1
|
Chen R, Sabeel Z, Ying L, Liang Y, Guo R, Hao M, Chen X, Zhang W, Dong J, Liu Y, Yu C, Yang Z. Rhoifolin Suppresses Cell Proliferation and Induces Apoptosis in Hepatocellular Carcinoma Cells In Vitro and In Vivo. Pharmaceuticals (Basel) 2025; 18:79. [PMID: 39861143 PMCID: PMC11768873 DOI: 10.3390/ph18010079] [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: 12/03/2024] [Revised: 12/15/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the most prevalent malignant tumor, ranking fifth in terms of fatality with poor prognosis and a low survival rate. Rhoifolin (ROF), a flavonoid constituent, has previously been shown to suppress the proliferation of breast and pancreatic cancer cells. However, its inhibitory effect on HCC has remained unexplored. Objectives: Exploring the potent inhibitory activities and underlying mechanisms of ROF on HCC cells. Methods: The suppressive effect of ROF on HCC cells were assessed via CCK8 assay, apoptosis assay, cell cycle analysis and xenograft tumor mouse model. Furthermore, quantitative real-time PCR and western blot were applied to analyze the underlying mechanisms of ROF on HCC cells. Results: Firstly, the IC50 values of ROF in HepG2 and HuH7 cells were 373.9 and 288.7 µg/mL at 24 h and 208.9 and 218.0 µg/mL at 48 h, respectively. Moreover, the apoptosis rates of HepG2 and HuH7 cells increased from 6.63% and 6.59% to 17.61% and 21.83% at 24 h and increased from 6.63% and 6.59% to 30.04% and 37.90% at 48 h, respectively. Additionally, ROF induced cell cycle arrest at the S phase in HCC cells. Furthermore, ROF suppressed the tumor growth of HCC cells in vivo without obvious toxicity. Mechanically, ROF facilitated apoptosis by upregulating the expression of PIDD1, CASP8, CASP9, BID, BAX, BIM, and BAK1 in HCC cells. Conclusions: ROF significantly restrains the growth of HCC cells in vitro and in vivo, which could be an effective supplement for HCC therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Changyuan Yu
- Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (R.C.); (Z.S.); (L.Y.); (Y.L.); (R.G.); (M.H.); (X.C.); (W.Z.); (J.D.); (Y.L.)
| | - Zhao Yang
- Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (R.C.); (Z.S.); (L.Y.); (Y.L.); (R.G.); (M.H.); (X.C.); (W.Z.); (J.D.); (Y.L.)
| |
Collapse
|
2
|
Liu Y, Jin Z, Sun D, Zheng J, Xu B, Lan T, Zhao Q, He Y, Li J, Zhang Y, Cui Y. Preparation of monoclonal antibody against rhoifolin and its application in enzyme-linked immunosorbent assay of rhoifolin and diosmin. Talanta 2025; 281:126871. [PMID: 39276572 DOI: 10.1016/j.talanta.2024.126871] [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: 06/07/2024] [Revised: 08/07/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Both rhoifolin and diosmin belong to flavonoids, which are widely present in citrus. Diosmin is not only used in the medical field in the world, but also used as a dietary supplement in the United States. Rhoifolin has a similar structure to diosmin and also exhibits antioxidant and anti-inflammatory properties. In this study, an anti-rhoifolin monoclonal antibody was prepared and an indirect competitive enzyme-linked immunosorbent assay (icELISA) method was established. The half-maximal inhibitory concentration (IC50) of icELISA was determined to be 4.83 ng/mL, and the detection range was 0.97-33.87 ng/mL. The results of UPLC-MS/MS and icELISA generally demonstrate consistency. Moreover, by exploiting the cross-reactivity of the antibody, diosmin in tablets can be detected by icELISA. The results demonstrate that the developed method has good accuracy, reproducibility, and broad application prospects.
Collapse
Affiliation(s)
- Yifan Liu
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Zihui Jin
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Di Sun
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Jiexin Zheng
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Bo Xu
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Tianyu Lan
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Qiyang Zhao
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Yue He
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Jing Li
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China
| | - Yaohai Zhang
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China.
| | - Yongliang Cui
- Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, China; National Citrus Engineering Research Center, Chongqing, 400712, China; Laboratory of Quality & Safety Risk Assessment for Citrus Products (Chongqing), Ministry of Agriculture, Citrus Research Institute, Southwest University, Chongqing, 400712, China.
| |
Collapse
|
3
|
Xia N, Xue H, Li Y, Liu J, Lou Y, Li S, Wang Y, Lu J, Chen X. Potential Mechanisms and Effects of Dai Bai Jie Ethanol Extract in Preventing Acute Alcoholic Liver Injury. Curr Issues Mol Biol 2024; 47:3. [PMID: 39852118 PMCID: PMC11763393 DOI: 10.3390/cimb47010003] [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: 11/23/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025] Open
Abstract
This study investigated the protective effect of Dai Bai Jie (DBJ) extract against acute alcoholic liver injury (AALI) and elucidated its potential mechanism. The total saponin level in the DBJ extracts was measured using vanillin-chloroform acid colorimetry. To observe the preventive and protective effects of DBJ on AML-12 cells in an ethanol environment, the effective components of DBJ were identified. An alcohol-induced AALI mouse model was used to evaluate the efficacy of DBJ against AALI. For this purpose, alcohol dehydrogenase (ADH) and acetaldehyde dehydrogenase (ALDH) levels were assessed, liver function indices and oxidative and inflammatory markers were determined, and histopathological examinations were performed. Mechanistic investigations were conducted using RT-qPCR assays and immunohistochemical analysis to determine the protective effects of DBJ. The samples (DBJ-1, DBJ-2, and DBJ-3) were obtained by extracting DBJ with water, 50% ethanol, and 95% ethanol, yielding total saponin contents of 5.35%, 6.64%, and 11.83%, respectively. DBJ-3 was isolated and purified, and its components were identified by Ultra Performance Liquid Chromatography-Mass Spectrometry (UPLC-MS). DBJ-3 had the greatest effect on cell viability in an ethanol environment. Moreover, DBJ-3 reduced inflammatory infiltration, liver cell degeneration, and hemorrhage, while increasing ADH and ALDH levels in liver tissues. Additionally, DBJ-3 considerably decreased the serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol (TC), and triglyceride (TG) levels. DBJ-3 reduced malondialdehyde (MDA), reactive oxygen species (ROS), and inflammatory factors, such as tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin 6 (IL-6), while increasing superoxide dismutase (SOD) and glutathione S-transferase (GST) activities. Furthermore, DBJ-3 significantly increased alcohol dehydrogenase 1b (ADH1B) and aldehyde dehydrogenase 2 (ALDH2) expression at the gene and protein levels within alcohol metabolism pathways and reduced the nuclear factor kappa-B (NF-κB) gene and protein levels. These findings suggest that DBJ-3 can prevent AALI by enhancing alcohol metabolism via the regulation of ADH1B and ALDH2 and the modulation of the NF-κB pathway to improve antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Niantong Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Hongwei Xue
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Yihang Li
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong 666100, China;
| | - Jia Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Yang Lou
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Shuyang Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Yutian Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Juan Lu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
| | - Xi Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China; (N.X.); (H.X.); (J.L.); (Y.L.); (S.L.); (Y.W.)
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Jinghong 666100, China;
| |
Collapse
|
4
|
Chen M, Chen X, Chen Q, Chu C, Yang S, Wu C, You Y, Hung A, Yang AWH, Sun X, Zhou L, Zhao X, Li H, Liu Y. Potential candidates from a functional food Zanthoxyli Pericarpium (Sichuan pepper) for the management of hyperuricemia: high-through virtual screening, network pharmacology and dynamics simulations. Front Endocrinol (Lausanne) 2024; 15:1436360. [PMID: 39722812 PMCID: PMC11668583 DOI: 10.3389/fendo.2024.1436360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Hyperuricemia (HUA) is a metabolic syndrome caused by purine metabolism disorders. Zanthoxyli Pericarpium (ZP) is a medicinal and food homologous plant, and its ripe peel is used to treat diseases and as a spice for cooking. Some studies have shown that ZP can inhibit the formation of xanthine oxidase and reduce the production of uric acid. Methods Through network pharmacology, ZP's potential targets and mechanisms for HUA treatment were identified. Databases like TCMSP, UniProt, and Swiss Target Prediction were utilized for ZP's active ingredients and targets. HUA-related targets were filtered using GeneCards, Drugbank, and Open Targets. Core targets for ZP's HUA treatment were mapped in a PPI network and analyzed with Cytoscape. GO and KEGG pathway enrichments were conducted on intersected targets via DAVID. Molecular docking and virtual screening were performed to find optimal binding pockets, and ADMET screening assessed compound safety. Molecular dynamics simulations confirmed compound stability in binding sites. Results We identified 81 ZP active ingredient targets, 140 HUA-related targets, and 6 drug targets, with xanthine dehydrogenase (XDH) as the top core target. Molecular docking revealed ZP's active ingredients had strong binding to XDH. Virtual screening via Protein plus identified 48 compounds near the optimal binding pocket, with 2'-methylacetophenone, ledol, beta-sitosterol, and ethyl geranate as the most promising. Molecular dynamics simulations confirmed binding stability, suggesting ZP's potential in HUA prevention and the need for further experimental validation. Conclusion Our study provides foundations for exploring the mechanism of the lowering of uric acid by ZP and developing new products of ZP. The role of ZP in the diet may provide a new dietary strategy for the prevention of HUA, and more experimental studies are needed to confirm our results in the future.
Collapse
Affiliation(s)
- Meilin Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaomei Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Qinghong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chenyang Chu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shuxuan Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chuanghai Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yanting You
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne, VIC, Australia
| | - Angela Wei Hong Yang
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC, Australia
| | - Xiaomin Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lin Zhou
- Endocrinology Department, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Traditional Chinese Medicine Department, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- School of Science, STEM College, RMIT University, Melbourne, VIC, Australia
| | - Yanyan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- Traditional Chinese Medicine Department, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Zhao G, Qi H, Liu M, Zhou T, Chen L, Wu C, Zhang X, Zeng N, Tong Y. Rhoifolin Attenuates Concanavalin A-Induced Autoimmune Hepatitis in Mice via JAKs/STATs Mediated Immune and Apoptotic Processes. ACS OMEGA 2024; 9:43233-43251. [PMID: 39464476 PMCID: PMC11500133 DOI: 10.1021/acsomega.4c07915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/27/2024] [Accepted: 10/03/2024] [Indexed: 10/29/2024]
Abstract
Rhoifolin (ROF) exhibits a diverse range of biological activities, encompassing anticancer, hepatoprotective, antidiabetic, antirheumatic, and antiviral properties. However, the specific protective effects and possible mechanisms of the compound against T-cell-mediated autoimmune hepatitis have not been previously elucidated. In the present study, adult male mice were administered Con A (20 mg/kg, intravenously) for 8 h. In the treated groups, mice were pretreated with ROF daily (20 mg/kg and 40 mg/kg, orally) for 7 days before Con A intoxication. The results showed that ROF significantly decreased serum biochemical indices (ALT, AST, ALP, and LDH) and regulated related oxidative stress indicators (MDA, SOD, and GSH), reduced hepatic necrosis areas and immune cells infiltration, inhibited the release of various inflammatory factors (TNF-α, IFN-γ, IL-2, and IL-17), and improved hepatic tissue apoptosis, thereby alleviating hepatic damage induced by Con A. Additionally, we have also confirmed that ROF efficiently inhibited Th1/Th17 cells polarization via modulation of the JAK2/JAK3/STAT1/STAT3 signaling pathways both in vivo and in vitro. Moreover, the molecular mechanism examination also demonstrated that ROF regulated apoptotic cascade signaling through IL-6/JAK2/STAT1/STAT3 controlling BNIP3 activity in primary hepatocytes. These effects were in good agreement with the bioinformatics analysis of ROF treatment for AIH. In conclusion, our findings provide new insights into the potential use of ROF for AIH therapy, which may result from the specific regulation of the T cell subtype polarization and the apoptosis of liver cells via modulation of the JAKs/STATs signaling pathways.
Collapse
Affiliation(s)
- Ge Zhao
- Department
of Pharmacy, The Affiliated Hospital, Southwest
Medical University, Luzhou, Sichuan 646000, P. R. China
- State Key
Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P. R. China
| | - Hu Qi
- State Key
Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P. R. China
| | - Minghua Liu
- Department
of Pharmacology, School of Pharmacy, Southwest
Medical University, Luzhou, Sichuan 646000, P. R. China
| | - Ting Zhou
- State Key
Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P. R. China
| | - Li Chen
- Department
of Pharmacy, Clinical Medical College and
The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P. R. China
| | - Chunhong Wu
- Information
Centre, Chengdu University, Chengdu, Sichuan 610106, P. R. China
| | - Xiongwei Zhang
- State Key
Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P. R. China
| | - Nan Zeng
- State Key
Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P. R. China
| | - Yue Tong
- Department
of Gastroenterology, Xinqiao Hospital, Third
Military Medical University (Army Medical University), Chongqing 400037, P. R. China
| |
Collapse
|
6
|
Lu JF, Xing SP, Wei X, Yang CX, Zhao GS, Ma XL, Sun XM, Guo HW, Su ZH, Fang B, Lin J, Liu YY, Zhu D. Elucidating the role of 4-hydroxy-2(3H)-benzoxazolone in chronic alcoholic liver disease via transcriptomics and metabolomics. Front Pharmacol 2024; 15:1447560. [PMID: 39323644 PMCID: PMC11422225 DOI: 10.3389/fphar.2024.1447560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Background Chronic alcoholic liver disease (CALD) is a global health problem which includes multiple pathological processes such as immune inflammation and oxidative stress. 4-hydroxy-2(3H)-benzoxazolone (HBOA), an alkaloid isolated from Acanthus ilicifolius L, has been shown to exert hepatoprotective and immunomodulatory effects. However, its effects on CALD remain unclear. This study aimed to investigate the effects and underlying mechanisms of HBOA on CALD. Methods Rats were administered alcohol by gavage continuously for 12 weeks to establish the CALD model, and then treated with HBOA by gavage for 4 weeks. Transcriptomics and metabolomics were used to predict the potential mechanisms of the effects of HBOA on CALD. Liver histology and function, oxidative stress, inflammatory cytokines, and the TLR4/NF-κB pathway components were evaluated. Results HBOA significantly improved alcohol-induced liver injury and steatosis. It decreased the expression levels of pro-inflammatory cytokines (tumour necrosis factor-α [TNF-α], interleukin (IL)-1β, and IL-6), and increased the activities of antioxidant enzymes (superoxide dismutase [SOD], glutathione [GSH], and glutathione peroxidase [GSH-Px]). Western blotting confirmed that HBOA treatment largely diminished NF-κBp65 nuclear translocation. Comprehensive transcriptomics and metabolomics analyses indicated that HBOA regulated the glycerophospholipid metabolism pathway to achieve therapeutic effects in rats with CALD. Conclusion HBOA has a therapeutic effect on rats with CALD. Its mechanism of action mainly affects the glycerophospholipid metabolic pathway to promote lipid metabolism homeostasis by regulating the expression of Etnppl, Gpcpd1, and Pla2g4c. In addition, it may also inhibit the TLR4/NF-κB signaling pathway, thereby reducing the immune-inflammatory response.
Collapse
Affiliation(s)
- Jun-Fei Lu
- Pharmaceutical College, Guangxi Medical University, Nanning, China
- Department of Pharmacy, College and Hospital of Stomatology, Guangxi Medical University, Nanning, China
| | - Shang-Ping Xing
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Xia Wei
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Chun-Xia Yang
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Gen-Shi Zhao
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Xiao-Lin Ma
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Xue-Mei Sun
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Hong-Wei Guo
- Pharmaceutical College, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Geriatric Diseases, Ministry of Education, Guangxi Medical University, Nanning, China
| | - Zhi-Heng Su
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Bin Fang
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Jun Lin
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Yan-Ying Liu
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Dan Zhu
- Pharmaceutical College, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Bioactive Molecules Research and Evaluation, Nanning, China
- Guangxi Beibu Gulf Marine Biomedicine Precision Development and High-value Utilization Engineering Research Center, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Research on Antigeriatric Drugs, Nanning, China
| |
Collapse
|
7
|
Chen P, Huang P, Liang Y, Wang Q, Miao J. The antioxidant peptides from walnut protein hydrolysates and their protective activity against alcoholic injury. Food Funct 2024; 15:5315-5328. [PMID: 38605685 DOI: 10.1039/d4fo00091a] [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: 04/13/2024]
Abstract
In this study, walnut protein was hydrolyzed, separated by ultrafiltration, purified by RP-HPLC, identified by LC-MS/MS, and screened by molecular docking to finally obtain three novel antioxidant peptides HGEPGQQQR (1189.584 Da), VAPFPEVFGK (1089.586 Da) and HNVADPQR (949.473 Da). These three peptides exhibited excellent cellular antioxidant activity (CAA) with EC50 values of 0.0120 mg mL-1, 0.0068 mg mL-1, and 0.0069 mg mL-1, respectively, which were superior to that of the positive control GSH (EC50: 0.0122 mg mL-1). In the ethanol injury model, three antioxidant peptides enhanced the survival of cells treated with ethanol from 47.36% to 62.69%, 57.06% and 71.64%, respectively. Molecular docking results showed that the three antioxidant peptides could effectively bind to Keap1, CYP2E1 and TLR4 proteins. These results suggested that walnut-derived antioxidant peptides could be potential antioxidants and hepatoprotective agents for application in functional foods.
Collapse
Affiliation(s)
- Peihang Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Pantian Huang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Yingyan Liang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Qiaoe Wang
- China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University, Beijing, 100048, China
| | - Jianyin Miao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
8
|
Rezaee A, Rahmanian P, Nemati A, Sohrabifard F, Karimi F, Elahinia A, Ranjbarpazuki A, Lashkarbolouki R, Dezfulian S, Zandieh MA, Salimimoghadam S, Nabavi N, Rashidi M, Taheriazam A, Hashemi M, Hushmandi K. NF-ĸB axis in diabetic neuropathy, cardiomyopathy and nephropathy: A roadmap from molecular intervention to therapeutic strategies. Heliyon 2024; 10:e29871. [PMID: 38707342 PMCID: PMC11066643 DOI: 10.1016/j.heliyon.2024.e29871] [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: 12/03/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic illness defined by elevated blood glucose levels, mediating various tissue alterations, including the dysfunction of vital organs. Diabetes mellitus (DM) can lead to many consequences that specifically affect the brain, heart, and kidneys. These issues are known as neuropathy, cardiomyopathy, and nephropathy, respectively. Inflammation is acknowledged as a pivotal biological mechanism that contributes to the development of various diabetes consequences. NF-κB modulates inflammation and the immune system at the cellular level. Its abnormal regulation has been identified in several clinical situations, including cancer, inflammatory bowel illnesses, cardiovascular diseases, and Diabetes Mellitus (DM). The purpose of this review is to evaluate the potential impact of NF-κB on complications associated with DM. Enhanced NF-κB activity promotes inflammation, resulting in cellular harm and compromised organ performance. Phytochemicals, which are therapeutic molecules, can potentially decline the NF-κB level, therefore alleviating inflammation and the progression of problems correlated with DM. More importantly, the regulation of NF-κB can be influenced by various factors, such as TLR4 in DM. Highlighting these factors can facilitate the development of novel therapies in the future.
Collapse
Affiliation(s)
- Aryan Rezaee
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Amirreza Nemati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farima Sohrabifard
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Fatemeh Karimi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Ali Elahinia
- Department of Clinical Science, Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Ali Ranjbarpazuki
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rozhin Lashkarbolouki
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sadaf Dezfulian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
9
|
Zhang L, Ma Z, Zhang X, Wang J, Tian W, Ren Y, Liu Y, Wang T, Li Y, Liu Y, Shen W, Li T, Liu J, Ma J, Zhang X, Yang S, Wang H. Butyrate alleviates alcoholic liver disease-associated inflammation through macrophage regulation and polarization via the HDAC1/miR-155 axis. Int Immunopharmacol 2024; 131:111852. [PMID: 38492338 DOI: 10.1016/j.intimp.2024.111852] [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: 11/25/2023] [Revised: 02/25/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND We recently found that butyrate could ameliorate inflammation of alcoholic liver disease (ALD) in mice. However, the exact mechanism remains incompletely comprehended. Here, we examined the role of butyrate on ALD-associated inflammation through macrophage (Mψ) regulation and polarization using in vivo and in vitro experiments. METHODS For in vivo experiments, C57BL/6J mice were fed modified Lieber-DeCarli liquid diets supplemented with or without ethanol and sodium butyrate (NaB). After 6 weeks of treatment, mice were euthanized and associated indicators were analyzed. For in vitro experiments, lipopolysaccharide (LPS)-induced inflammatory murine RAW264.7 cells were treated with NaB or miR-155 inhibitor/mimic to verify the anti-inflammatory effect and underlying mechanism. RESULTS The administration of NaB alleviated pathological damage and associated inflammation, including LPS, tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β levels in ALD mice. NaB intervention restored the imbalance of macrophage polarization by inhibiting inducible nitric oxide synthase (iNOS) and elevating arginase-1 (Arg-1). Moreover, NaB reduced histone deacetylase-1 (HDAC1), nuclear factor kappa-B (NF-κB), NOD-like receptor thermal protein domain associated protein 3 (NLRP3), and miR-155 expression in ALD mice, but also increased peroxisome proliferator-activated receptor-γ (PPAR-γ). Thus, MiR-155 was identified as a strong regulator of ALD. To further penetrate the role of miR-155, LPS-stimulated RAW264.7 cells co-cultured with NaB were treated with the specific inhibitor or mimic. Intriguingly, miR-155 was capable of negatively regulated inflammation with NaB intervention by targeting SOCS1, SHIP1, and IRAK-M genes. CONCLUSION Butyrate suppresses the inflammation in mice with ALD by regulating macrophage polarization via the HDAC1/miR-155 axis, which may potentially contribute to the novel therapeutic treatment for the disease.
Collapse
Affiliation(s)
- Lina Zhang
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Zhiguo Ma
- Yinchuan Hospital of Traditional Chinese Medicine, Yinchuan 750004 Ningxia, China
| | - Xiaoxu Zhang
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Jing Wang
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Wenyan Tian
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Yi Ren
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Yajuan Liu
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Ting Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China
| | - Yiwei Li
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China
| | - Yuanyuan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China
| | - Wenke Shen
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China
| | - Ting Li
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Jian Liu
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China
| | - Junbai Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Shaoqi Yang
- General Hospital of Ningxia Medical University (the First Clinical Medical College of Ningxia Medical University), Yinchuan 750004 Ningxia, China.
| | - Hao Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| |
Collapse
|
10
|
Kim DK, Rajan P, Cuong DM, Choi JH, Yoon TH, Go GM, Lee JW, Noh SW, Choi HK, Cho SK. Melosira nummuloides Ethanol Extract Ameliorates Alcohol-Induced Liver Injury by Affecting Metabolic Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8476-8490. [PMID: 38588403 DOI: 10.1021/acs.jafc.3c06261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Melosira nummuloides is a microalga with a nutritionally favorable polyunsaturated fatty acid profile. In the present study, M. nummuloides ethanol extract (MNE) was administered to chronic-binge alcohol-fed mice and alcohol-treated HepG2 cells, and its hepatoprotective effects and underlying mechanisms were investigated. MNE administration reduced triglyceride (TG), total cholesterol (T-CHO), and liver injury markers, including aspartate transaminase (AST) and alanine transaminase (ALT), in the serum of chronic-binge alcohol-fed mice. However, MNE administration increased the levels of phosphorylated adenosine monophosphate-activated protein kinase (P-AMPK/AMPK) and PPARα, which was accompanied by a decrease in SREBP-1; this indicates that MNE can inhibit adipogenesis and improve fatty acid oxidation. Moreover, MNE administration upregulated the expression of antioxidant enzymes, including SOD, NAD(P)H quinone dehydrogenase 1, and GPX, and ameliorated alcohol-induced inflammation by repressing the Akt/NFκB/COX-2 pathway. Metabolomic analysis revealed that MNE treatment modulated many lipid metabolites in alcohol-treated HepG2 cells. Our study findings provide evidence for the efficacy and mechanisms of MNE in ameliorating alcohol-induced liver injury.
Collapse
Affiliation(s)
- Dae Kyeong Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Priyanka Rajan
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Republic of Korea
| | - Do Manh Cuong
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Jae Ho Choi
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon 16499, Republic of Korea
| | - Tae Hyeon Yoon
- College of Applied Life Sciences, SARI, Jeju National University, Jeju 63243, Republic of Korea
| | - Gyung Min Go
- JDKBIO lnc., Jeju-si, Jeju 63023, Republic of Korea
| | - Ji Won Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Soon-Wook Noh
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Somi Kim Cho
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Republic of Korea
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Republic of Korea
- College of Applied Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
11
|
Rabelo ACS, Andrade AKDL, Costa DC. The Role of Oxidative Stress in Alcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis of Preclinical Studies. Nutrients 2024; 16:1174. [PMID: 38674865 PMCID: PMC11055095 DOI: 10.3390/nu16081174] [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: 03/14/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Alcoholic Fatty Liver Disease (AFLD) is characterized by the accumulation of lipids in liver cells owing to the metabolism of ethanol. This process leads to a decrease in the NAD+/NADH ratio and the generation of reactive oxygen species. A systematic review and meta-analysis were conducted to investigate the role of oxidative stress in AFLD. A total of 201 eligible manuscripts were included, which revealed that animals with AFLD exhibited elevated expression of CYP2E1, decreased enzymatic activity of antioxidant enzymes, and reduced levels of the transcription factor Nrf2, which plays a pivotal role in the synthesis of antioxidant enzymes. Furthermore, animals with AFLD exhibited increased levels of lipid peroxidation markers and carbonylated proteins, collectively contributing to a weakened antioxidant defense and increased oxidative damage. The liver damage in AFLD was supported by significantly higher activity of alanine and aspartate aminotransferase enzymes. Moreover, animals with AFLD had increased levels of triacylglycerol in the serum and liver, likely due to reduced fatty acid metabolism caused by decreased PPAR-α expression, which is responsible for fatty acid oxidation, and increased expression of SREBP-1c, which is involved in fatty acid synthesis. With regard to inflammation, animals with AFLD exhibited elevated levels of pro-inflammatory cytokines, including TNF-a, IL-1β, and IL-6. The heightened oxidative stress, along with inflammation, led to an upregulation of cell death markers, such as caspase-3, and an increased Bax/Bcl-2 ratio. Overall, the findings of the review and meta-analysis indicate that ethanol metabolism reduces important markers of antioxidant defense while increasing inflammatory and apoptotic markers, thereby contributing to the development of AFLD.
Collapse
Affiliation(s)
- Ana Carolina Silveira Rabelo
- Postgraduate Program in Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35402-163, Brazil
- Department of Biochemistry, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | | | - Daniela Caldeira Costa
- Postgraduate Program in Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35402-163, Brazil
| |
Collapse
|
12
|
Jing Y, Hu J, Zhang Y, Sun J, Guo J, Zheng Y, Zhang D, Wu L. Structural characterization and preventive effect on alcoholic gastric mucosa and liver injury of a novel polysaccharide from Dendrobium officinale. Nat Prod Res 2024; 38:1140-1147. [PMID: 36239522 DOI: 10.1080/14786419.2022.2134363] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/17/2022] [Accepted: 10/01/2022] [Indexed: 10/17/2022]
Abstract
In this study, a polysaccharide (DOP) with molecular weight of 8.25 × 105 Da and monosaccharide composition of mannose (Man) and glucose (Glc) at a molar ratio of 4.2: 1 was isolated from Dendrobium officinale. The preventive effect on alcoholic gastric mucosa and liver injury of DOP was also investigated. In vitro data exhibited that the IC50 values of 1, 1-diphenyl-2-picrylhydrazy (DPPH) radical scavenging ability and Fe2+ chelating capacity were 2.762 mg/mL and 6.667 mg/mL, respectively. Both the alcoholic gastric mucosal injury (AGMI) and alcoholic liver injury (ALI) animal models were used to investigate the gastrotrophic and hepatoprotective abilities of DOP. After administration of DOP, both gastric mucosal index (TNF-α, IL-6, PGE2, SOD, and MDA) and hepatic indicators (ALT, AST, SOD and MDA) improved compared to non-DOP groups. Histopathological results displayed that the DOP groups improved gastric epithelial defect and inflammatory cell redness caused by AGMI, and decreased vacuolization, hepatocyte necrosis and fibrosis caused by ALI. The results might be related to adjusting inflammatory factors, eliminating free radicals, and inhibiting lipid peroxidation capacities. These results manifested that DOP may be a therapeutic reagent to attenuate alcohol gastric mucosal and liver injury.
Collapse
Affiliation(s)
- Yongshuai Jing
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Jinying Hu
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yameng Zhang
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Junxiang Sun
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Jinyun Guo
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yuguang Zheng
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Danshen Zhang
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Lanfang Wu
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
13
|
Fu J, Deng W, Ge J, Fu S, Li P, Wu H, Wang J, Gao Y, Gao H, Wu T. Sirtuin 1 alleviates alcoholic liver disease by inhibiting HMGB1 acetylation and translocation. PeerJ 2023; 11:e16480. [PMID: 38034869 PMCID: PMC10688304 DOI: 10.7717/peerj.16480] [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: 07/10/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Background Alcoholic liver disease (ALD) encompasses a spectrum of liver disorders resulting from prolonged alcohol consumption and is influenced by factors such as oxidative stress, inflammation, and apoptosis. High Mobility Group Box 1 (HMGB1) plays a pivotal role in ALD due to its involvement in inflammation and immune responses. Another key factor, Sirtuin 1 (SIRT1), an NAD+-dependent deacetylase, is known for its roles in cellular stress responses and metabolic regulation. Despite individual studies on HMGB1 and SIRT1 in ALD, their specific molecular interactions and combined effects on disease advancement remain incompletely understood. Methods Alcohol-induced liver injury (ALI) models were established using HepG2 cells and male C57BL/6 mice. HMGB1 and SIRT1 expressions were assessed at the mRNA and protein levels usingreverse transcription-quantitative polymerase chain reaction, western blot, and immunofluorescence staining. The physical interaction between HMGB1 and SIRT1 was investigated using co-immunoprecipitation and immunofluorescence co-expression analyses. Cellular viability was evaluated using the CCK-8 assay. Results In patients with clinical ALI, HMGB1 mRNA levels were elevated, while SIRT1 expression was reduced, indicating a negative correlation between the two. ALI models were successfully established in cells and mice, as evidenced by increased markers of cellular and liver damage. HMGB1 acetylation and translocation were observed in both ALI cells and mouse models. Treatment with the SIRT1 agonist, SRT1720, reversed the upregulation of HMGB1 acetylation, nuclear translocation, and release in the ethyl alcohol (EtOH) group. Furthermore, SIRT1 significantly attenuated ALI. Importantly, in vivo binding was confirmed between SIRT1 and HMGB1. Conclusions SIRT1 alleviates HMGB1 acetylation and translocation, thereby ameliorating ALI.
Collapse
Affiliation(s)
- Juan Fu
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wei Deng
- Department of Oral and Maxillofacial Surgery, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jun Ge
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shengqi Fu
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Panpan Li
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Huazhi Wu
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jiao Wang
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yi Gao
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Hui Gao
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Tao Wu
- Department of Infectious Diseases, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
14
|
Yang R, Wei L, Wang J, Huang S, Mo P, Chen Q, Zheng P, Chen J, Zhang S, Chen J. Chemical characterization and metabolic profiling of Xiao-Er-An-Shen Decoction by UPLC-QTOF/MS. Front Pharmacol 2023; 14:1219866. [PMID: 38027020 PMCID: PMC10652787 DOI: 10.3389/fphar.2023.1219866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Xiao-Er-An-Shen decoction (XEASD), a TCM formula composed of sixteen Chinese medicinal herbs, has been used to alleviate tic disorders (TD) in clinical practice for many years. However, the chemical basis underlying the therapeutic effects of XEASD in the treatment of TD remains unknown. Purpose: The present study aimed to determine the major chemical components of XEASD and its prototype compounds and metabolites in mice biological samples. Methods: The chemical constituents in XEASD were identified using ultra-high Performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS/MS). Following this, XEASD was orally administered to mice, and samples of plasma, urine, feces, bile, and tissue were collected in order to identify effective compounds for the prevention or treatment of TD. Result: Of the total 184 compounds identified to be discriminated in the XEASD, comprising 44 flavonoids, 26 phenylpropanoids, 16 coumarins, 16 triterpenoids, 14 amino acids, 13 organic acids, 13 alkaloids, 13 ketones, 10 cyclic enol ether terpenes, 7 citrullines, 3 steroids, and 5 anthraquinones, and others. Furthermore, we summarized 54 prototype components and 78 metabolic products of XEASD, measured with biological samples, by estimating metabolic principal components, with four prototype compounds detected in plasma, 58 prototypes discriminated in urine, and 40 prototypes identified in feces. These results indicate that the Oroxylin A glucuronide from Citri reticulatae pericarpium (CRP) is a major compound with potential therapeutic effects identified in brain, while operating positive effect in inhibiting oxidative stress in vitro. Conclusion: In summary, our work delineates the chemical basis underlying the complexity of XEASD, providing insights into the therapeutic and metabolic pathways for TD. Various types of chemicals were explored in XEASD, including flavonoids, phenylpropanoids, coumarins, organic acids, triterpenoid saponins, and so on. This study can promote the further pharmacokinetic and pharmacological evaluation of XEASD.
Collapse
Affiliation(s)
- Ruipei Yang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
- KMHD GeneTech Co., Ltd., Shenzhen, Guangdong, China
| | - Lifang Wei
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jie Wang
- Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Shiying Huang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Pingli Mo
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Qiugu Chen
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Ping Zheng
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jihang Chen
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Shangbin Zhang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jianping Chen
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
15
|
Zhang D, Zhang S, He Z, Chen Y. Cytosine-phosphate-guanine oligodeoxynucleotides alleviate radiation-induced kidney injury in cervical cancer by inhibiting DNA damage and oxidative stress through blockade of PARP1/XRCC1 axis. J Transl Med 2023; 21:679. [PMID: 37773127 PMCID: PMC10541701 DOI: 10.1186/s12967-023-04548-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/20/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Radiotherapy can cause kidney injury in patients with cervical cancer. This study aims to investigate the possible molecular mechanisms by which CpG-ODNs (Cytosine phosphate guanine-oligodeoxynucleotides) regulate the PARP1 (poly (ADP-ribose) polymerase 1)/XRCC1 (X-ray repair cross-complementing 1) signaling axis and its impact on radiation kidney injury (RKI) in cervical cancer radiotherapy. METHODS The GSE90627 dataset related to cervical cancer RKI was obtained from the Gene Expression Omnibus (GEO) database. Bioinformatics databases and R software packages were used to analyze the target genes regulated by CpG-ODNs. A mouse model of RKI was established by subjecting C57BL/6JNifdc mice to X-ray irradiation. Serum blood urea nitrogen (BUN) and creatinine levels were measured using an automated biochemical analyzer. Renal tissue morphology was observed through HE staining, while TUNEL staining was performed to detect apoptosis in renal tubular cells. ELISA was conducted to measure levels of oxidative stress-related factors in mouse serum and cell supernatant. An in vitro cell model of RKI was established using X-ray irradiation on HK-2 cells for mechanism validation. RT-qPCR was performed to determine the relative expression of PARP1 mRNA. Cell proliferation activity was assessed using the CCK-8 assay, and Caspase 3 activity was measured in HK-2 cells. Immunofluorescence was used to determine γH2AX expression. RESULTS Bioinformatics analysis revealed that the downstream targets regulated by CpG-ODNs in cervical cancer RKI were primarily PARP1 and XRCC1. CpG-ODNs may alleviate RKI by inhibiting DNA damage and oxidative stress levels. This resulted in significantly decreased levels of BUN and creatinine in RKI mice, as well as reduced renal tubular and glomerular damage, lower apoptosis rate, decreased DNA damage index (8-OHdG), and increased levels of antioxidant factors associated with oxidative stress (SOD, CAT, GSH, GPx). Among the CpG-ODNs, CpG-ODN2006 had a more pronounced effect. CpG-ODNs mediated the inhibition of PARP1, thereby suppressing DNA damage and oxidative stress response in vitro in HK-2 cells. Additionally, PARP1 promoted the formation of the PARP1 and XRCC1 complex by recruiting XRCC1, which in turn facilitated DNA damage and oxidative stress response in renal tubular cells. Overexpression of either PARP1 or XRCC1 reversed the inhibitory effects of CpG-ODN2006 on DNA damage and oxidative stress in the HK-2 cell model and RKI mouse model. CONCLUSION CpG-ODNs may mitigate cervical cancer RKI by blocking the activation of the PARP1/XRCC1 signaling axis, inhibiting DNA damage and oxidative stress response in renal tubule epithelial cells.
Collapse
Affiliation(s)
- Deyu Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, China
| | - Shitai Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, China
| | - Zheng He
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, China
| | - Ying Chen
- Department of Nephrology, The First Hospital of China Medical University, No. 155 Nanjing Bei Street, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
16
|
Deng G, Liu C, Zhao J, Wang M, Li Y, Yang M, Ye H, Li J, Qin M, Wu C, Shi H, Liao Y, Zhou Z, Zhang S, Lam Yung KK, Gao L. Exocarpium Citri Grandis alleviates the aggravation of NAFLD by mitigating lipid accumulation and iron metabolism disorders. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116559. [PMID: 37116730 DOI: 10.1016/j.jep.2023.116559] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Exocarpium Citri grandis (ECG, Huajuhong in Chinese), the epicarp of C. grandis 'Tomentosa', has been used for hundreds of years as an anti-inflammatory, expectorant, hypoglycemic, and lipid-lowering medication in China. Nevertheless, there have been few papers that have explored the mechanism behind ECG's hypolipidemic characteristics from the perspective of treating nonalcoholic fatty liver disease (NAFLD). AIM OF STUDY The purpose of our study was to confirm the therapeutic and preventative effects of ECG in NAFLD by regulating lipid accumulation and iron metabolism, and to explore the specific mechanism of ECG in enhancing hepatic iron transport and excretion capabilities. STUDY DESIGN We constructed a NAFLD model by feeding male C57BL/6 J mice with a high-fat diet for 12 weeks. Mice were gavaged with ECG beginning in the seventh week of modeling, and three dosage gradients were established: low dose group (2.5 g/kg/d), medium dose group (5 g/kg/d) y, and high dose group (10 g/kg/d) until the end of model construction in week 12. MATERIALS AND METHODS We used network pharmacology to analyze the relationship between ECG and NAFLD. In addition, we constructed a nonalcoholic fatty liver disease model by feeding male C57BL/6 J mice a high-fat diet for 12 weeks. Finally, lipid accumulation, iron accumulation, inflammation and oxidative stress were evaluated by serological index detection, histological detection, immunofluorescent and immunohistochemical staining, and western blotting. RESULTS Network pharmacology confirmed the treatment effect of ECG in NAFLD. Three active components of ECG, including Naringenin, Naringin and Neohesperidin, were detected by UHPLC-HRMS analysis. The results of serum TC, TG, LDL concentration, HE staining, Oil red staining and Nile red staining demonstrated that ECG could improve lipid metabolism disorders. The results of serum iron concentration, liver tissue iron concentration, iron metabolism-related proteins Ferritin light chain, Ferroportin1, Transferrin receptor, and Transferrin demonstrated that ECG improved the iron transport and storage capacities of hepatic cells. CONCLUSIONS Our results demonstrated that ECG relieved liver injury by inhibiting lipid accumulation and iron accumulation in NAFLD.
Collapse
Affiliation(s)
- Guanghui Deng
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Gastroenterology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Chang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jiamin Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Ming Wang
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yunjia Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Menghan Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Haixin Ye
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Junjie Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Mengchen Qin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Chaofeng Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Hao Shi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yuxin Liao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhaoxi Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shiqing Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Ken Kin Lam Yung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region of China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region of China.
| | - Lei Gao
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Gastroenterology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
17
|
Wen S, Ma Z, Tripathi AS. Rhoifolin protects cecal ligation and puncture induced sepsis mice model by regulating inflammatory pathway. Microb Pathog 2023; 180:106112. [PMID: 37059211 DOI: 10.1016/j.micpath.2023.106112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
Sepsis is a systemic infection affects several organs, which needs novel therapy for the management of it, thus protective effect of Rhoifolin was estimated against sepsis. Cecal ligation and puncture (CLP) method was used to induce sepsis and thereafter mice were treated with rhoifolin (10 and 20 mg/kg, p.o.) for one week. Food intake and survival rate was determined sepsis mice, moreover liver function test and cytokines was estimated in the serum of sepsis mice. In the lung tissue homogenate, oxidative stress parameters were determined, histopathological analysis was performed in liver and lung tissue of sepsis mice. Food intake and percentage of survival was improved in rhoifolin treated group than sham group. Level of liver function enzyme and cytokine was reduced significantly in the serum of rhoifolin treated sepsis mice. Treatment with rhoifolin ameliorates the altered oxidative stress parameters, and mRNA expression of Toll-like receptor 4 (TLR-4) in lung tissue of sepsis mice. Histopathological changes were also reverse in rhoifolin treated group than sham group of mice. In conclusion, result of report indicates Rhoifolin treatment reduces oxidative stress and inflammation in CLP induced sepsis mice, as it regulates TLR4/MyD88/NF-κB pathway.
Collapse
Affiliation(s)
- Shun Wen
- Department of Emergency and Intensive Care Unit, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, 215500, China
| | - Zhen Ma
- Department of Intensive Care Unit, Shandong Provincial Third Hospital, Shandong University, 25000, Jinan, China.
| | | |
Collapse
|