeutic effect of CGA in IPF remain unclear. The purpose of this study was to analyze the pharmacological impact and underlying mechanisms of CGA in IPF. MAIN METHODS Using network pharmacology analysis, genes associated with IPF and potential molecular targets of CGA were identified through specialized databases, and a protein-protein interaction (PPI) network was constructed. Molecular docking was performed to accurately select potential therapeutic targets. To investigate the effects of CGA on lung histology and key gene expression, a murine model of bleomycin-induced lung fibrosis was used. KEY FINDINGS Network pharmacology analysis identified 384 were overlapped between CGA and IPF. Key targets including AKT1, TP53, JUN, CASP3, BCL2, MMP9, NFKB1, EGFR, HIF1A, and IL1B were identified. Pathway analysis suggested the involvement of cancer, atherosclerosis, and inflammatory processes. Molecular docking confirmed the stable binding between CGA and targets. CGA regulated the expression mRNA of EGFR, MMP9, AKT1, BCL2 and IL1B and attenuated pulmonary fibrosis in the mouse model. SIGNIFICANCE CGA is a promising multi-target therapeutic agent for IPF, which is supported by its efficacy in reducing fibrosis through the modulation of key pathways. This evidence provides a basis to further investigate CGA as an IPF potential treatment.
Collapse
Affiliation(s)
- Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico.
| | - Jovito Cesar Santos-Álvarez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Alma Aurora Ramírez-Hernández
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Edilburga Reyes-Jiménez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Laura Pérez-Campos Mayoral
- Facultad Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - María de Los Ángeles Romero-Tlalolini
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Cristian Jiménez-Martínez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City, 07738, Mexico
| | - Jaime Arellanes-Robledo
- Laboratorio de Enfermedades Hepáticas, Instituto Nacional de Medicina Genómica - INMEGEN, México City, 14610, Mexico; Dirección Adjunta de Investigación Humanística y Científica, Consejo Nacional de Humanidades, Ciencias y Tecnologías - CONAHCYT, México City, 03940, Mexico
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, C.P. 07360, Mexico
| | - Verónica Rocío Vásquez-Garzón
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico; CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico
| | - Rafael Baltiérrez-Hoyos
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico; CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Sur, San Felipe del Agua, Oaxaca, C.P. 68020, Mexico.
| |
Collapse
|
3
|
Xu Q, Shi MF, Han YF, Liu MY, Liu XB, Ma XN, Feng W, Lin CS, Liu QP. Kunduan Yimu Decoction affected Th17/Treg balance through microRNA-124 to improve rheumatoid arthritis pathology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156129. [PMID: 39427523 DOI: 10.1016/j.phymed.2024.156129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune condition characterized by inflammation and the deterioration of joints. Current treatments often have side effects, highlighting the need for safer options. This study investigates the therapeutic effects of Kunduan Yimu Decoction (KDYMD) on RA, focusing on the role of miR-124 in regulating Th17/Treg differentiation. METHODS PBMCs from RA patients were analyzed before and after KDYMD treatment. RT-qPCR was used to measure the miR-124 expressions. Flow cytometry was used to assess the ratios of Th17 to Treg cells. ELISA was used to quantify the cytokine concentrations. The effects of KDYMD on JAK2/STAT3 signaling were evaluated by western blot analysis. A CIA mouse model was used to validate the in vivo effects of KDYMD. RESULTS MiR-124 expression was significantly upregulated in PBMCs of RA patients after KDYMD treatment. This upregulation was associated with increased Tip60 and Foxp3 expression and decreased RORγt expression. In the cytokine analysis, IL-1, IL-6, and IL-17A were decreased, and IL-10 and TGF- were increased after treatment. Flow cytometry showed a restoration of the Th17/Treg balance, with a decrease in Th17 and an increase in Treg cells. In vivo, KDYMD treatment ameliorated ankle swelling and arthritis index in CIA mice, comparable to methotrexate (MTX). In addition, KDYMD modulated JAK2/STAT3 signaling and enhanced anti-inflammatory responses. CONCLUSIONS KDYMD exerts significant anti-inflammatory effects in RA by upregulating miR-124, which in turn regulates Th17/Treg differentiation and modulates JAK2/STAT3 signaling. A novel mechanism involving miR-124 and immune cell balance suggests KDYMD could be a promising therapeutic agent for RA.
Collapse
Affiliation(s)
- Qiang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China.
| | - Mei-Feng Shi
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu-Feng Han
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Min-Ying Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Xiao-Bao Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Xiao-Na Ma
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Feng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang-Song Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China.
| | - Qing-Ping Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
4
|
Zhang H, Xiong Z, He Y, Su H, Jiao Y. Cimifugin improves intestinal barrier dysfunction by upregulating SIRT1 to regulate the NRF2/HO-1 signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03433-9. [PMID: 39302422 DOI: 10.1007/s00210-024-03433-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Irritable bowel syndrome (IBS) is a prevalent gastrointestinal dysfunction. Cimifugin is an active component of Radix saposhnikoviae which is effective for maintaining intestinal barrier integrity and intestinal function. This study aimed to investigate the treatment efficacy of Cimifugin on intestinal barrier dysfunction and to unveil the relevant mechanism through network pharmacology and experimental verification as well as molecular docking. Through SuperPred and Pubchem databases, the targets of Cimifugin were obtained. The disease targets were screened using Disgenet and GEO databases. With STRING database and Cytoscape software, the analysis of PPI network was performed. In DAVID database, the hub genes of Cimifugin were analyzed using GO and Pathway enrichment analyses. To validate the binding of Cimifugin with core targets, molecular docking was performed. The in vitro cellular model of intestinal barrier was established via the induction of Caco2 cells with LPS. TEER was used to detect epithelial barrier function and permeability was measured using FITC-dextran (FD4). Western blotting was used to measure the expressions of SIRT1, tight junction proteins, and NRF2/HO-1 signaling pathway-related proteins. The fluorescence intensity of ZO-1, Occludin, and Claudin-1 was detected using immunofluorescence staining. ELISA was used to detect the expression levels of inflammatory cytokines. Through the integration of all targets of IBS and Cimifugin, 94 frequent drug-disease-related targets were identified. These targets were enriched in some signaling pathways, like cellular responses to stress, cellular responses to stimuli, and VEGFA-VEGFR2. Ten hub genes including PTGS2, ANPRP, TGFB1, ACACA, SIRT1, NEF2L2, APEX1, IL6, AKT1, and HSP90AB1 were obtained. Cimifugin showed strong affinity with four key genes, including AKT1, SIRT1, IL6, and NFE2L2 (NRF2), which were obtained through the intersection of hug genes with cellular responses to stimuli. In vitro experiments showed that Cimifugin ameliorated LPS-induced intestinal barrier injury in Caco2 cells via upregulating SIRT1 to modulate NRF2/HO-1 signaling pathway. Cimifugin could alleviate intestinal barrier dysfunction in IBS by upregulating SIRT1 to regulate the NRF2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Gastroenterology, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, 528401, Guangdong, China
| | - Zhekun Xiong
- Department of Gastroenterology, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, 528401, Guangdong, China
| | - Yanshan He
- Department of Gastroenterology, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, 528401, Guangdong, China
| | - Huixia Su
- Department of Gastroenterology, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, 528401, Guangdong, China
| | - Yali Jiao
- Xinzhuang Community Health Center, No. 115 Xinjian Road, Minhang District, Shanghai, 201199, China.
| |
Collapse
|
5
|
He Y, Amer HM, Xu Z, Liu L, Wu S, He B, Liu J, Kai G. Exploration of the underlying mechanism of Astragaloside III in attenuating immunosuppression via network pharmacology and vitro/vivo pharmacological validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118235. [PMID: 38648891 DOI: 10.1016/j.jep.2024.118235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus mongholicus Bunge (AM, recorded in http://www.worldfloraonline.org, 2023-08-03) is a kind of medicine food homology plant with a long medicinal history in China. Astragaloside III (AS-III) has immunomodulatory effects and is one of the most active components in AM. However, its underlying mechanism of action is still not fully explained. AIM OF THE STUDY The research was designed to discuss the protective effects of AS-III on immunosuppression and to elucidate its prospective mechanism. MATERIALS AND METHODS Molecular docking methods and network pharmacology analysis were used to comprehensively investigate potential targets and relative pathways for AS-III and immunosuppression. In order to study and verify the pharmacological activity and mechanism of AS-III in alleviating immunosuppression, immunosuppression mouse model induced by cyclophosphamide (CTX) in vivo and macrophage RAW264.7 cell model induced by hypoxia/lipopolysaccharide (LPS) in vitro were used. RESULTS A total of 105 common targets were obtained from the AS-III-related and immunosuppression-related target networks. The results of network pharmacology and molecular docking demonstrate that AS-III may treat immunosuppression through by regulating glucose metabolism-related pathways such as regulation of lipolysis in adipocytes, carbohydrate digestion and absorption, cGMP-PKG signaling pathway, central carbon metabolism in cancer together with HIF-1 pathway. The results of molecular docking showed that AS-III has good binding relationship with LDHA, AKT1 and HIF1A. In CTX-induced immunosuppressive mouse model, AS-III had a significant protective effect on the reduction of body weight, immune organ index and hematological indices. It can also protect immune organs from damage. In addition, AS-III could significantly improve the expression of key proteins involved in energy metabolism and serum inflammatory factors. To further validate the animal results, an initial inflammatory/immune response model of macrophage RAW264.7 cells was constructed through hypoxia and LPS. AS-III improved the immune function of macrophages, reduced the release of NO, TNF-α, IL-1β, PDHK-1, LDH, lactate, HK, PK and GLUT-1, and restored the decrease of ATP caused by hypoxia. Besides, AS-III was also demonstrated that it could inhibit the increase of HIF-1α, PDHK-1 and LDH by adding inhibitors and agonists. CONCLUSIONS In this study, the main targets of AS-III for immunosuppressive therapy were initially analyzed. AS-III was systematically confirmed to attenuates immunosuppressive state through the HIF-1α/PDHK-1 pathway. These findings offer an experimental foundation for the use of AS-III as a potential candidate for the treatment of immunosuppression.
Collapse
Affiliation(s)
- Yining He
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Heba M Amer
- Medicinal and Aromatic Plants Research Dept, National Research Centre, 12622, Dokki, Cairo, Egypt
| | - Zonghui Xu
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lin Liu
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Shujing Wu
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Beihui He
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Junqiu Liu
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Guoyin Kai
- Zhejiang Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 311402, China; The Third Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
6
|
Yang H, Cao J, Li JM, Li C, Zhou WW, Luo JW. Exploration of the molecular mechanism of tea polyphenols against pulmonary hypertension by integrative approach of network pharmacology, molecular docking, and experimental verification. Mol Divers 2024; 28:2603-2616. [PMID: 37486473 DOI: 10.1007/s11030-023-10700-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
Pulmonary hypertension, a common complication of chronic obstructive pulmonary disease, is a major global health concern. Green tea is a popular beverage that is consumed all over the world. Green tea's active ingredients are epicatechin derivatives, also known as "polyphenols," which have anti-carcinogenic, anti-inflammatory, and antioxidant properties. This study aimed to explore the possible mechanism of green tea polyphenols in the treatment of pulmonary hypertension using network pharmacology, molecular docking, and experimental verification. A total of 316 potential green tea polyphenols-related targets were obtained from the PharmMapper, SwissTargetPrediction, and TargetNet databases. A total of 410 pulmonary hypertension-related targets were predicted by the CTD, DisGeNET, pharmkb, and GeneCards databases. Green tea polyphenols-related targets were hit by the 49 targets associated with pulmonary hypertension. AKT1 and HIF1-α were identified through the FDA drugs-target network and PPI network combined with GO functional annotation and KEGG pathway enrichment. Molecular docking results showed that green tea polyphenols had strong binding abilities to AKT1 and HIF1-α. In vitro experiments showed that green tea polyphenols inhibited the proliferation and migration of hypoxia stimulated pulmonary artery smooth muscle cells by decreasing AKT1 phosphorylation and downregulating HIF1α expression. Collectively, green tea polyphenols are promising phytochemicals against pulmonary hypertension.
Collapse
Affiliation(s)
- Huan Yang
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Jun Cao
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Jian-Min Li
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Cheng Li
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Wen-Wu Zhou
- Department of Cardiovascular Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Jin-Wen Luo
- Department of Cardio-Thoracic Surgery, Hunan Children's Hospital, Changsha, 410007, Hunan, China.
| |
Collapse
|
7
|
Hu R, Xue X, Sun X, Mi Y, Wen H, Xi H, Li F, Zheng P, Liu S. Revealing the role of metformin in gastric intestinal metaplasia treatment. Front Pharmacol 2024; 15:1340309. [PMID: 39101145 PMCID: PMC11294171 DOI: 10.3389/fphar.2024.1340309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/01/2024] [Indexed: 08/06/2024] Open
Abstract
Objective Gastric intestinal metaplasia (IM) is a precancerous stage associated with gastric cancer. Despite the observed beneficial effects of metformin on IM, its molecular mechanism remains not fully elucidated. This study aims to reveal the effects and potential mechanisms of metformin in treating IM based on both bioinformatics and in vivo investigations. Methods The seven public databases (GeneCards, DisGeNET, OMIM, SuperPred, Pharm Mapper, Swiss Target Prediction, TargetNet) were used in this work to identify targeted genes related to intestinal metaplasia (IM) and metformin. The shared targeted genes between metformin and IM were further analyzed by network pharmacology, while the interactions in-between were investigated by molecular docking. In parallel, the therapeutic effect of metformin was evaluated in IM mice model, while the core targets and pathways effected by metformin were verified in vivo. Results We screened out 1,751 IM-related genes and 318 metformin-targeted genes, 99 common genes identified in between were visualized by constructing the protein-protein interaction (PPI) network. The top ten core targeted genes were EGFR, MMP9, HIF1A, HSP90AA1, SIRT1, IL2, MAPK8, STAT1, PIK3CA, and ICAM1. The functional enrichment analysis confirmed that carcinogenesis and HIF-1 signaling pathways were primarily involved in the metformin treatment of IM. Based on molecular docking and dynamics, we found metformin affected the function of its targets by inhibiting receptor binding. Furthermore, metformin administration reduced the progression of IM lesions in Atp4a-/- mice model significantly. Notably, metformin enhanced the expression level of MUC5AC, while inhibited the expression level of CDX2. Our results also showed that metformin modulated the expression of core targets in vivo by reducing the activity of NF-κB and the PI3K/AKT/mTOR/HIF-1α signaling pathway. Conclusion This study confirms that metformin improves the efficacy of IM treatment by regulating a complex molecular network. Metformin plays a functional role in inhibiting inflammation/apoptosis-related pathways of further IM progression. Our work provides a molecular foundation for understanding metformin and other guanidine medicines in IM treatment.
Collapse
Affiliation(s)
- Ruoyu Hu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangdong Sun
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huijuan Wen
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huayuan Xi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fuhao Li
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Simeng Liu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Guo P, Li X, Xue Y, Lu Q, Liu Y, Xiong J, Wu Z, Fu S, Ye C, Wang X, Qiu Y. Using network pharmacology and molecular docking to uncover the mechanism by which quercetin alleviates deoxynivalenol-induced porcine intestinal injury. Toxicon 2024; 243:107709. [PMID: 38615996 DOI: 10.1016/j.toxicon.2024.107709] [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: 11/20/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Deoxynivalenol is a widespread feed contaminant that leads to vomit, which results in serious symptom such as increased intestinal permeability and even intestinal mucosal necrosis. Recent studies have reported the role of quercetin in alleviating deoxynivalenol-induced intestinal injury; however, the mechanisms and targets remain unclear. Thus, we aimed to identify the mechanisms of action by using a combination of network pharmacology and molecular docking. We identified 151 quercetin targets, 235 deoxynivalenol targets and 47 porcine intestinal injury targets by searching compound database and PubMed database, among which there were two common targets. The PPI network showed that the key proteins involved are NQO1 and PPAR-γ. The PPI network showed that the key proteins involved were NQO1 and PPARG. GO analysis found that genes were enriched primarily in response to oxidative stress. The PPI network showed that the key proteins involved are NQO1 and PPAR-γ. The genes are enriched primarily in response to oxidative stress. KEGG analysis showed enrichment of the HIF, reactive oxygen species and other signaling pathways. The molecular docking results indicated key binding activity between NQO1-quercetin and PPAR-γ-quercetin. By using network pharmacology, we have revealed the potential molecular mechanisms by which quercetin alleviates deoxynivalenol-induced porcine intestinal injury, which lays the foundation for the development of drugs to treat deoxynivalenol-induced intestinal injury in pigs.
Collapse
Affiliation(s)
- Pu Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xuemin Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yunda Xue
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Qirong Lu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Jianglin Xiong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
9
|
Guo Y, Wu Y, Huang T, Huang D, Zeng Q, Wang Z, Hu Y, Liang P, Chen H, Zheng Z, Liang T, Zhai D, Jiang C, Liu L, Zhu H, Liu Q. Licorice flavonoid ameliorates ethanol-induced gastric ulcer in rats by suppressing apoptosis via PI3K/AKT signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117739. [PMID: 38301986 DOI: 10.1016/j.jep.2024.117739] [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: 11/13/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 02/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Licorice is the dry roots and rhizomes of Glycyrrhiza uralensis Fisch., Glycyrrhiza glabra L. and Glycyrrhiza inflata Bat., which was first recorded in Shengnong's herbal classic. Licorice flavonoid (LF) is the main compound isolated from licorice with an indispensable action in treating gastric ulcer (GU). However, the underlying mechanisms need to be further explored. AIM OF THE STUDY This study aimed to investigate and further elucidate the mechanisms of LF against ethanol-induced GU using an integrated approach. MATERIALS AND METHODS The anti-GU effects of LF were evaluated in an ethanol-induced gastric injury rat model. Then, the metabolomics approach was applied to explore the specific metabolites and metabolic pathways. Next, the network pharmacology combined with metabolomics strategy was employed to predict the targets and pathways of LF for GU. Finally, these predictions were validated by molecular docking, RT-qPCR, and western blotting. RESULTS LF had a positive impact on gastric injury and regulated the expression of GU-related factors. Upon serum metabolomics analysis, 25 metabolic biomarkers of LF in GU treatment were identified, which were primarily involved in amino acid metabolism, carbohydrate metabolism, and other related processes. Subsequently, a "components-targets-metabolites" network was constructed, revealing six key targets (HSP90AA1, AKT1, MAPK1, EGFR, ESR1, PIK3CA) that may be associated with GU treatment. More importantly, KEGG analysis highlighted the importance of the PI3K/AKT pathway including key targets, as a critical route through which LF exerted its anti-GU effects. Molecular docking analyses confirmed that the core components of LF exhibited a strong affinity for key targets. Furthermore, RT-qPCR and western blotting results indicated that LF could reverse the expression of these targets, activate the PI3K/AKT pathway, and ultimately reduce apoptosis. CONCLUSION LF exerted a gastroprotective effect against gastric ulcer induced by ethanol, and the therapeutic mechanism may involve improving metabolism and suppressing apoptosis through the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Yinglin Guo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yufan Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Tairun Huang
- Faculty of Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
| | - Dehao Huang
- Huizhou Jiuhui Pharmaceutical Co., Ltd., Huizhou, 516000, China
| | - Quanfu Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhuxian Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yi Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Peiyi Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hongkai Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zeying Zheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Tao Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Dan Zhai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Cuiping Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Li Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hongxia Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
10
|
Li P, Wang D, Yang X, Liu C, Li X, Zhang X, Liu K, Zhang Y, Zhang M, Wang C, Wang R. Anti-Tumor Activity and Mechanism of Silibinin Based on Network Pharmacology and Experimental Verification. Molecules 2024; 29:1901. [PMID: 38675723 PMCID: PMC11054111 DOI: 10.3390/molecules29081901] [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: 03/24/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Silibinin is a flavonoid compound extracted from the seeds of Silybum marianum (L.) Gaertn. It has the functions of liver protection, blood-lipid reduction and anti-tumor effects. However, the potential molecular mechanism of silibinin against tumors is still unknown. This study aimed to assess the anti-tumor effects of silibinin in adenoid cystic carcinoma (ACC2) cells and Balb/c nude mice, and explore its potential mechanism based on network pharmacology prediction and experimental verification. A total of 347 targets interacting with silibinin were collected, and 75 targets related to the tumor growth process for silibinin were filtrated. Based on the PPI analysis, CASP3, SRC, ESR1, JAK2, PRKACA, HSPA8 and CAT showed stronger interactions with other factors and may be the key targets of silibinin for treating tumors. The predicted target proteins according to network pharmacology were verified using Western blot analysis in ACC2 cells and Balb/c nude mice. In the pharmacological experiment, silibinin was revealed to significantly inhibit viability, proliferation, migration and induce the apoptosis of ACC2 cells in vitro, as well as inhibit the growth and development of tumor tissue in vivo. Western blot analysis showed that silibinin affected the expression of proteins associated with cell proliferation, migration and apoptosis, such as MMP3, JNK, PPARα and JAK. The possible molecular mechanism involved in cancer pathways, PI3K-Akt signaling pathway and viral carcinogenesis pathway via the inhibition of CASP3, MMP3, SRC, MAPK10 and CDK6 and the activation of PPARα and JAK. Overall, our results provided insight into the pharmacological mechanisms of silibinin in the treatment of tumors. These results offer a support for the anti-tumor uses of silibinin.
Collapse
Affiliation(s)
- Peihai Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Dexu Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Xueliang Yang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Changyu Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Xiaobin Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Xuanming Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Kechun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Yun Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Mengqi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Changyun Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Rongchun Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| |
Collapse
|
11
|
Nong Y, Zhou X, Li S, Liu Q, Zhang Y, Liang J, Zhang Y, Liu C. Efficient and fast screening and separation based on computer-aided screening and complex chromatography methods for lipoxygenase inhibitors from Ganoderma lucidum. PHYTOCHEMICAL ANALYSIS : PCA 2024; 35:599-616. [PMID: 38287705 DOI: 10.1002/pca.3316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/31/2024]
Abstract
INTRODUCTION Accurate screening and targeted preparative isolation of active substances from natural medicines have long been technical challenges in natural medicine research. OBJECTIVES This study outlines a new approach for improving the efficiency of natural product preparation, focusing on the rapid and accurate screening of potential active ingredients in Ganoderma lucidum and efficient preparation of lipoxidase inhibitors, with the aim of providing new ideas for the treatment of Alzheimer's disease with G. lucidum. METHODS The medicinal plant G. lucidum was selected through ultrafiltration coupled with liquid chromatography and mass spectrometry (UF-LC-MS) and computer-assisted screening for lipoxygenase (LOX) inhibitors. In addition, the inhibitory effect of the active compounds on LOX was studied using enzymatic reaction kinetics, and the underlying mechanism is discussed. Finally, based on the earlier activity screening guidelines, the identified ligands were isolated and purified through complex chromatography (high-speed countercurrent chromatography and semi-preparative high-performance liquid chromatography). RESULTS Five active ingredients, ganoderic acids A, B, C2, D2, and F, were identified and isolated from G. lucidum. We improved the efficiency and purity of active compound preparation using virtual computer screening and enzyme inhibition assays combined with complex chromatography. CONCLUSION The innovative methods of UF-LC-MS, computer-aided screening, and complex chromatography provide powerful tools for screening and separating LOX inhibitors from complex matrices and provide a favourable platform for the large-scale production of bioactive substances and nutrients.
Collapse
Affiliation(s)
- Yuyu Nong
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Xu Zhou
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Sainan Li
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Qiang Liu
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Yutong Zhang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Jiaqi Liang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Yuchi Zhang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Chunming Liu
- Central Laboratory, Changchun Normal University, Changchun, China
| |
Collapse
|
12
|
Han F, Chen S, Zhang K, Zhang K, Wang M, Wang P. Single-cell transcriptomic sequencing data reveal aberrant DNA methylation in SMAD3 promoter region in tumor-associated fibroblasts affecting molecular mechanism of radiosensitivity in non-small cell lung cancer. J Transl Med 2024; 22:288. [PMID: 38493128 PMCID: PMC10944599 DOI: 10.1186/s12967-024-05057-2] [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: 12/27/2023] [Accepted: 03/02/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVE Non-small cell lung cancer (NSCLC) often exhibits resistance to radiotherapy, posing significant treatment challenges. This study investigates the role of SMAD3 in NSCLC, focusing on its potential in influencing radiosensitivity via the ITGA6/PI3K/Akt pathway. METHODS The study utilized gene expression data from the GEO database to identify differentially expressed genes related to radiotherapy resistance in NSCLC. Using the GSE37745 dataset, prognostic genes were identified through Cox regression and survival analysis. Functional roles of target genes were explored using Gene Set Enrichment Analysis (GSEA) and co-expression analyses. Gene promoter methylation levels were assessed using databases like UALCAN, DNMIVD, and UCSC Xena, while the TISCH database provided insights into the correlation between target genes and CAFs. Experiments included RT-qPCR, Western blot, and immunohistochemistry on NSCLC patient samples, in vitro studies on isolated CAFs cells, and in vivo nude mouse tumor models. RESULTS Fifteen key genes associated with radiotherapy resistance in NSCLC cells were identified. SMAD3 was recognized as an independent prognostic factor for NSCLC, linked to poor patient outcomes. High expression of SMAD3 was correlated with low DNA methylation in its promoter region and was enriched in CAFs. In vitro and in vivo experiments confirmed that SMAD3 promotes radiotherapy resistance by activating the ITGA6/PI3K/Akt signaling pathway. CONCLUSION High expression of SMAD3 in NSCLC tissues, cells, and CAFs is closely associated with poor prognosis and increased radiotherapy resistance. SMAD3 is likely to enhance radiotherapy resistance in NSCLC cells by activating the ITGA6/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Fushi Han
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai, 200065, China
| | - Shuzhen Chen
- Department of Nuclear Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Kangwei Zhang
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai, 200065, China
| | - Kunming Zhang
- Department of Internal Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Meng Wang
- Department of Radiotherapy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Peijun Wang
- Department of Medical Imaging, Tongji Hospital, School of Medicine, Tongji University, No. 389, Xincun Road, Putuo District, Shanghai, 200065, People's Republic of China.
- Institute of Medical Imaging Artificial Intelligence, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
13
|
He Y, Liu F, He M, Long F, Hu D, Chen J, Fang M, Wang Z. Molecular mechanism of resveratrol promoting differentiation of preosteoblastic MC3T3-E1 cells based on network pharmacology and experimental validation. BMC Complement Med Ther 2024; 24:108. [PMID: 38424533 PMCID: PMC10905894 DOI: 10.1186/s12906-024-04396-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/10/2024] [Indexed: 03/02/2024] Open
Abstract
The purpose of this study was to investigate the mechanism by which resveratrol (Res) inhibits apoptosis and promotes proliferation and differentiation of pre-osteoblastic MC3T3-E1 cells, laying the groundwork for the treatment of osteoporosis (OP). The TCMSP database was used to find the gene targets for Res. The GeneCards database acquire the gene targets for OP. After discovering the potential target genes, GO, KEGG, and Reactome enrichment analysis were conducted. Verifying the major proteins involved in apoptosis can bind to Res using molecular docking. CCK8 measured the proliferative activity of mouse pre-osteoblasts in every group following Res intervention. Alkaline phosphatase staining (ALP) and alizarin red staining to measure the ability of osteogenic differentiation. RT-qPCR to determine the expression levels of Runx2 and OPG genes for osteogenic differentiation ability of cells. Western blot to measure the degree of apoptosis-related protein activity in each group following Res intervention. The biological processes investigated for GO of Res therapeutic OP involved in cytokine-mediated signaling pathway, negative regulation of apoptotic process, Aging, extrinsic apoptotic signaling pathway in absence of ligand, according to potential therapeutic target enrichment study. Apoptosis, FoxO signaling pathway, and TNF signaling pathway are the primary KEGG signaling pathways. Recactome pathways are primarily engaged in Programmed Cell Death, Apoptosis, Intrinsic Apoptotic Pathway, and Caspase activation via extrinsic apoptotic signaling pathways. This research established a new approach for Res treatment of OP by demonstrating how Res controls the apoptosis-related proteins TNF, IL6, and CASP3 to suppress osteoblast death and increase osteoclastogenesis.
Collapse
Affiliation(s)
- Yu He
- Department of Spine Surgery, Chengdu Second People's Hospital, No.2, Huatai Road, Chenghua District, Chengdu, 610000, Sichuan, China
| | - Fei Liu
- Department of Spine Surgery, RuiKang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, 530011, Guangxi, China
- Department of orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, China
| | - Mingjuan He
- Department of Spine Surgery, Chengdu Second People's Hospital, No.2, Huatai Road, Chenghua District, Chengdu, 610000, Sichuan, China
| | - Fayu Long
- Department of Spine Surgery, Chengdu Second People's Hospital, No.2, Huatai Road, Chenghua District, Chengdu, 610000, Sichuan, China
| | - Ding Hu
- Department of Spine Surgery, Chengdu Second People's Hospital, No.2, Huatai Road, Chenghua District, Chengdu, 610000, Sichuan, China
| | - Jingwen Chen
- Department of orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, China
| | - Miao Fang
- Department of Spine Surgery, Chengdu Second People's Hospital, No.2, Huatai Road, Chenghua District, Chengdu, 610000, Sichuan, China.
| | - Zhenlong Wang
- Department of orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No.182, Chunhui Road, Longmatan District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
14
|
Jeong E, Hong H, Lee YA, Kim KS. Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development. Int J Mol Sci 2024; 25:2682. [PMID: 38473928 PMCID: PMC11154459 DOI: 10.3390/ijms25052682] [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: 01/22/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by swelling in at least one joint. Owing to an overactive immune response, extra-articular manifestations are observed in certain cases, with interstitial lung disease (ILD) being the most common. Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is characterized by chronic inflammation of the interstitial space, which causes fibrosis and the scarring of lung tissue. Controlling inflammation and pulmonary fibrosis in RA-ILD is important because they are associated with high morbidity and mortality. Pirfenidone and nintedanib are specific drugs against idiopathic pulmonary fibrosis and showed efficacy against RA-ILD in several clinical trials. Immunosuppressants and disease-modifying antirheumatic drugs (DMARDs) with anti-fibrotic effects have also been used to treat RA-ILD. Immunosuppressants moderate the overexpression of cytokines and immune cells to reduce pulmonary damage and slow the progression of fibrosis. DMARDs with mild anti-fibrotic effects target specific fibrotic pathways to regulate fibrogenic cellular activity, extracellular matrix homeostasis, and oxidative stress levels. Therefore, specific medications are required to effectively treat RA-ILD. In this review, the commonly used RA-ILD treatments are discussed based on their molecular mechanisms and clinical trial results. In addition, a computational approach is proposed to develop specific drugs for RA-ILD.
Collapse
Affiliation(s)
- Eunji Jeong
- Department of Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Hyunseok Hong
- Yale College, Yale University, New Haven, CT 06520, USA;
- Department of Clinical Pharmacology and Therapeutics, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon-Ah Lee
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea;
| | - Kyoung-Soo Kim
- Department of Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Clinical Pharmacology and Therapeutics, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- East-West Bone & Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| |
Collapse
|
15
|
Zhang M, Chen G, Chen Y, Sui Y, Zhang Y, Yang W, Yu X. Synthesis, biological activities and mechanism studies of 1,3,4-oxadiazole analogues of petiolide A as anticancer agents. Mol Divers 2024:10.1007/s11030-023-10773-w. [PMID: 38300352 DOI: 10.1007/s11030-023-10773-w] [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: 10/12/2023] [Accepted: 11/17/2023] [Indexed: 02/02/2024]
Abstract
In order to develop new natural product-based anticancer agents, a series of 1,3,4-oxadiazole analogues based on petiolide A were prepared and evaluated for their anticancer activities by MTT method. The structures of all analogues were characterized by various spectral analyses, and B9 was further confirmed by X-ray crystallography. Among all the synthesized compounds, B1 displayed the most promising growth inhibitory effect on colon cancer cells (HCT116) with the IC50 value of 8.53 μM. Flow cytometric analysis exhibited that B1 arrested the cell cycle at G2 phase and induced apoptosis. Additionally, network pharmacology analysis calculated that B1 might target several key proteins, including AKT serine/threonine kinase 1 (AKT1), SRC proto-oncogene, non-receptor tyrosine kinase (SRC) and epidermal growth factor receptor (EGFR). Furthermore, molecular docking study indicated that B1 had potentially high binding affinity to these three target proteins. Given these results, analogue B1 could be deeply developed as potential anticancer agents.
Collapse
Affiliation(s)
- Minjie Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Guifen Chen
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yafang Chen
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yi Sui
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
- Guizhou Joint Laboratory for International Cooperation in Ethnic Medicine (Ministry of Education), Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Wude Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
- Guizhou Joint Laboratory for International Cooperation in Ethnic Medicine (Ministry of Education), Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| | - Xiang Yu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
- Guizhou Joint Laboratory for International Cooperation in Ethnic Medicine (Ministry of Education), Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| |
Collapse
|
16
|
Xing W, Liu G, Zhang Y, Zhang T, Lou H, Fan P. Selective Antitumor Effect and Lower Toxicity of Mitochondrion-Targeting Derivatization of Triptolide. J Med Chem 2024; 67:1093-1114. [PMID: 38169372 DOI: 10.1021/acs.jmedchem.3c01508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Triptolide has a significant antitumor activity, but its toxicity limits its clinical application. As the mitochondrion-targeting strategy showed an advantage in selective antitumor effect based on the higher mitochondrial membrane potential (MMP) in tumor cells than normal cells, the lipophilic cations triphenylphosphonium and E-4-(1H-indol-3-yl vinyl)-N-methylpyridinium iodide (F16) were selected as targeting carriers for structural modification of triptolide. The derivatives bearing F16 generally retained most antitumor activities, overcame its inhibition plateau phenomena, and enhanced its selective antitumor effect in lung cancer. The representative derivative F9 could accumulate in the mitochondria of NCI-H1975 cells, inducing apoptosis and a dose-dependent increase in intracellular reactive oxygen species and reducing MMP. Moreover, no effects were observed in normal cells BEAS-2B. In vivo studies showed that the developmental, renal, and liver toxicities of F9 to zebrafish were significantly lower than those of triptolide. This study provides a promising idea to relieve the toxicity problem of triptolide.
Collapse
Affiliation(s)
- Wenlan Xing
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Guoliang Liu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Yue Zhang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Tao Zhang
- Shandong Provincial Key Laboratory of Neuroprotective Drugs, Shandong Qidu Pharmaceutical Research Institute, Zibo 255400, P. R. China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Peihong Fan
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| |
Collapse
|
17
|
Wan M, Sun S, Di X, Zhao M, Lu F, Zhang Z, Li Y. Icariin improves learning and memory function in Aβ 1-42-induced AD mice through regulation of the BDNF-TrκB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117029. [PMID: 37579923 DOI: 10.1016/j.jep.2023.117029] [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: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedium brevicornu Maxim. is a traditional medicinal Chinese herb that is enriched with flavonoids, which have remarkably high medicinal value. Icariin (ICA) is a marker compound isolated from the total flavonoids of Epimedium brevicornu Maxim. It has been shown to improve Neurodegenerative disease, therefore, ICA is probably a potential drug for treating AD. MATERIALS AND METHODS The 6-8-week-old SPF-class male ICR mice were randomly divided into 8 groups for modeling, and then the mice were administered orally with ICA for 21 days. The behavioral experiments were conducted to evaluate if learning and memory behavior were absent in mice, confirming that infusion of Amyloid β-protein (Aβ)1-42 caused significant memory impairment. The morphological changes and damage of neurons in the mice's brains were observed by HE and Nissl staining. The spinous protrusions (dendritic spines) on neuronal dendrites were investigated by Golgi-Cox staining. The molecular mechanism of ICA was examined by Western Blot. The protein docking of ICA and Donepezil with BDNF were analyzed to determine their interaction. RESULTS The behavioral experimental results showed that in Aβ1-42-induced AD mice, the learning and memory abilities were improved after using ICA. At the same time, the low, medium, and high doses of ICA could reduce the content of Aβ1-42 in the hippocampus of AD mice, repair neuronal damage, enhance synaptic plasticity, as well as increase the expression of BDNF, TrκB, CREB, Akt, GAP43, PSD95, and SYN proteins in the hippocampus of mice. However, the effect with high doses of ICA is more pronounced. The high-dose administration of ICA has the best therapeutic effect on AD mice. After administering the inhibitor k252a, the therapeutic effect of ICA was reversed. The macromolecular docking results of ICA and BDNF protein demonstrated a strong interaction of -7.8 kcal/mol, which indicates that ICA plays a therapeutic role in AD mice by regulating the BDNF-TrκB signaling pathway. CONCLUSIONS The results confirm that ICA can repair neuronal damage, enhance synaptic plasticity, as well as ultimately improve learning and memory impairment through the regulation of the BDNF-TrκB signaling pathway.
Collapse
Affiliation(s)
- Meiyu Wan
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Shengqi Sun
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Xiaoke Di
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Minghui Zhao
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Fengjuan Lu
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Zhifei Zhang
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China
| | - Yang Li
- School of Pharmacy, North China University of Science and Technology, Tangshan, 063210, People's Republic of China.
| |
Collapse
|
18
|
Ren J, Zhao S, Lai J. Triptolide restrains the growth, invasion, stemness, and glycolysis of non-small cell lung cancer cells by PFKFB2-mediated PI3K/AKT pathway. Chem Biol Drug Des 2024; 103:e14450. [PMID: 38230789 DOI: 10.1111/cbdd.14450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/14/2023] [Accepted: 12/31/2023] [Indexed: 01/18/2024]
Abstract
Triptolide (TP) has been found to have anti-tumor effects. However, more potential molecular mechanisms of TP in the progression of non-small cell lung cancer (NSCLC) deserve further investigation. Cell proliferation, apoptosis, invasion, and stemness were detected by cell counting kit 8 assay, EdU assay, flow cytometry, transwell assay, and sphere formation assay. Cell glycolysis was evaluated by corresponding assay kits. 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 (PFKFB2) expression was measured by western blot (WB), qRT-PCR and immunohistochemical staining. PI3K/AKT pathway-related markers were determined by WB. Besides, xenograft tumor model was conducted to evaluate the anti-tumor effect of TP in NSCLC. Our results revealed that TP treatment suppressed NSCLC cell proliferation, invasion, stemness, glycolysis, and enhanced apoptosis. PFKFB2 was upregulated in NSCLC tissues and cells, and its expression was decreased by TP. PFKFB2 knockdown restrained NSCLC cell functions, and its overexpression also eliminated TP-mediated NSCLC cell functions inhibition. TP decreased PFKFB2 expression to inactivate PI3K/AKT pathway. Moreover, PI3K/AKT pathway inhibitor LY294002 also could reverse the promoting effect of PFKFB2 on NSCLC cell functions. In addition, TP suppressed NSCLC tumorigenesis by inhibiting PFKFB2/PI3K/AKT pathway. In conclusion, TP exerted anti-tumor role in NSCLC, which was achieved by reducing PFKFB2 expression to inactivate PI3K/AKT pathway.
Collapse
Affiliation(s)
- Jiankun Ren
- Nursing School, Hebi Polytechnic, Hebi, Henan, China
| | - Songwei Zhao
- Nursing School, Hebi Polytechnic, Hebi, Henan, China
| | - Junyu Lai
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Liu Q, Zhuang S, Li S, Nong Y, Zhang Y, Liang J, Zhang Y, Liu C. Rapid screening, isolation, and activity evaluation of potential xanthine oxidase inhibitors in Polyporus umbellatus and mechanism of action in the treatment of gout. PHYTOCHEMICAL ANALYSIS : PCA 2024; 35:116-134. [PMID: 37798938 DOI: 10.1002/pca.3279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 08/16/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Studies show that Polyporus umbellatus has some pharmacological effects in enhancing immunity and against gout. OBJECTIVES We aimed to establish new techniques for extraction, biological activity screening, and preparation of xanthine oxidase inhibitors (XODIs) from P. umbellatus. METHODS First, the extraction of P. umbellatus was investigated using the back propagation (BP) neural network genetic algorithm mathematical regression model, and the extraction variables were optimised to maximise P. umbellatus yield. Second, XODIs were rapidly screened using ultrafiltration, and the change of XOD activity was tested by enzymatic reaction kinetics experiment to reflect the inhibitory effect of active compounds on XOD. Meanwhile, the potential anti-gout effects of the obtained active substances were verified using molecular docking, molecular dynamics simulations, and network pharmacology analysis. Finally, with activity screening as guide, a high-speed countercurrent chromatography (HSCCC) method combined with consecutive injection and two-phase solvent system preparation using the UNIFAC mathematical model was successfully developed for separation and purification of XODIs, and the XODIs were identified using MS and NMR. RESULTS The results verified that polyporusterone A, polyporusterone B, ergosta-4,6,8(14),22-tetraen-3-one, and ergosta-7,22-dien-3-one of P. umbellatus exhibited high biological affinity towards XOD. Their structures have been further identified by NMR, indicating that the method is effective and applicable for rapid screening and identification of XODIs. CONCLUSION This study provides new ideas for the search for natural XODIs active ingredients, and the study provide valuable support for the further development of functional foods with potential therapeutic benefits.
Collapse
Affiliation(s)
- Qiang Liu
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Siyuan Zhuang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Sainan Li
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Yuyu Nong
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Yutong Zhang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Jiaqi Liang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Yuchi Zhang
- Central Laboratory, Changchun Normal University, Changchun, China
| | - Chunming Liu
- Central Laboratory, Changchun Normal University, Changchun, China
| |
Collapse
|
20
|
Zhang X, Cai W, Yan Y. Agrimonolide Inhibits the Malignant Progression of Non-small Cell Lung Cancer and Induces Ferroptosis through the mTOR Signaling Pathway. Anticancer Agents Med Chem 2024; 24:1371-1381. [PMID: 39129291 DOI: 10.2174/0118715206305421240715042502] [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: 03/15/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Non-Small Cell Lung Cancer (NSCLC), a prevalent type of lung cancer, has a poor prognosis and contributes to a high mortality rate. Agrimonolide, which belongs to the Rosaceae family, possesses various biomedical activities. This study aimed to explore the efficacy and mechanism of agrimonolide in NSCLC. METHODS The viability, proliferation, and tumor-forming ability of A549 cells were detected using the Cell Counting Kit-8 assay (CCK-8) assay, EdU staining, and colony formation assay. The cell cycle was detected using flow cytometry. Cell migration and invasion were detected using wound healing and transwell assays. Western blot was used to detect Epithelial-Mesenchymal Transition (EMT)-, ferroptosis-, and mechanistic targets of rapamycin (mTOR) signaling pathway-related proteins. Lipid peroxidation was detected using the thiobarbituric acid reactive substances (TBARS) assay kit, while lipid Reactive Oxygen Species (ROS) was detected using a BODIPY 581/591 C11 kit. The level of Fe2+ was detected using corresponding assay kits. RESULTS In this study, agrimonolide with varying concentrations (10, 20, and 40 μM) could inhibit the proliferation, induce cycle arrest, suppress metastasis, induce ferroptosis, and block the mTOR signaling pathway in NSCLC cells. To further reveal the mechanism of agrimonolide associated with the mTOR signaling pathway in NSCLC, mTOR agonist MHY1485 (10 μM) was used to pre-treat A549 cells, and functional experiments were conducted again. It was found that the protective effects of AM on NSCLC cells were all partially abolished by MHY1485 pre-treatment. CONCLUSION Agrimonolide inhibited the malignant progression of NSCLC and induced ferroptosis by blocking the mTOR signaling pathway, thus indicating the potential of agrimonolide as a prospective candidate for treating NSCLC.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Department of Pathology, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Wei Cai
- Department of Pathology, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Yiguang Yan
- Department of Cardiothoracic Surgery, Wuxi No.2 People's Hospital (Jiangnan University Medical Center), Wuxi, Jiangsu, 214002, China
| |
Collapse
|
21
|
Li T, Ling J, Du X, Zhang S, Yang Y, Zhang L. Exploring the underlying mechanisms of fisetin in the treatment of hepatic insulin resistance via network pharmacology and in vitro validation. Nutr Metab (Lond) 2023; 20:51. [PMID: 37996895 PMCID: PMC10666360 DOI: 10.1186/s12986-023-00770-z] [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: 06/07/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
OBJECTIVE To characterize potential mechanisms of fisetin on hepatic insulin resistance (IR) using network pharmacology and in vitro validation. METHODS Putative targets of fisetin were retrieved from the Traditional Chinese Medicine Systems Pharmacology database, whereas the potential genes of hepatic IR were obtained from GeneCards database. A protein-protein interaction (PPI) network was constructed according to the intersection targets of fisetin and hepatic IR using the Venn diagram. The biological functions and potential pathways related to genes were determined using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Cell experiments were also conducted to further verify the mechanism of fisetin on hepatic IR. RESULTS A total of 118 potential targets from fisetin were associated with hepatic IR. The areas of nodes and corresponding degree values of TP53, AKT1, TNF, IL6, CASP3, CTNNB1, JUN, SRC, epidermal growth factor receptor (EGFR), and HSP90AA1 were larger and could be easily found in the PPI network. Furthermore, GO analysis revealed that these key targets were significantly involved in multiple biological processes that participated in oxidative stress and serine/threonine kinase activity. KEGG enrichment analysis showed that the PI3K/AKT signaling pathway was a significant pathway involved in hepatic IR. Our in vitro results demonstrated that fisetin treatment increased the expressions of EGFR and IRS in HepG2 and L02 cells under normal or IR conditions. Western blot results revealed that p-AKT/AKT levels were significantly up-regulated, suggesting that fisetin was involved in the PI3K/AKT signaling pathway to regulate insulin signaling. CONCLUSION We explored the pharmacological actions and the potential molecular mechanism of fisetin in treating hepatic IR from a holistic perspective. Our study lays a theoretical foundation for the development of fisetin for type 2 diabetes.
Collapse
Affiliation(s)
- Tian Li
- Metabilic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, China
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China
| | - Junjun Ling
- Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, China
| | - Xingrong Du
- Metabilic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, China
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, China
| | - Siyu Zhang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, 646000, China
| | - Yan Yang
- Chongqing Tongnan NO.1 Middle School, Tongnan, 402660, China
| | - Liang Zhang
- Metabilic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, China.
- Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, China.
| |
Collapse
|
22
|
Fan W, Jiang ZZ, Wan SR. Based on network pharmacology and molecular docking to explore the molecular mechanism of Ginseng and Astragalus decoction against postmenopausal osteoporosis. Medicine (Baltimore) 2023; 102:e35887. [PMID: 37986389 PMCID: PMC10659622 DOI: 10.1097/md.0000000000035887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 11/22/2023] Open
Abstract
Traditional Chinese medicine suggests that Ginseng and Astragalus Decoction (GAD) may effectively treat postmenopausal osteoporosis (PMO). However, the exact mechanism of action for GAD remains unclear. This study aims to utilize network pharmacology and molecular docking technology to explore the potential mechanism of GAD in treating PMO. The main chemical components of GAD were identified by consulting literature and traditional Chinese medicine systems pharmacology database. GeneCards and online mendelian inheritance in man were used to identify PMO disease targets, and Cytoscape 3.8.2 software was used to construct a herb-disease-gene-target network. The intersection of drug targets and disease targets was introduced into the search tool for the retrieval of interacting genes platform to construct a protein-protein interaction network. Additionally, we further conducted gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses, followed by molecular docking between active ingredients and core protein targets. We have identified 59 potential targets related to the treatment of PMO by GAD, along with 33 effective components. Quercetin and kaempferol are the compounds with higher degree. In the protein-protein interaction network, IL6, AKT1, and IL1B are proteins with high degree. The enrichment analysis of gene ontology and KEEG revealed that biological processes involved in treating PMO with GAD mainly include response to hormones, positive regulation of phosphorylation, and regulation of protein homodimerization activity. The signal pathways primarily include Pathways in cancer, PI3K-Akt signaling pathway, and AGE-RAGE signaling pathway. Molecular docking results indicate that kaempferol and quercetin have a high affinity for IL6, AKT1, and IL1B. Our research predicts that IL6, AKT1, and IL1B are highly likely to be potential targets for treating PMO with GAD. PI3K/AKT pathway and AGE-ARGE pathway may play an important role in PMO.
Collapse
Affiliation(s)
- Wei Fan
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, Sichuan, China
| | - Zong-Zhe Jiang
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Sheng-Rong Wan
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
23
|
Gu X, Bao N, Zhang J, Huang G, Zhang X, Zhang Z, Du Y, Meng H, Liu J, Wu P, Wang X, Wang G. Muscone ameliorates myocardial ischemia‒reperfusion injury by promoting myocardial glycolysis. Heliyon 2023; 9:e22154. [PMID: 38045159 PMCID: PMC10692826 DOI: 10.1016/j.heliyon.2023.e22154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 12/05/2023] Open
Abstract
Objective The incidence of acute myocardial infarction (AMI) is increasing yearly. With the use of thrombolysis or percutaneous coronary intervention (PCI), the mortality rate of acute myocardial infarction has been significantly reduced. However, reperfusion can cause additional myocardial injury. There is still a lack of effective drugs to treat I/R injury, and it is urgent to find new therapeutic drugs. Methods In this study, network pharmacology was used to predict potential targets and biological processes involved in Muscone-mediated treatment of acute myocardial infarction. To model ischemia‒reperfusion injury, a hypoxia-reoxygenation model and in vivo ischemia‒reperfusion injury C57BL/6 mice model was constructed. Mice were treated with Muscone i.p. for 4 weeks. We detected the cardiac function on day 28.The expression levels of the apoptotic proteins Caspase-3 and Bax and the anti-apoptotic protein Bcl-2 were detected by immunoblotting after Muscone treatment of AC16 cells and in vivo. Additionally, the gene expression levels of the PUMA and p53 were analyzed by qRT‒PCR. Molecular docking was used to evaluate the binding energy between Muscone and NLRP3-related proteins. Immunoblotting and qRT‒PCR were used to assess the expression levels of NLRP3 signaling pathway-related proteins (NLRP3, ASC, and Caspase-1) and the NLRP3 gene, respectively. Moreover, the extracellular acidification rate of AC16 cells was measured using the Seahorse system to evaluate glycolysis levels after Muscone treatment. The expression of the key glycolytic enzyme PKM2 was analyzed by immunoblotting and qRT‒PCR. Finally, ChIP‒qPCR was performed to determine the levels of histone modifications (H3K4me3, H3K27me3, and H2AK119Ub) in the PKM2 promoter region. Results GO functional enrichment analysis revealed that muscone was involved in regulating the biological processes (BP) of AMI, which mainly included negative regulation of the apoptosis signaling pathway, the response to lipopolysaccharide, and blood pressure regulation. The cellular components (CC) involved in muscone-mediated regulation of AMI mainly included lipid rafts, membrane microdomains, and membrane regions. The molecular functions (MF) involved in muscone-mediated regulation of AMI mainly included oxidoreductase activity, nuclear receptor activity, and transcription factor activity. In vitro results indicated that muscone treatment could inhibit the expression levels of Bax and Caspase-3 in AC16 cells after ischemia‒reperfusion while increasing the expression level of the antiapoptotic protein Bcl-2. Muscone significantly suppressed the transcription levels of p53 and PUMA in AC16 cells. Molecular docking suggested that muscone could bind well with the Cryo-EM structure of NEK7(PDB ID:6NPY). Further investigation of inflammatory pathways revealed that muscone could inhibit the expression level of NLRP3 in AC16 cells and reduce the expression levels of Caspase-1 and Caspase recruitment domain. Fluorescent quantitative PCR experiments showed that muscone significantly inhibited the transcription of NLRP3. Moreover, we found that muscone could enhance the glycolytic efficiency of AC16 cells, which may be related to the increased protein expression of PKM2 in AC16 cells. Fluorescent quantitative PCR showed that muscone could increase the transcription level of PKM2. Chromatin immunoprecipitation assays showed that muscone treatment increased the expression level of H3K4me3 in the PKM2 promoter region and inhibited the levels of H3K27me3 and H2AK119Ub in the PKM2 promoter region. Conclusion Muscone promoted myocardial glycolysis and inhibited NLRP3 pathway activation to improve myocardial ischemia‒reperfusion injury.
Collapse
Affiliation(s)
- Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Neng Bao
- Department of Nephrology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Jing Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Guangyi Huang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Xiaodong Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Zhixuan Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Yinqiang Du
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215008, China
| | - Haoyu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Jiabao Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Peng Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| | - Guangyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, China
| |
Collapse
|
24
|
Li H, Shi W, Shen T, Hui S, Hou M, Wei Z, Qin S, Bai Z, Cao J. Network pharmacology-based strategy for predicting therapy targets of Ecliptae Herba on breast cancer. Medicine (Baltimore) 2023; 102:e35384. [PMID: 37832105 PMCID: PMC10578738 DOI: 10.1097/md.0000000000035384] [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: 04/13/2023] [Accepted: 09/04/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer is a prevalent malignancy affecting women globally, characterized by significant morbidity and mortality rates. Ecliptae Herba is a traditional herbal medicine commonly used in clinical practice, has recently been found to possess antitumor properties. In order to explore the underlying material basis and molecular mechanisms responsible for the anti-breast cancer effects of Ecliptae Herba, we used network pharmacology and experimental verification. UPLC-MS/MS was utilized to identify compounds present in Ecliptae Herba. The active components of Ecliptae Herba and its breast cancer targets were screened using public databases. Hub genes were identified using the STRING and Metascape database. The R software was utilized for visual analysis of GO and KEGG pathways. The affinity of the hub targets for the active ingredients was assessed by molecular docking analysis, which was verified by experimental assessment. A total of 178 targets were obtained from the 10 active components of Ecliptae Herba, while 3431 targets associated with breast cancer were screened. There were 144 intersecting targets between the components and the disease. Targets with a higher degree, namely EGFR and TGFB1, were identified through the hub subnetwork of PPI. GO and KEGG analyses revealed that Ecliptae Herba plays an important role in multiple cancer therapeutic mechanisms. Moreover, molecular docking results showed that the core components had good binding affinity with key targets. Finally, it was confirmed that TGF-β1 might be a potential crucial target of Ecliptae Herba in the treatment of breast cancer by cytological experiments, and the TGF-β1/Smad signaling pathway might be an important pathway for Ecliptae Herba to exert its therapeutic effects. This study elucidated the active ingredients, key targets, and molecular mechanisms of Ecliptae Herba in the treatment of breast cancer, providing a scientific foundation and therapeutic mechanism for the prevention and treatment of breast cancer with Traditional Chinese medicine.
Collapse
Affiliation(s)
- Hui Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wei Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tingming Shen
- Ningde Hospital of Traditional Chinese Medicine, Ningde, China
| | - Siwen Hui
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Manting Hou
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ziying Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuanglin Qin
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junling Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Luoyang Branch of Dongzhimen Hospital Afiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Hu J, Feng Y, Li B, Wang F, Qian Q, Tian W, Niu L, Wang X. Identification of quality markers for Cyanotis arachnoidea and analysis of its physiological mechanism based on chemical pattern recognition, network pharmacology, and experimental validation. PeerJ 2023; 11:e15948. [PMID: 37719108 PMCID: PMC10501370 DOI: 10.7717/peerj.15948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 08/01/2023] [Indexed: 09/19/2023] Open
Abstract
Cyanotis arachnoidea C. B. Clarke is a traditional Chinese medicinal herb that has a limited clinical use in the treatment of diabetes mellitus (DM) in minority areas of Guizhou in China. However, few prior reports are available on the quality control of Cyanotis arachnoidea, and its quality markers and hypoglycemic mechanism are still unclear. The purpose of this study is to explore the quality markers (Q-markers) of Cyanotis arachnoidea and predict its hypoglycemic mechanism. In this study, ultra-high-performance liquid chromatography (UHPLC) fingerprint combined with chemical pattern recognition were performed, and four differential components were screened out as quality markers, including 20-Hydroxyecdysone, 3-O-acetyl-20-hydroxyecdysone, Ajugasterone C, and 2-O-acetyl-20-hydroxyecdysone. Network pharmacology analysis revealed 107 therapeutic target genes of Cyanotis arachnoidea in DM treatment, and the key targets were Akt1, TNF, IL-6, MAPK3, and JUN. The hypoglycemic mode of action of Cyanotis arachnoidea may be mediated by tumor necrosis factor (TNF) signaling, cancer, insulin resistance, and JAK-STAT pathways. Molecular docking analysis disclosed that the foregoing quality markers effectively bound their key target genes. An in vitro experiment conducted on pancreatic islet β-cells indicated that the forenamed active components of Cyanotis arachnoidea had hypoglycemic efficacy by promoting PI3K/Akt and inhibiting MAPK signaling. UHPLC also accurately quantified the quality markers. The identification and analysis of quality markers for Cyanotis arachnoidea is expected to provide references for the establishment of a quality control evaluation system and clarify the material basis and hypoglycemic mechanisms of this traditional Chinese medicine (TCM).
Collapse
Affiliation(s)
- Jingnan Hu
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yu Feng
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Baolin Li
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Fengxia Wang
- Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qi Qian
- Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Traditional Chinese Medicine Formula Granule Engineering & Technology Innovate Center, Shijiazhuang, China
- Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Shijiazhuang, China
| | - Wei Tian
- Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Traditional Chinese Medicine Formula Granule Engineering & Technology Innovate Center, Shijiazhuang, China
- Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Shijiazhuang, China
| | - Liying Niu
- Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Traditional Chinese Medicine Formula Granule Engineering & Technology Innovate Center, Shijiazhuang, China
- Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Shijiazhuang, China
| | - Xinguo Wang
- Hebei University of Chinese Medicine, Shijiazhuang, China
- Hebei Traditional Chinese Medicine Formula Granule Engineering & Technology Innovate Center, Shijiazhuang, China
- Quality Evaluation & Standardization Hebei Province Engineering Research Center of Traditional Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
26
|
Gu JY, Li FJ, Hou CZ, Zhang Y, Bai ZX, Zhang Q. Mechanism of icariin for the treatment of osteoarthritis based on network pharmacology and molecular docking method. Am J Transl Res 2023; 15:5071-5084. [PMID: 37692948 PMCID: PMC10492078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/24/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Icarin's mechanism of action in osteoarthritis (OA) was explored using network pharmacology and the GEO database, and then further validated using molecular docking. METHODS GEO database using network pharmacology identified differential genes in OA based on Icariin's possible targets predicted by pharmmapper database. Combining the differentially expressed genes in OA with the OA-related targets, the overlapping targets were removed. In order to determine what Icariin's core targets are for treating OA, PPI network analysis was performed using OA-related targets and possible Icariin targets. Furthermore, molecular docking was used to verify the chemical's binding to the targets. Final steps included Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of genes and genomes (KEGG) pathway enrichment analysis. Cytoscape was used to construct a network of compound-target-pathway-disease. RESULTS Protein-protein interactions between overlapping targets revealed 151 intersection targets based on a network analysis. The top ten targets with the highest enrichment scores were SRC, MAPK1, HSP90AA1, AKT1, PTPN11, ESR1, EGFR, RhoA, JAK2, and MAPK14. KEGG enrichment analysis showed that the pathways at which Icariin intervention occurs include the OA including FOXO signaling pathway, and estrogen signaling pathway. The GO analysis result showed that various biologic processes such as proteolysis, angiogenesis, innate immune response, and positive regulation of inflammatory response were involved in treatment. Molecular docking analysis confirmed that Icariin could bind well to the targets through intermolecular forces. CONCLUSION With its multi-targeting and multi-pathway characteristics, Icariin is a promising candidate drug for treating OA.
Collapse
Affiliation(s)
- Jin-Yu Gu
- Department of Orthopaedic, The Hospital of Xi YuanBeijing, China
| | - Fa-Jie Li
- Department of Orthopaedic, The Hospital of Wang Jing, China Academy of Chinese Medical SciencesBeijing, China
| | - Cheng-Zhi Hou
- Department of Orthopaedic, The Hospital of Wang Jing, China Academy of Chinese Medical SciencesBeijing, China
| | - Yue Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical SciencesBeijing, China
| | - Zi-Xing Bai
- Department of Orthopaedic, The Hospital of Wang Jing, China Academy of Chinese Medical SciencesBeijing, China
| | - Qing Zhang
- Department of Orthopaedic, The Hospital of Wang Jing, China Academy of Chinese Medical SciencesBeijing, China
| |
Collapse
|
27
|
Fan W, Lan S, Yang Y, Liang J. Network pharmacology prediction and molecular docking-based strategy to discover the potential pharmacological mechanism of Huang-Qi-Gui-Zhi-Wu-Wu decoction against deep vein thrombosis. J Orthop Surg Res 2023; 18:475. [PMID: 37391801 DOI: 10.1186/s13018-023-03948-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Huangqi Guizhi Wuwu decoction (HQGZWWD) has been used to treat and prevent deep vein thrombosis (DVT) in China. However, its potential mechanisms of action remain unclear. This study aimed to utilize network pharmacology and molecular docking technology to elucidate the molecular mechanisms of action of HQGZWWD in DVT. METHODS We identified the main chemical components of HQGZWWD by reviewing the literature and using a Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. We used GeneCards and Online Mendelian Inheritance in Man databases to identify the targets of DVT. Herb-disease-gene-target networks using Cytascape 3.8.2 software; a protein-protein interaction (PPI) network was constructed by combining drug and disease targets on the STRING platform. Additionally, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Finally, molecular docking verification of active components and core protein targets was conducted. RESULTS A total of 64 potential targets related to DVT were identified in HQGZWWD, with 41 active components; quercetin, kaempferol, and beta-sitosterol were the most effective compounds. The PPI network analysis revealed that AKT1, IL1B, and IL6 were the most abundant proteins with the highest degree. GO analysis indicated that DVT treatment with HQGZWWD could involve the response to inorganic substances, positive regulation of phosphorylation, plasma membrane protein complexes, and signaling receptor regulator activity. KEGG analysis revealed that the signaling pathways included pathways in cancer, lipid and atherosclerosis, fluid shear stress and atherosclerosis, and the phosphatidylinositol 3-kinases/protein kinase B(PI3K-Akt) and mitogen-activated protein kinase (MAPK) signaling pathways. The molecular docking results indicated that quercetin, kaempferol, and beta-sitosterol exhibited strong binding affinities for AKT1, IL1B, and IL6. CONCLUSION Our study suggests that AKT1, IL1B, and IL6 are promising targets for treating DVT with HQGZWWD. The active components of HQGZWWD likely responsible for its effectiveness against DVT are quercetin, kaempferol, and beta-sitosterol, they may inhibit platelet activation and endothelial cell apoptosis by regulating the PI3K/Akt and MAPK signaling pathways, slowing the progression of DVT.
Collapse
Affiliation(s)
- Wei Fan
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China
| | - Shuangli Lan
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yunkang Yang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China.
| | - Jie Liang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
- Sichuan Provincial Laboratory of Orthopaedic Engineering, Luzhou, China.
| |
Collapse
|
28
|
Chen L, Yu T, Zhai Y, Nie H, Li X, Ding Y. Luteolin Enhances Transepithelial Sodium Transport in the Lung Alveolar Model: Integrating Network Pharmacology and Mechanism Study. Int J Mol Sci 2023; 24:10122. [PMID: 37373270 DOI: 10.3390/ijms241210122] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Luteolin (Lut), a natural flavonoid compound existing in Perilla frutescens (L.) Britton, has been proven to play a protective role in the following biological aspects: inflammatory, viral, oxidant, and tumor-related. Lut can alleviate acute lung injury (ALI), manifested mainly by preventing the accumulation of inflammation-rich edematous fluid, while the protective actions of Lut on transepithelial ion transport in ALI were seldom researched. We found that Lut could improve the lung appearance/pathological structure in lipopolysaccharide (LPS)-induced mouse ALI models and reduce the wet/dry weight ratio, bronchoalveolar protein, and inflammatory cytokines. Meanwhile, Lut upregulated the expression level of the epithelial sodium channel (ENaC) in both the primary alveolar epithelial type 2 (AT2) cells and three-dimensional (3D) alveolar epithelial organoid model that recapitulated essential structural and functional aspects of the lung. Finally, by analyzing the 84 interaction genes between Lut and ALI/acute respiratory distress syndrome using GO and KEGG enrichment of network pharmacology, we found that the JAK/STAT signaling pathway might be involved in the network. Experimental data by knocking down STAT3 proved that Lut could reduce the phosphorylation of JAK/STAT and enhance the level of SOCS3, which abrogated the inhibition of ENaC expression induced by LPS accordingly. The evidence supported that Lut could attenuate inflammation-related ALI by enhancing transepithelial sodium transport, at least partially, via the JAK/STAT pathway, which may offer a promising therapeutic strategy for edematous lung diseases.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yiman Zhai
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Xin Li
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Center of Forensic Investigation, China Medical University, Shenyang 110122, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| |
Collapse
|
29
|
He X, Zhang W, Cao Q, Li Y, Bao G, Lin T, Bao J, Chang C, Yang C, Yin Y, Xu J, Ren Z, Jin Y, Lu F. Global Downregulation of Penicillin Resistance and Biofilm Formation by MRSA Is Associated with the Interaction between Kaempferol Rhamnosides and Quercetin. Microbiol Spectr 2022; 10:e0278222. [PMID: 36354319 PMCID: PMC9769653 DOI: 10.1128/spectrum.02782-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
The rapid development of methicillin-resistant Staphylococcus aureus (MRSA) drug resistance and the formation of biofilms seriously challenge the clinical application of classic antibiotics. Extracts of the traditional herb Chenopodium ambrosioides L. were found to have strong antibiofilm activity against MRSA, but their mechanism of action remains poorly understood. This study was designed to investigate the antibacterial and antibiofilm activities against MRSA of flavonoids identified from C. ambrosioides L. in combination with classic antibiotics, including ceftazidime, erythromycin, levofloxacin, penicillin G, and vancomycin. Liquid chromatography-mass spectrometry (LC-MS) was used to analyze the nonvolatile chemical compositions. Reverse transcription (RT)-PCR was used to investigate potential multitargets of flavonoids based on global transcriptional responses of virulence and antibiotic resistance. A synergistic antibacterial and biofilm-inhibiting activity of the alcoholic extract of the ear of C. ambrosioides L. in combination with penicillin G was observed against MRSA, which proved to be closely related to the interaction of the main components of kaempferol rhamnosides with quercetin. In regard to the mechanism, the increased sensitivity of MRSA to penicillin G was shown to be related to the downregulation of penicillinase with SarA as a potential drug target, while the antibiofilm activity was mainly related to downregulation of various virulence factors involved in the initial and mature stages of biofilm development, with SarA and/or σB as drug targets. This study provides a theoretical basis for further exploration of the medicinal activity of kaempferol rhamnosides and quercetin and their application in combination with penicillin G against MRSA biofilm infection. IMPORTANCE In this study, the synergistic antibacterial and antibiofilm effects of the traditional herb C. ambrosioides L. and the classic antibiotic penicillin G on MRSA provide a potential strategy to deal with the rapid development of MRSA antibiotic resistance. This study also provides a theoretical basis for further optimizing the combined effect of kaempferol rhamnosides, quercetin, and penicillin G and exploring anti-MRSA biofilm infection research with SarA and σB as drug targets.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Wenwen Zhang
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Qingchao Cao
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yinyue Li
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Guangyu Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Tao Lin
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiaojiao Bao
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Caiwang Chang
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
| | - Changshui Yang
- Department of Pharmacy, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yi Yin
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Jiahui Xu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Zhenyu Ren
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| | - Yingshan Jin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
| | - Feng Lu
- Department of Pathogenic Biology, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Affiliated Hospital of Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
30
|
JIAN L, GUO J, ZHANG Y, LIU J, LIU Y, XU J. Using integrated GC-MS analysis, in vitro experiments, network pharmacology: exploring migao fatty oil active components/mechanisms against coronary heart disease. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.89322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Lina JIAN
- Guizhou University of Traditional Chinese Medicine, China
| | - Jiangtao GUO
- Guizhou University of Traditional Chinese Medicine, China
| | - Yongping ZHANG
- Guizhou University of Traditional Chinese Medicine, China; National Engineering Research Center of Miao's Medicines, China; Guizhou Chinese Medicine Processing and Preparation Engineering Technology Research Center, China
| | - Jie LIU
- Guizhou University of Traditional Chinese Medicine, China; National Engineering Research Center of Miao's Medicines, China; Guizhou Chinese Medicine Processing and Preparation Engineering Technology Research Center, China
| | - Yao LIU
- Guizhou University of Traditional Chinese Medicine, China
| | - Jian XU
- Guizhou University of Traditional Chinese Medicine, China; National Engineering Research Center of Miao's Medicines, China; Guizhou Chinese Medicine Processing and Preparation Engineering Technology Research Center, China
| |
Collapse
|