1
|
Ye C, Shi D, Zhu Y, Shi P, Zhao N, Sun Z, Zhang Z, Zhang D, Lv Y, Wu W, Yu J, Karimi-Maleh H, Li H, Fu L, Jiang N, Liu J, Lin CT. Graphene electrochemical biosensors combining effervescent solid-phase extraction (ESPE) for rapid, ultrasensitive, and simultaneous determination of DA, AA, and UA. Biosens Bioelectron 2025; 268:116899. [PMID: 39499969 DOI: 10.1016/j.bios.2024.116899] [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: 08/04/2024] [Revised: 10/02/2024] [Accepted: 10/29/2024] [Indexed: 11/25/2024]
Abstract
Simultaneous monitoring of key metabolites like dopamine, ascorbic acid, and uric acid is essential for early disease diagnosis and evaluating treatment. Electrochemical techniques are increasingly used for precise, point-of-care testing (POCT) of these metabolites. Herein, a sample pretreatment method called effervescent solid-phase extraction (ESPE) was proposed for efficient enrichment of trace analytes for electrochemical detection. In an ESPE process, effervescent tablets made of gold nanoparticle-decorated graphene oxide (Au/GO) were first self-dispersed in a test solution to promote the enrichment of analytes on the Au/GO adsorbents, followed by the addition of flocculant effervescent tablets to cause Au/GO sheets to form self-assembled aggregates, which then can be efficiently collected by the foam electrodes. The entire sample pretreatment process operates without external power and takes only 5 min. With the assistance of the ESPE method, our electrochemical sensors achieve an ultralow detection limit for dopamine, ascorbic acid, and uric acid of 80 pM, 1.8 nM, and 460 pM, respectively, which are two to three orders of magnitude lower than the results obtained by the drop casting technique. The enhancement mechanism of our approach is based on increasing the contact probability with analytes through dynamic dispersion of the Au/GO adsorbents, in contrast to the static diffusion mechanism relied on Brownian motion. We also show that combining the ESPE solution kit with a portable micro-electrochemical workstation can attain the same detection level as HPLC in real urine samples. The proposed ESPE approach holds great promise for POCT applications in 2D material-based biosensors.
Collapse
Affiliation(s)
- Chen Ye
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Diwei Shi
- School of Naval Architecture and Maritime, Zhejiang Ocean University, Zhoushan, Zhejiang 316022, PR China
| | - Yangguang Zhu
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Peizheng Shi
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Ningbin Zhao
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Zhuang Sun
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Zhe Zhang
- Shenzhen Refresh Biosensing Technology Co., Ltd., Shenzhen, 518129, PR China
| | - Diming Zhang
- Research Center for Intelligent Sensing Systems, Zhejiang Laboratory, Hangzhou, 311100, PR China
| | - Yaokang Lv
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Wenqi Wu
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangdong, 510260, PR China; Guangdong Key Laboratory of Urology, Guangzhou Medical University, Guangdong, 511495, PR China
| | - Jiancheng Yu
- Institute of Mass Spectrometry, Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Ningbo University, Ningbo, 315211, PR China; Zhenhai Institute of Mass Spectrometry, Ningbo, 315211, PR China; Faculty of Electrical Engineering and Computer Science, Ningbo University, Ningbo, 315211, PR China
| | - Hassan Karimi-Maleh
- School of Resources and Environment, University of Electronic Science and Technology of China, Chengdu, 611731, PR China; School of Engineering, Lebanese American University, Byblos, 1102-2801, Lebanon
| | - He Li
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China
| | - Li Fu
- College of Materials and Environmental Engineering, Hangzhou Dianzi University, Hangzhou 310018, PR China.
| | - Nan Jiang
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China.
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Cheng-Te Lin
- Qianwan Institute, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Marine Materials and Related Technologies, Zhejiang Key Laboratory of Marine Materials and Protective Technologies, Ningbo Institute of Materials Technology and Engineering (NIMTE), Chinese Academy of Sciences, Ningbo, 315201, PR China.
| |
Collapse
|
2
|
Ren L, Dang L, Wang D, Jiang Y, Wang T, Liu Z, Li X, Cui F, Li T, Li J. Natural polysaccharides in the prevention of hyperuricemia: Source, classification, mechanism, application in food industry. Int J Biol Macromol 2025; 286:138421. [PMID: 39645137 DOI: 10.1016/j.ijbiomac.2024.138421] [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: 07/19/2024] [Revised: 11/13/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Hyperuricemia (HUA) is one of the major threats to human health. In recent years, with the gradual increase in the incidence rate of Hua, the prevention and treatment of HUA has attracted more and more attention. Clinical pharmaceutical interventions, such as Allopurinol, Febuxostat, and so on, though effective, are usually accompanied by notable adverse effects. Therefore, alternative therapy with high-safety natural components has received more and more attention from scholars. The natural polysaccharides showed a significant potential in HUA therapy and more and more natural polysaccharides for treating HUA were being obtained. Therefore, in this review, the recent progress on natural polysaccharides in preventing HUA was presented focusing on the sources, classification, and biological activities (oxidative stress, anti-inflammatory, and UA-lowering) of natural polysaccharides. Furthermore, this review explores the mechanisms of action and application. It is beneficial to the development of polysaccharides for natural HUA therapy and the results of this review could offer guidance on preventing the occurrence of HUA in daily life.
Collapse
Affiliation(s)
- Likun Ren
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, Jinzhou, Liaoning 121013, China
| | - Lingling Dang
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, Jinzhou, Liaoning 121013, China
| | - Dangfeng Wang
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, Jinzhou, Liaoning 121013, China
| | - Yang Jiang
- School of Public Health, Dali University, Dali 671000, China
| | - Tian Wang
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, Jinzhou, Liaoning 121013, China
| | - Zhiteng Liu
- Dalian Food Co., Ltd., Jinzhou, Liaoning 121209, China
| | - Xuepeng Li
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, Jinzhou, Liaoning 121013, China
| | - Fangchao Cui
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, Jinzhou, Liaoning 121013, China.
| | - Tingting Li
- Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, Dalian, Liaoning 116029, China.
| | - Jianrong Li
- College of Food Science and Technology, Bohai University, National & Local Joint Engineering Research Center of Storage, Processing and Safety Control Technology for Fresh Agricultural and Aquatic Products, China Light Industry Key Laboratory of Marine Fish Processing, Jinzhou, Liaoning 121013, China
| |
Collapse
|
3
|
Yan X, Feng B, Song H, Wang L, Wang Y, Sun Y, Cai X, Rong Y, Wang X, Wang Y. Identification and mechanistic study of piceatannol as a natural xanthine oxidase inhibitor. Int J Biol Macromol 2024; 293:139231. [PMID: 39732228 DOI: 10.1016/j.ijbiomac.2024.139231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Natural Xanthine oxidase (XOD) inhibitors represent promising therapeutic agents for hyperuricemia (HUA) treatment due to their potent efficacy and favorable safety profiles. This study involved the construction of a comprehensive database of 315 XOD inhibitors and development of 28 machine learning-based QSAR models. The ChemoPy light gradient boosting machine model exhibited the best performance (AUC = 0.9371 and MCC = 0.7423). This model identified three potential XOD inhibitors from the FooDB database: daphnetin, 7-hydroxycoumarin, and piceatannol. Molecular docking and dynamics simulations revealed favorable interactions, with piceatannol showing a remarkable stability through hydrogen bonding and hydrophobic interactions. ADME predictions suggested that all three compounds possess desirable drug-like properties and safety characteristics. Subsequent in vitro enzyme inhibition assays validated computational predictions, with piceatannol exhibiting the strongest inhibitory activity (IC50 = 8.80 ± 0.05 μM). Multispectroscopic analyses revealed that piceatannol-XOD binding was predominantly mediated by hydrogen bonding and van der Waals forces, which induced conformational changes characterized by decreased α-helical content and increased proportions of β-sheets, β-turns, and random coils. This study presents an efficient strategy for the identification of natural XOD inhibitors, elucidates the molecular mechanism of piceatannol-mediated XOD inhibition, and establishes a foundation for its therapeutic application in HUA treatment.
Collapse
Affiliation(s)
- Xinxu Yan
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China; Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji 831100, PR China
| | - Baolong Feng
- Center for Education Technology, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Hongjie Song
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Lili Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Yehui Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Yulin Sun
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Xiaoshuang Cai
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Yating Rong
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China
| | - Xibo Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, Heilongjiang, China.
| | - Yutang Wang
- Institute of Agro-Products Processing Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture, Beijing 100193, China; Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji 831100, PR China.
| |
Collapse
|
4
|
Zhao Z, Zhu B, Li X, Cao J, Qi M, Zhou L, Su B. Microneedle Electrode Patch Modified with Graphene Oxide and Carbon Nanotubes for Continuous Uric Acid Monitoring and Diet Management in Hyperuricemia. ACS APPLIED BIO MATERIALS 2024; 7:8456-8464. [PMID: 39636040 DOI: 10.1021/acsabm.4c01286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Hyperuricemia is a common disorder induced by purine metabolic abnormality, which will further cause chronic kidney disease, cardiovascular disease, and gout. Its main pathological characteristic is the high uric acid (UA) level in the blood, so that the detection of UA is highly important for hyperuricemia diagnosis and therapy. Herein, we report a biocompatible and minimally invasive microneedle electrode patch (MEP) for continuous UA monitoring and diet management in hyperuricemia. The composite of graphene oxide and carboxylated multiwalled carbon nanotubes was modified on the microneedle electrode surface to enhance its sensitivity, selectivity, and stability, thus realizing the continuous detection of UA in the interstitial fluid to accurately predict the UA level in the blood. This further allowed us to study the hypouricemic effect of anthocyanins on the hyperuricemia model mouse. It was found that anthocyanins extracted from blueberry can effectively inhibit the activity of xanthine oxidase to reduce the production of UA. The UA level of hyperuricemia model mice fed with anthocyanins is ∼1.7 fold lower than that of the control group. We believe that this MEP offers enormous promise for continuous UA monitoring and diet management in hyperuricemia.
Collapse
Affiliation(s)
- Ziyi Zhao
- Department of Chemistry, Institute of Analytical Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Boyu Zhu
- Department of Chemistry, Institute of Analytical Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Xinru Li
- Department of Chemistry, Institute of Analytical Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Jiayi Cao
- Department of Chemistry, Institute of Analytical Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Min Qi
- Department of Chemistry, Institute of Analytical Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Lin Zhou
- Department of Chemistry, Institute of Analytical Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Bin Su
- Department of Chemistry, Institute of Analytical Chemistry, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
5
|
Guo W, Zhang J, Deng H, Hao Y, Chen H, Li Y, Zhang W, Zhang M, Chen Y. Structural characterization and anti-hyperuricemic effect of a mannogalactan from Armillariella tabescens mycelium. Int J Biol Macromol 2024; 283:137152. [PMID: 39489252 DOI: 10.1016/j.ijbiomac.2024.137152] [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: 03/13/2024] [Revised: 10/21/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Hyperuricemia and its complications caused by purine metabolism disorder continue to occur, which require effective drug treatment with fewer side effects. This study explored the positive effects of a natural homogeneous polysaccharide (AT-W) from the mycelium of Armillariella tabescens in a mouse hyperuricemia model. Structural characterization showed that the average molecular weight of AT-W is 25.6 kDa, and it is composed of mannose, galactose, arabinose, and fucose, with molar percentages of 11.46:70.9:4.96:12.67. The main backbone of AT-W is composed of partially 3-O-methylated →6)-α-Galp-(1→, and the branches are mainly composed of α-Manp-(1→, →3)-α-Fucp-(1→. In vivo bioactivity evaluation showed AT-W could not only reduce serum uric acid levels by inhibiting the activity of xanthine oxidase, upregulating the expression of ABCG2 and OAT1, and downregulating the expression of URAT1 but also have a significant protective effect on renal damage caused by hyperuricemia. These findings indicate that AT-W has therapeutic potential for hyperuricemia diseases.
Collapse
Affiliation(s)
- Wenhua Guo
- Anhui Key Laboratory for Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Junqiang Zhang
- Tongling Jieya Biologic Technology Stock Co., Ltd., Tongling 244031, Anhui, China
| | - Hailan Deng
- Anhui Key Laboratory for Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Yunbo Hao
- Anhui Key Laboratory for Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Hao Chen
- Anhui Key Laboratory for Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Yong Li
- Anhui Key Laboratory for Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Wenna Zhang
- Anhui Key Laboratory for Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230031, Anhui, China; The Traditional and Western Medicine (TCM)-Integrated Cancer Center of Anhui Medical University, Hefei 230031, Anhui, China.
| | - Yan Chen
- Anhui Key Laboratory for Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China.
| |
Collapse
|
6
|
Wang Z, Wu G, Niu T, Guo Y, Wang C, Wang X, Yu J. Polysaccharide isolated from Dioscorea septemloba improves hyperuricemia and alleviates renal fibrosis through gut-kidney axis in mice. Int J Biol Macromol 2024; 282:137112. [PMID: 39489240 DOI: 10.1016/j.ijbiomac.2024.137112] [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: 08/25/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Hyperuricemia (HUA) is a common metabolic disorder that often accompanies kidney diseases such as tubule damage and renal interstitial fibrosis. The preventive and therapeutic effects of Dioscorea septemloba, an anti-HUA herb, polysaccharide of which was considered as the main active ingredient on HUA, need to be explored. The major polysaccharide component, BXP, was purified from Dioscorea septemloba, with an average molecular weight of 10.432 kDa. Structural analysis inferred that BXP backbone was composed of t-β-D-Glcp-(1→4)-α-D-Glcp-(1 → 4)-α-D-Glcp-(1 → 4, 3)-β-D-Glcp-(1 → , along with the side chain of →1)-α-D-Glcp-(6, 4 → 3, 4)-β-D-Glcp-(1→. The HUA mouse model was further established to clarify the underlying effect of BXP on HUA alleviation. As results shown, BXP decreased serum uric acid by inhibiting XOD and regulating urate transporter expression (GLUT9, OAT3, OAT1, URAT1 and ABCG2) in HUA mice, as well as relieving kidney and liver damage. Moreover, results of microbiome and metabolomics indicated that BXP improved the abundance of gut bacteria and reversed the Lipids-related metabolism disorder caused by HUA. This study indicated that BXP had potential to alleviate HUA and kidney disease through the gut-kidney axis in mice.
Collapse
Affiliation(s)
- Zhenqiang Wang
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Guozhen Wu
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Tong Niu
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Yingjian Guo
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Chuangchuang Wang
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Xiao Wang
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Jinqian Yu
- Shandong Engineering Research Center for Innovation and Application of General Technology for Separation of Natural Products, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China; Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China.
| |
Collapse
|
7
|
Wu C, Wu B, Qu Y, Fu H, Chen Y, Lu Y, Ji S, Ding L, Li Z, Sun Q, Zhang M, Zhang X, Ying B, Zhao F, Zheng X, Qiu Y, Zhang Z, Li K, Zhu Y, Cao Z, Lv Y, Shi X. Blood mercury mediates the associations between fish consumption and serum uric acid levels among Chinese adults: A nationally representative study. ENVIRONMENTAL RESEARCH 2024; 260:119612. [PMID: 39004394 DOI: 10.1016/j.envres.2024.119612] [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: 04/27/2024] [Revised: 06/17/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Fish consumption can increase purine load in human body, and the enrichment of mercury in fish may affect the glomerular filtration function, both resulting in increased serum uric acid (SUA) levels. The data of blood mercury (BHg), fish consumption frequency and SUA levels of 7653 participants aged 18 years or older was from China National Human Biomonitoring (2017-2018). The associations between fish consumption frequency, ln-transformed BHg and SUA levels were explored through weighted multiple linear regressions. The mediating effect of BHg levels between fish consumption frequency and SUA levels was evaluated by mediation analysis. We found that both the fish consumption frequency and BHg were positively associated with SUA levels in both sexes. Compared to participants who had never consumed fish, participants who consumed fish once a week or more had higher SUA levels [β (95% confidence interval, CI): 20.39 (2.16, 38.62) in males; β (95% CI): 10.06 (0.76, 19.37) in females] and ln-transformed BHg [β (95% CI): 0.97 (0.61, 1.34) in males; β (95% CI): 0.84 (0.63, 1.05) in females]. Each 1-unit increase in ln-transformed BHg, the SUA levels rose by 4.78 (95% CI: 0.01, 9.54) μmol/L for males and 3.81 (95% CI: 1.60, 6.03) μmol/L for females. The association between fish consumption with SUA levels was mediated by ln-transformed BHg with the percent mediated of 34.66% in males and 26.58% in females. It revealed that BHg played mediating roles in the elevation of SUA levels caused by fish consumption. This study's findings could promote the government to intervene in mercury pollution in fish, so as to ensure the safety of fish consumption.
Collapse
Affiliation(s)
- Changzi Wu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Bing Wu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yingli Qu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Fu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yue Chen
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yifu Lu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Saisai Ji
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liang Ding
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zheng Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qi Sun
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Miao Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xu Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Bo Ying
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Feng Zhao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xulin Zheng
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yidan Qiu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Big Data in Health Science, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zheng Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Kexin Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ying Zhu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhaojin Cao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yuebin Lv
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Xiaoming Shi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Ye J, Yao J, Xu S, Xiao G, Jia Y, Xie N, Yan J, Ying X, Zhang H. Elucidating the substance basis and pharmacological mechanism of Fufang Qiling granules in modulating xanthine oxidase for intervention in hyperuricemia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118410. [PMID: 38848973 DOI: 10.1016/j.jep.2024.118410] [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: 02/02/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang Qiling granules (FQG), derived from the traditional Qiling Decoction with a longstanding clinical history, is utilized for the treatment of hyperuricemia (HUA). FQG is formulated with a combination of seven Chinese herbs based on the principles of traditional Chinese medicine (TCM) theories. Clinical evidence indicates that FQG exhibits favorable therapeutic effects in reducing uric acid (UA) levels and attenuating renal damage. AIM OF THIS STUDY To elucidate the potential active components and pharmacological mechanism of FQG in the treatment of HUA, and to provide an experimental basis for the development of efficient and low-toxicity TCM for HUA treatment. MATERIALS AND METHODS A HUA rat model induced by potassium oxonate and adenine was established to initially evaluate the hypouricemic effects of FQG. Chemical analyses were conducted using an ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS). Network pharmacology was used to investigate the active components and mechanism of FQG in the treatment of HUA. Potential Xanthine oxidase (XOD) inhibitors were screened from FQG based on ultrafiltration liquid chromatography and mass spectrometry (UF-LC-MS). Molecular docking, surface plasmon resonance (SPR) and circular dichroism (CD) spectroscopy were applied to validate the interactions between the active components and XOD. RESULTS In comparison to the model group, treatment with FQG significantly decreased serum UA, serum creatinine (CREA), serum blood urea nitrogen (BUN), and liver XOD activity. Additionally, the FQG administration notably ameliorated HUA-induced renal injury in rats. Through the pharmacodynamics of the HUA rat models and network pharmacology, it was found that XOD was a key pathway enzyme in UA metabolism. 18 XOD inhibitors were screened from FQG by UF-LC-MS, and 11 compounds with strong affinity were verified by SPR, molecular docking and CD spectroscopy. CONCLUSION In summary, flavonoids, organic acids and saponins may be the active components in FQG that alleviate HUA. The primary mechanism of FQG involves inhibiting XOD enzyme activity in the plasma to reduce UA production, alleviating renal tubular epithelial cell necrosis, tubulointerstitial injury, fibrosis, and urate deposition, ultimately exerting a therapeutic effect on HUA.
Collapse
Affiliation(s)
- Jiamin Ye
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China
| | - Jiangyu Yao
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China
| | - Shaojing Xu
- Research Institute of Chiatai Qingchunbao Pharmaceutical Co., Ltd., Hangzhou, 310030, China
| | - Guyu Xiao
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China
| | - Yuwei Jia
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China
| | - Ningjun Xie
- Research Institute of Chiatai Qingchunbao Pharmaceutical Co., Ltd., Hangzhou, 310030, China
| | - Jizhong Yan
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China
| | - Xuhui Ying
- Research Institute of Chiatai Qingchunbao Pharmaceutical Co., Ltd., Hangzhou, 310030, China.
| | - Hui Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, No. 18, Chaowang Road, Hangzhou, 310014, China.
| |
Collapse
|
9
|
Ullah Z, Yue P, Mao G, Zhang M, Liu P, Wu X, Zhao T, Yang L. A comprehensive review on recent xanthine oxidase inhibitors of dietary based bioactive substances for the treatment of hyperuricemia and gout: Molecular mechanisms and perspective. Int J Biol Macromol 2024; 278:134832. [PMID: 39168219 DOI: 10.1016/j.ijbiomac.2024.134832] [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: 07/11/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024]
Abstract
Hyperuricemia (HUA) has attained a considerable global health concern, related to the development of other metabolic syndromes. Xanthine oxidase (XO), the main enzyme that catalyzes xanthine and hypoxanthine into uric acid (UA), is a key target for drug development against HUA and gout. Available XO inhibitors are effective, but they come with side effects. Recent, research has identified new XO inhibitors from dietary sources such as flavonoids, phenolic acids, stilbenes, alkaloids, polysaccharides, and polypeptides, effectively reducing UA levels. Structural activity studies revealed that -OH groups and their substitutions on the benzene ring of flavonoids, polyphenols, and stilbenes, cyclic rings in alkaloids, and the helical structure of polysaccharides are crucial for XO inhibition. Polypeptide molecular weight, amino acid sequence, hydrophobicity, and binding mode, also play a significant role in XO inhibition. Molecular docking studies show these bioactive components prevent UA formation by interacting with XO substrates via hydrophobic, hydrogen bonds, and π-π interactions. This review explores the potential bioactive substances from dietary resources with XO inhibitory, and UA lowering potentials detailing the molecular mechanisms involved. It also discusses strategies for designing XO inhibitors and assisting pharmaceutical companies in developing safe and effective treatments for HUA and gout.
Collapse
Affiliation(s)
- Zain Ullah
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Panpan Yue
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Guanghua Mao
- School of the Environment and Safety Engineering, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Min Zhang
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Peng Liu
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Xiangyang Wu
- School of the Environment and Safety Engineering, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Ting Zhao
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China.
| | - Liuqing Yang
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China.
| |
Collapse
|
10
|
Jiang Y, Zeng X, Dai H, Luo S, Zhang X. Polygonatum sibiricum polysaccharide regulation of gut microbiota: A viable approach to alleviate cognitive impairment. Int J Biol Macromol 2024; 277:134494. [PMID: 39111476 DOI: 10.1016/j.ijbiomac.2024.134494] [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: 03/19/2024] [Revised: 07/19/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
Polygonatum sibiricum has anti-inflammatory effects and is one of the well-known functional foods. Polygonatum sibiricum polysaccharide (PSP), as a traditional medicinal and food homologous substance, can regulate the balance of intestinal flora and short chain fatty acid levels, reduce intestinal permeability and serum endotoxin levels, and inhibit the activation of astrocytes and microglia. It can significantly alleviate neurological diseases and improve cognitive impairment. Current evidence suggests that bidirectional communication between the central nervous system and the gastrointestinal tract may affect the human nervous system, cognition, and behavior through the gut-brain axis. This article provides a systematic review, detailing the biological activity of PSP, and explores the pathogenesis of gut microbiota signaling in cognitive impairment, providing a promising strategy for improving cognitive impairment.
Collapse
Affiliation(s)
- Yuhan Jiang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Xiaoxiong Zeng
- Department of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Haochen Dai
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Songmei Luo
- Department of Pharmacy, Lishui Central Hospital, Lishui 323000, PR China; The Fifth Hospital Affiliated to Wenzhou Medical University, Lishui 323000, PR China.
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China.
| |
Collapse
|
11
|
Liu X, Zhang L, Lai B, Li J, Zang J, Ma L. Harnessing Protein Hydrolysates and Peptides for Hyperuricemia Management: Insights into Sources, Mechanisms, Techniques, and Future Directions. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18758-18773. [PMID: 39161084 DOI: 10.1021/acs.jafc.4c03605] [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: 08/21/2024]
Abstract
Hyperuricemia (HUA) is a metabolic disorder characterized by an imbalance in uric acid production and excretion, frequently leading to gout and various chronic conditions. Novel bioactive compounds offer effective alternatives for managing HUA, reducing side effects of traditional medications. Recent studies have highlighted the therapeutic potential of protein hydrolysates and peptides in managing HUA. This review focuses on preparing and applying protein hydrolysates to treat HUA and explores peptides for xanthine oxidase inhibition. Particularly, we discuss their origins, enzymatic approaches, and mechanisms of action in detail. The review provides an updated understanding of HUA pathogenesis, current pharmacological interventions, and methodologies for the preparation, purification, identification, and assessment of these compounds. Furthermore, to explore the application of protein hydrolysates and peptides in the food industry, we also address challenges and propose solutions related to the safety, bitterness, oral delivery, and the integration of artificial intelligence in peptide discovery. Bridging traditional pharmacological approaches and innovative dietary interventions, this study paves the way for future research and development in HUA management, contributing to the utilization of proteins from different food sources. In conclusion, protein hydrolysates and peptides show significant promise as safe agents and dietary interventions for preventing and treating HUA.
Collapse
Affiliation(s)
- Xiaoyu Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lei Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Boyin Lai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jingming Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jiachen Zang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Liyan Ma
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
12
|
Li K, Wang Y, Liu W, Zhang C, Xi Y, Zhou Y, Li H, Liu X. Structure-Activity Relationships and Changes in the Inhibition of Xanthine Oxidase by Polyphenols: A Review. Foods 2024; 13:2365. [PMID: 39123556 PMCID: PMC11312107 DOI: 10.3390/foods13152365] [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: 06/20/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Hyperuricemia (HUA), or elevated uric acid in the blood, has become more prevalent in recent years. Polyphenols, which are known to have good inhibitory activity on xanthine oxidoreductase (XOR), are effective in uric acid reduction. In this review, we address the structure-activity relationship of flavonoids that inhibit XOR activity from two perspectives: the key residues of XOR and the structural properties of flavonoids. Flavonoids' inhibitory effect is enhanced by their hydroxyl, methoxy, and planar structures, whereas glycosylation dramatically reduces their activity. The flavonoid structure-activity relationship informed subsequent discussions of the changes that occur in polyphenols' XOR inhibitory activity during their extraction, processing, gastrointestinal digestion, absorption, and interactions. Furthermore, gastrointestinal digestion and heat treatment during processing can boost the inhibition of XOR. Polyphenols with comparable structures may have a synergistic effect, and their synergy with allopurinol thus provides a promising future research direction.
Collapse
Affiliation(s)
- Kexin Li
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Yumei Wang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Wanlu Liu
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | | | - Yu Xi
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Yanv Zhou
- The Product Makers Co., Ltd., Shanghai 200444, China
| | - He Li
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Xinqi Liu
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| |
Collapse
|
13
|
Ferdiansyah MK, Kim YH, Kim KP, Kim MK. Quercetin as the primary xanthine oxidase inhibitor compound in Maclura tricuspidata leaf. Nat Prod Res 2024:1-5. [PMID: 39004844 DOI: 10.1080/14786419.2024.2377317] [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: 03/28/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024]
Abstract
Maclura tricuspidata (MT) leaf demonstrated various health benefits, notably the inhibition of xanthine oxidase (XOD) activity, which is crucial in the management of hyperuricaemia and many diseases related to oxidative stress. This study aimed to identify the primary compound responsible for this inhibitory effect. Through a systematic investigation, MT leaf extracts were subjected to solvent-solvent partitioning using ethyl acetate, n-hexane, n-butanol, and dichloromethane. Further purification involved adsorption and desorption using Amberlite XAD-2 resin, followed by column chromatography on Silica Gel and Sephadex LH-20. The purified compounds were analysed using UPLC-QTOF-MS coupled with NMR spectroscopy. Our findings identified quercetin, a phenolic compound, as the most significant inhibitor of XOD activity in MT leaf, with an IC50 value of 212.92 μg/ml. This is the first report of purifying and identifying a single compound responsible for XOD inhibition in MT.
Collapse
Affiliation(s)
- Mokhammad Khoiron Ferdiansyah
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Young-Hoi Kim
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Kwang Pyo Kim
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
- Department of Agricultural Convergence Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
- Department of FoodTech, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Myung-Kon Kim
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
14
|
AlAseeri AA, Al-Kuraishy HM, Al-Gareeb AI, Ali NH, Alexiou A, Papadakis M, Bahaa MM, Alruwaili M, Batiha GES. The compelling role of allopurinol in hyperuricemia-induced epilepsy: Unrecognized like tears in rain. Brain Res Bull 2024; 213:110973. [PMID: 38723694 DOI: 10.1016/j.brainresbull.2024.110973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/23/2024]
Abstract
Epilepsy is a common neurological disease characterized by the recurrent, paroxysmal, and unprovoked seizures. It has been shown that hyperuricemia enhances and associated with the development and progression of epilepsy through induction of inflammation and oxidative stress. In addition, uric acid is released within the brain and contributes in the development of neuronal hyperexcitability and epileptic seizure. Brain uric acid acts as damage associated molecular pattern (DAMP) activates the immune response and induce the development of neuroinflammation. Therefore, inhibition of xanthine oxidase by allopurinol may reduce hyperuricemia-induced epileptic seizure and associated oxidative stress and inflammation. However, the underlying mechanism of allopurinol in the epilepsy was not fully elucidated. Therefore, this review aims to revise from published articles the link between hyperuricemia and epilepsy, and how allopurinol inhibits the development of epileptic seizure.
Collapse
Affiliation(s)
- Ali Abdullah AlAseeri
- Department of Internal Medicine, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Naif H Ali
- Department of Internal Medicine, Medical College, Najran University, Najran, Saudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens 11741, Greece; Department of Research & Development, AFNP Med, Wien 1030, Austria; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Mostafa M Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511, Egypt
| |
Collapse
|
15
|
Chen Y, Li H, Cai Y, Wang K, Wang Y. Anti-hyperuricemia bioactive peptides: a review on obtaining, activity, and mechanism of action. Food Funct 2024; 15:5714-5736. [PMID: 38752330 DOI: 10.1039/d4fo00760c] [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: 06/05/2024]
Abstract
Hyperuricemia, a disorder of uric acid metabolism, serves as a significant risk factor for conditions such as hypertension, diabetes mellitus, renal failure, and various metabolic syndromes. The main contributors to hyperuricemia include overproduction of uric acid in the liver or impaired excretion in the kidneys. Despite traditional clinical drugs being employed for its treatment, significant health concerns persist. Recently, there has been growing interest in utilizing protein peptides sourced from diverse food origins to mitigate hyperuricemia. This article provides a comprehensive review of bioactive peptides with anti-hyperuricemia properties derived from animals, plants, and their products. We specifically outline the methods for preparing these peptides from food proteins and elucidate their efficacy and mechanisms in combating hyperuricemia, supported by in vitro and in vivo evidence. Uric acid-lowering peptides offer promising prospects due to their safer profile, enhanced efficacy, and improved bioavailability. Therefore, this review underscores significant advancements and contributions in identifying peptides capable of metabolizing purine and/or uric acid, thereby alleviating hyperuricemia. Moreover, it offers a theoretical foundation for the development of functional foods incorporating uric acid-lowering peptides.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing, China
| | - Hongyan Li
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing, China
| | - Yunfei Cai
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing, China
| | - Ke Wang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing, China
- Institute of Modern Fermentation Engineering and Future Foods, Guangxi University, Nanning, 530004, China
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co. Ltd., Rizhao, 276800, China
| | - Yousheng Wang
- Institute of Modern Fermentation Engineering and Future Foods, Guangxi University, Nanning, 530004, China
- College of Light Industry and Food Engineering, Guangxi University, Nanning, 530004, China
| |
Collapse
|
16
|
Yang JJ, Yu H, Wu K, He D, Zhang H, Cui ZX, Chai X, Duan X. Potential Anti-Gouty Arthritis of Citronella Essential Oil and Nutmeg Essential Oil through Reducing Oxidative Stress and Inhibiting PI3K/Akt/mTOR Activation-Induced NLRP3 Activity. Chem Biodivers 2024; 21:e202400448. [PMID: 38498112 DOI: 10.1002/cbdv.202400448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/20/2024]
Abstract
Citronella and Nutmeg are two common spices used for seasoning and medicinal purposes, both of which have significant economic value. This study aimed to investigate whether Citronella essential oil and Nutmeg essential oil (NEO) can ameliorate monosodium urate (MSU)-induced gouty arthritis in rats and the potential mechanisms. The results showed that CEO and NEO reduced swelling and redness at joint sites, inhibited neutrophil infiltration, and limited proinflammatory mediator secretion in mice with MSU-induced gouty arthritis. Based on the results of network pharmacology, molecular docking, and western blotting, CEO and NEO may exert anti-gouty arthritis effects by reducing the expression of reactive oxygen species and oxidative stress and downregulating the phosphorylation of the PI3K/AKT/mTOR signaling pathway, thereby inhibiting the production of the NLRP3 inflammasome and inhibiting the production of inflammatory cytokines. Therefore, these two essential oils show potential for use as adjuvant treatments for gouty arthritis in specific aromatherapy products or food seasonings.
Collapse
Affiliation(s)
- Jin Jin Yang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Hongpeng Yu
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, Guangzhou, 510006, China
| | - Kegang Wu
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, Guangzhou, 510006, China
| | - Dong He
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, Guangzhou, 510006, China
| | - Huadan Zhang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Zheng Xiang Cui
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Xianghua Chai
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
| | - Xuejuan Duan
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, No. 100 Waihuan Xi Road, Panyu District, Guangzhou, 510006, People's Republic of China
| |
Collapse
|
17
|
Dziki-Michalska K, Tajchman K, Kowalik S, Wójcik M. The Levels of Cortisol and Selected Biochemical Parameters in Red Deer Harvested during Stalking Hunts. Animals (Basel) 2024; 14:1108. [PMID: 38612347 PMCID: PMC11010865 DOI: 10.3390/ani14071108] [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: 02/29/2024] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
As a reactive species, the red deer is sensitive to both negative exogenous and endogenous stimuli. An intensive hunting period may have a particularly negative impact on game animals. The aim of this study was to determine the plasma cortisol level and biochemical parameters in 25 wild red deer (Cervus elaphus) harvested during stalking hunts in correlation with the sex and age of the animals. The mean cortisol concentrations in the stags and hinds analyzed in this study were similar (20.2 and 21.5 ng/mL, respectively). Higher HDL cholesterol values were found in the blood of the hinds than in stags (p < 0.05). Similarly, the mean levels of LDL cholesterol, lactate dehydrogenase, and alanine aminotransferase were higher by 21%, 16%, and 42%, respectively, in the blood of the hinds. In contrast, the levels of alkaline phosphatase, bilirubin, and aspartate aminotransferase were higher in the stags (by 30%, 49%, and 36%, respectively). There was a negative correlation of the cortisol concentration with urea and bilirubin and a positive correlation between cortisol and aspartate aminotransferase in the stags (p < 0.05). In turn, a negative correlation was found between the cortisol and urea levels in the hinds (p < 0.05). In summary, the stress caused by stalking hunts and the characteristic behavior of red deer during the mating season had an impact on chosen biochemical parameters. The increased concentration of cortisol resulted in a decrease in the carcass mass, which may lead to the deterioration of the physical condition of animals on hunting grounds.
Collapse
Affiliation(s)
- Katarzyna Dziki-Michalska
- Department of Animal Ethology and Wildlife Management, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland;
| | - Katarzyna Tajchman
- Department of Animal Ethology and Wildlife Management, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland;
| | - Sylwester Kowalik
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12, 20-033 Lublin, Poland
| | - Maciej Wójcik
- Regional Directorate of the State Forests in Lublin, Czechowska 4, 20-950 Lublin, Poland;
| |
Collapse
|
18
|
Zhao X, Cai P, Xiong S, Wei B, Du T, Huang T, Yu Q, Xie M, Xiong T. Lacticaseibacillus rhamnosus NCUH061012 alleviates hyperuricemia via modulating gut microbiota and intestinal metabolites in mice. FOOD BIOSCI 2024; 58:103699. [DOI: 10.1016/j.fbio.2024.103699] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Cunha Matosinhos R, Frézard F, Mendes Silva Araújo S, Magalhães Barbosa A, de Souza IF, de Souza Filho JD, de Souza J, Corrêa Oliveira Bahia AP, Ietta F, Magnani A, Saúde-Guimarães DA. Development and characterization of liposomal formulations containing sesquiterpene lactones for the treatment of chronic gout. Sci Rep 2024; 14:6991. [PMID: 38523180 PMCID: PMC10961318 DOI: 10.1038/s41598-024-57663-1] [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: 12/30/2023] [Accepted: 03/20/2024] [Indexed: 03/26/2024] Open
Abstract
Gout and hyperuricemia are characterized by high uric acid levels, and their treatment involves medications that have adverse effects. In this study, we evaluated oral liposomal formulations with eremantholide C and goyazensolide as a novel approach to reduce the toxicity associated with these substances while maintaining their anti-hyperuricemic activity. We characterized the formulations and evaluated them based on encapsulation efficiency and stability over 12 months and under simulated physiological environments. We determined the toxicity of the liposomal formulations in Caco-2 cells and the anti-hyperuricemic activity in rats. The formulations exhibited nanometric size, a narrow size distribution, and a negative zeta potential, indicating their stability and uniformity. The efficient encapsulation of the sesquiterpene lactones within the liposomes emphasizes their potential for sustained release and therapeutic efficacy. Stability evaluation revealed a small decrease in the eremantholide C concentration and a remarkable stability in the goyazensolide concentration. In Caco-2 cells, the liposomes did not exert toxicity, but did exhibit an antiproliferative effect. In vivo assays demonstrated that the liposomes reduced serum uric acid levels. Our study represents an advancement in gout and hyperuricemia treatment. The liposomal formulations effectively reduced the toxicity associated with the sesquiterpene lactones while maintaining their therapeutic effects.
Collapse
Affiliation(s)
- Rafaela Cunha Matosinhos
- Laboratório de Plantas Medicinais (LAPLAMED), Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Frédéric Frézard
- Laboratório de Biofísica e Sistemas Nanoestruturados (LabNano), Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Sabrina Mendes Silva Araújo
- Laboratório de Biofísica e Sistemas Nanoestruturados (LabNano), Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Andressa Magalhães Barbosa
- Laboratório de Plantas Medicinais (LAPLAMED), Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Isabela Fernanda de Souza
- Laboratório de Plantas Medicinais (LAPLAMED), Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - José Dias de Souza Filho
- Laboratório Multiusuário de Caracterização de Moléculas (LMCM), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Jacqueline de Souza
- Laboratório de Controle de Qualidade de Insumos, Fármacos e Medicamentos (LCQ), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Ana Paula Corrêa Oliveira Bahia
- Laboratório de Biofísica e Sistemas Nanoestruturados (LabNano), Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Francesca Ietta
- Dipartimento Scienze della Vita, Università degli Studi di Siena, 53100, Siena, Tuscany, Italy
| | - Agnese Magnani
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, 53100, Siena, Tuscany, Italy
| | - Dênia Antunes Saúde-Guimarães
- Laboratório de Plantas Medicinais (LAPLAMED), Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil.
| |
Collapse
|
20
|
Wang Y, Jia X, An S, Yin W, Huang J, Jiang X. Nanozyme-Based Regulation of Cellular Metabolism and Their Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301810. [PMID: 37017586 DOI: 10.1002/adma.202301810] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Metabolism is the sum of the enzyme-dependent chemical reactions, which produces energy in catabolic process and synthesizes biomass in anabolic process, exhibiting high similarity in mammalian cell, microbial cell, and plant cell. Consequently, the loss or gain of metabolic enzyme activity greatly affects cellular metabolism. Nanozymes, as emerging enzyme mimics with diverse functions and adjustable catalytic activities, have shown attractive potential for metabolic regulation. Although the basic metabolic tasks are highly similar for the cells from different species, the concrete metabolic pathway varies with the intracellular structure of different species. Here, the basic metabolism in living organisms is described and the similarities and differences in the metabolic pathways among mammalian, microbial, and plant cells and the regulation mechanism are discussed. The recent progress on regulation of cellular metabolism mainly including nutrient uptake and utilization, energy production, and the accompanied redox reactions by different kinds of oxidoreductases and their applications in the field of disease therapy, antimicrobial therapy, and sustainable agriculture is systematically reviewed. Furthermore, the prospects and challenges of nanozymes in regulating cell metabolism are also discussed, which broaden their application scenarios.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xiaodan Jia
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Shangjie An
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Wenbo Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Jiahao Huang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Xiue Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
21
|
Peng X, Liu K, Hu X, Gong D, Zhang G. Hesperitin-Copper(II) Complex Regulates the NLRP3 Pathway and Attenuates Hyperuricemia and Renal Inflammation. Foods 2024; 13:591. [PMID: 38397567 PMCID: PMC10888018 DOI: 10.3390/foods13040591] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Hyperuricaemia (HUA) is a disorder of purine metabolism in the body. We previously synthesized a hesperitin (Hsp)-Cu(II) complex and found that the complex possessed strong uric acid (UA)-reducing activity in vitro. In this study we further explored the complex's UA-lowering and nephroprotective effects in vivo. METHODS A mouse with HUA was used to investigate the complex's hypouricemic and nephroprotective effects via biochemical analysis, RT-PCR, and Western blot. RESULTS Hsp-Cu(II) complex markedly decreased the serum UA level and restored kidney tissue damage to normal in HUA mice. Meanwhile, the complex inhibited liver adenosine deaminase (ADA) and xanthine oxidase (XO) activities to reduce UA synthesis and modulated the protein expression of urate transporters to promote UA excretion. Hsp-Cu(II) treatment significantly suppressed oxidative stress and inflammatory in the kidney, reduced the contents of cytokines and inhibited the activation of the nucleotide-binding oligomerization domain (NOD)-like receptor thermal protein domain associated protein 3 (NLRP3) inflammatory pathway. CONCLUSIONS Hsp-Cu(II) complex reduced serum UA and protected kidneys from renal inflammatory damage and oxidative stress by modulating the NLRP3 pathway. Hsp-Cu(II) complex may be a promising dietary supplement or nutraceutical for the therapy of hyperuricemia.
Collapse
Affiliation(s)
- Xi Peng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.P.); (K.L.); (X.H.); (D.G.)
- Department of Biological Engineering, Jiangxi Biotech Vocational College, Nanchang 330200, China
| | - Kai Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.P.); (K.L.); (X.H.); (D.G.)
| | - Xing Hu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.P.); (K.L.); (X.H.); (D.G.)
| | - Deming Gong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.P.); (K.L.); (X.H.); (D.G.)
| | - Guowen Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.P.); (K.L.); (X.H.); (D.G.)
| |
Collapse
|
22
|
Yu D, Du J, He P, Wang N, Li L, Liu Y, Yang C, Xu H, Li Y. Identification of natural xanthine oxidase inhibitors: Virtual screening, anti-xanthine oxidase activity, and interaction mechanism. Int J Biol Macromol 2024; 259:129286. [PMID: 38216015 DOI: 10.1016/j.ijbiomac.2024.129286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/14/2024]
Abstract
Xanthine oxidase (XO) is a crucial target for hyperuricemia treatment(s). Naturally occurred XO inhibitors with minimal toxicity and high efficacy have attracted researchers' attention. With the goal of quickly identifying natural XO inhibitors, an integrated computational screening strategy was constructed by molecular docking and calculating the free energy of binding. Twenty-seven hits were achieved from a database containing 19,377 natural molecules. This includes fourteen known XO inhibitors and four firstly-reported inhibitors (isolicoflavonol, 5,7-dihydroxycoumarin, parvifolol D and clauszoline M, IC50 < 40 μM). Iolicoflavonol (hit 8, IC50 = 8.45 ± 0.68 μM) and 5,7-dihydroxycoumarin (hit 25, IC50 = 10.91 ± 0.71 μM) displayed the great potency as mixed-type inhibitors. Docking study and molecular dynamics simulation revealed that both hits could interact with XO's primarily active site residues ARG880, MOS1328, and ASN768 of XO. Fluorescence spectroscopy studies showed that hit 8 bound to the active cavity region of XO, causing changes in XO's conformation and hydrophobicity. Hits 8 and 25 exhibit favorable Absorption, Distribution, Metabolism, and Excretion (ADME) properties. Additionally, no cytotoxicity against human liver cells was observed at their median inhibition concentrations against XO. Therefore, the present study offers isolicoflavonol and 5,7-dihydroxycoumarin with the potential to be disease-modifying agents for hyperuricemia.
Collapse
Affiliation(s)
- Dehong Yu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Jiana Du
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Pei He
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Na Wang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Lizi Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Yi Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Can Yang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Haiqi Xu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Yanfang Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
23
|
Ma N, Cai S, Sun Y, Chu C. Chinese Sumac ( Rhus chinensis Mill.) Fruits Prevent Hyperuricemia and Uric Acid Nephropathy in Mice Fed a High-Purine Yeast Diet. Nutrients 2024; 16:184. [PMID: 38257077 PMCID: PMC10819650 DOI: 10.3390/nu16020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Hyperuricemia (HUA) is a prevalent chronic disease, characterized by excessive blood uric acid levels, that poses a significant health risk. In this study, the preventive effects and potential mechanisms of ethanol extracts from Chinese sumac (Rhus chinensis Mill.) fruits on HUA and uric acid nephropathy were comprehensively investigated. The results demonstrated a significant reduction in uric acid levels in hyperuricemia mice after treatment with Chinese sumac fruit extract, especially in the high-dose group, where the blood uric acid level decreased by 39.56%. Visual diagrams of the kidneys and hematoxylin and eosin (H&E)-stained sections showed the extract's effectiveness in protecting against kidney damage caused by excessive uric acid. Further investigation into its mechanism revealed that the extract prevents and treats hyperuricemia by decreasing uric acid production, enhancing uric acid excretion, and mitigating the oxidative stress and inflammatory reactions induced by excessive uric acid in the kidneys. Specifically, the extract markedly decreased xanthine oxidase (XOD) levels and expression in the liver, elevated the expression of uric acid transporters ABCG2, and lowered the expression of uric acid reabsorption proteins URAT1 and SLC2A9. Simultaneously, it significantly elevated the levels of endogenous antioxidant enzymes (SOD and GSH) while reducing the level of malondialdehyde (MDA). Furthermore, the expression of uric-acid-related proteins NLRP3, ACS, and Caspase-3 and the levels of IL-1β and IL-6 were significantly reduced. The experimental results confirm that Chinese sumac fruit extract can improve HUA and uric acid nephropathy in mice fed a high-purine yeast diet. This finding establishes a theoretical foundation for developing Chinese sumac fruit as a functional food or medicine for preventing and treating HUA.
Collapse
Affiliation(s)
| | | | | | - Chuanqi Chu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (N.M.); (S.C.)
| |
Collapse
|
24
|
Lu C, Wang X, Ma J, Wang M, Liu W, Wang G, Ding Y, Lin Z, Li Y. Chemical substances and their activities in sea cucumber Apostichopus japonicus: A review. Arch Pharm (Weinheim) 2024; 357:e2300427. [PMID: 37853667 DOI: 10.1002/ardp.202300427] [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: 08/03/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023]
Abstract
Apostichopus japonicus, also known as Stichopus japonicus, with medicinal and food homologous figures, is a globally recognized precious ingredient with extremely high nutritional value. There is no relevant review available through literature search, so this article selects the research articles through the keywords "sea cucumber" and "Apostichopus japonicus (Stichopus japonicus)" in six professional databases, such as Wiley, PubMed, ScienceDirect, ACS, Springer, and Web of Science, from 2000 to the present, summarizing the extraction, isolation, and purification methods for the four major categories (polysaccharides, proteins and peptides, saponins, and other components) of the A. japonicus chemical substances and 10 effective biological activities of A. japonicus. Included are anticoagulation, anticancer/antitumor activities, hematopoiesis, regulation of gut microbiota, and immune regulatory activities that correspond to traditional efficacy. Literature support is provided for the development of medicines and functional foods and related aspects that play a leading role in future directions.
Collapse
Affiliation(s)
- Chang Lu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xueyu Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jiahui Ma
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Mengtong Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wei Liu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guangyue Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yuling Ding
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhe Lin
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yong Li
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
25
|
Chen DD, Li Q, Wu JC. Efficient removal of purine compounds from solutions via biomass carbons derived from pomelo peel. J Biosci Bioeng 2023; 136:383-390. [PMID: 37775439 DOI: 10.1016/j.jbiosc.2023.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/20/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023]
Abstract
The high purine diet could result in the increase of the level of blood uric acid, causing serious health problems such as hyperuricemia, gout, nephropathy and cardiovascular diseases. To find out a safe, cheap and super adsorption material for removing purines in stomach or pretreating high-purine beverages, we used different tissues of pomelo peel to prepare biomass carbon by drying, chemical modification and carbonization and then applied it to remove purine compounds in strong acidic solution, beer and soybean milk. The characteristic analysis of pomelo-peel-derived carbons (PPCs) indicated that the preparation methods significantly affected the structures and adsorption capacities of PPCs. Compared with the biomass carbon derived from bamboo, PPCs exhibited higher adsorption capabilities for purine compounds in strong acidic solution (adsorption rates > 99% in 15 min) and soybean milk (adsorption rates > 56% in 30 min) but slightly lower adsorption capabilities in beer (adsorption rates > 52% in 30 min). In addition, the adsorption capabilities of PPCs for purine compounds in beer and soybean milk were not obviously affected by temperatures. Therefore, PPCs are promising absorbents for applications in removing purine compounds from beverages to produce low-purine, healthier products for treating hyperuricemia. The strong adsorption capabilities of PPCs on purine compounds in strong acidic environment also provides a possibility of using the PPCs as food additives for removing purines in stomach for healthcare applications such as gout prevention after confirming their biosafety.
Collapse
Affiliation(s)
- Dai Di Chen
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Qingxin Li
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China
| | - Jin Chuan Wu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510316, China.
| |
Collapse
|
26
|
Čypienė A, Gimžauskaitė S, Rinkūnienė E, Jasiūnas E, Laucevičius A, Ryliškytė L, Badarienė J. Effect of Alcohol Consumption Habits on Early Arterial Aging in Subjects with Metabolic Syndrome and Elevated Serum Uric Acid. Nutrients 2023; 15:3346. [PMID: 37571284 PMCID: PMC10421141 DOI: 10.3390/nu15153346] [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: 07/04/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Hyperuricemia is perceived as one of the risk factors for developing and progressing cardiovascular disease and metabolic syndrome through various pathological mechanisms. Endogenous synthesis and exogenous factors such as diet and beverages consumed play a major role in determining serum uric acid (sUA) levels. The aim of this study was to evaluate the effect of alcohol consumption on early arterial aging in middle-aged patients with metabolic syndrome (MetS) and hyperuricemia. MATERIALS AND METHODS This study included 661 middle-aged subjects (241 men and 420 women) from the Lithuanian High Cardiovascular Risk (LitHiR) primary prevention program. Characteristics of subjects such as blood pressure, laboratory testing, and the specialized nutrition profile questionnaire were evaluated. As an early marker of arterial stiffness, carotid-femoral pulse wave velocity (cfPWV) was assessed using a non-invasive applanation tonometry technique. RESULTS Hyperuricemia was present in 29% of men and 34% of women. Hyperuricemic men reported 1.6 times higher rates of alcohol drinking compared to men with normal sUA levels. After analyzing the correlation between alcohol consumption and cfPWV, no statistically significant relationships were found at a significance level of α = 0.05 but lowering the significance level to 0.06 revealed significant associations in men with normal sUA (ε2ordinal = 0.05, p = 0.06) and in women with increased sUA levels (ε2ordinal = 0.05, p = 0.08). Regression analysis showed that hyperuricemic men, consuming more than one unit of alcohol per week, had a significant impact on increasing cfPWV, while men with normal sUA levels, abstaining from alcohol entirely, resulted in a statistically significant decrease in cfPWV. Our results showed statistically significant relationships only among a group of men, although the women in the hyperuricemic group had a statistically higher cfPWV than women with normal sUA levels. CONCLUSIONS Drinking alcohol is associated with increased arterial stiffness among hyperuricemic middle-aged men with MetS.
Collapse
Affiliation(s)
- Alma Čypienė
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania; (A.Č.); (A.L.)
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Silvija Gimžauskaitė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Egidija Rinkūnienė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Eugenijus Jasiūnas
- Center of Informatics and Development, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania;
| | - Aleksandras Laucevičius
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania; (A.Č.); (A.L.)
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Ligita Ryliškytė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| | - Jolita Badarienė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (E.R.); (L.R.); (J.B.)
| |
Collapse
|
27
|
Yang X, Qiu H, Zhang Y, Zhang P. Quantitative structure-activity relationship study of amide derivatives as xanthine oxidase inhibitors using machine learning. Front Pharmacol 2023; 14:1227536. [PMID: 37456753 PMCID: PMC10339742 DOI: 10.3389/fphar.2023.1227536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
The target of the study is to predict the inhibitory effect of amide derivatives on xanthine oxidase (XO) by building several models, which are based on the theory of the quantitative structure-activity relationship (QSAR). The heuristic method (HM) was used to linearly select descriptors and build a linear model. XGBoost was used to non-linearly select descriptors, and radial basis kernel function support vector regression (RBF SVR), polynomial kernel function SVR (poly SVR), linear kernel function SVR (linear SVR), mix-kernel function SVR (MIX SVR), and random forest (RF) were adopted to establish non-linear models, in which the MIX-SVR method gives the best result. The kernel function of MIX SVR has strong abilities of learning and generalization of established models simultaneously, which is because it is a combination of the linear kernel function, the radial basis kernel function, and the polynomial kernel function. In order to test the robustness of the models, leave-one-out cross validation (LOOCV) was adopted. In a training set, R2 = 0.97 and RMSE = 0.01; in a test set, R2 = 0.95, RMSE = 0.01, and Rcv2 = 0.96. This result is in line with the experimental expectations, which indicate that the MIX-SVR modeling approach has good applications in the study of amide derivatives.
Collapse
|
28
|
Lin X, Zhou Q, Zhou L, Sun Y, Han X, Cheng X, Wu M, Lv W, Wang J, Zhao W. Quinoa ( Chenopodium quinoa Willd) Bran Saponins Alleviate Hyperuricemia and Inhibit Renal Injury by Regulating the PI3K/AKT/NFκB Signaling Pathway and Uric Acid Transport. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6635-6649. [PMID: 37083411 DOI: 10.1021/acs.jafc.3c00088] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Triterpenoids derived from natural products can exert antihyperuricemic effects. Here, we investigated the antihyperuricemic activity and mechanism of quinoa bran saponins (QBSs) in hyperuricemic mouse and cell models. The QBS4 fraction, with the highest saponin content, was used. Fourier-transform infrared, high-performance liquid chromatography, and ultrahigh-performance liquid chromatography-mass spectrometry identified 11 individual saponins in QBS4, of which the main components were hederagenin and oleanolic acid. The QBS4 effects on hyperuricemic mice (induced by adenine and potassium oxonate) were then studied. QBS4 reduced the levels of uric acid (UA), serum urea nitrogen, creatinine, and lipids in mice with hyperuricemia (HUA) and decreased renal inflammation and renal damage. Molecular analysis revealed that QBS4 may alleviate HUA by regulating the expression of key genes involved in the transport of UA and by inhibiting the activation of the PI3K/AKT/NFκB inflammatory signaling pathway. In conclusion, QBS4 has promise for using as a natural dietary supplement to treat and prevent HUA.
Collapse
Affiliation(s)
- Xuan Lin
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Qian Zhou
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Liangfu Zhou
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Yasai Sun
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Xue Han
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Xinlong Cheng
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Mengying Wu
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Wei Lv
- National Engineering Research Center for Semi-arid Agriculture, Shijiazhuang 050000, Hebei Province, China
| | - Jie Wang
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Wen Zhao
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| |
Collapse
|
29
|
Zhao Q, Jiang X, Mao Z, Zhang J, Sun J, Mao X. Exploration, sequence optimization and mechanism analysis of novel xanthine oxidase inhibitory peptide from Ostrea rivularis Gould. Food Chem 2023; 404:134537. [DOI: 10.1016/j.foodchem.2022.134537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
|
30
|
Yu D, Fu J, Wang Y, Lu F, Chen P, Liu S. Integrative metabolomics and network pharmacology to study the preventative impact of dioscin treatment on hyperuricemia. Biomed Chromatogr 2023; 37:e5558. [PMID: 36468521 DOI: 10.1002/bmc.5558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/11/2022]
Abstract
This work aims to combine network pharmacology and metabolomics to explore the mechanism of action of dioscin on hyperuricemia (HUA). The preventative impact of dioscin on HUA and its putative mechanism were examined using network pharmacological analysis and metabonomics. Network pharmacology study further pointed out the potential targets of dioscin after a review of the relevant biomarker pathways discovered by metabolomic analysis. Molecular docking was then used to examine how the active chemicals interacted with the target proteins. The therapeutic effect of dioscin on HUA was shown to be mediated by 13 potentially important metabolites as a result of metabonomic research. Most of these metabolites are regulated after dioscin therapy to help patients recover. Based on network pharmacology, we identified 10 central genes, which is partly in agreement with metabolomics data. Using metabolomics and network pharmacology, this study investigated the primary targets and mechanisms of dioscin in the treatment of HUA. It is advantageous that dioscin has been developed as an additional drug for the treatment of HUA.
Collapse
Affiliation(s)
- Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jiaqi Fu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yu Wang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Pingping Chen
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
31
|
Zhang N, Zhou J, Zhao L, Zhao Z, Wang S, Zhang L, Zhou F. Ferulic acid supplementation alleviates hyperuricemia in high-fructose/fat diet-fed rats via promoting uric acid excretion and mediating the gut microbiota. Food Funct 2023; 14:1710-1725. [PMID: 36722874 DOI: 10.1039/d2fo03332a] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The prevalence of hyperuricemia (HUA) has been rising, and it is typically accompanied by renal injury and intestinal flora disorder, leading to a non-negligible health crisis. Ferulic acid (FA), as a familiar polyphenol, has been proven to exert anti-hyperuricemic properties via inhibiting uric acid (UA) synthesis; however, the detailed underlying mechanisms remain unclear. The aim of this study was to explore the regulatory effect of FA on UA excretion as a potential strategy for reducing UA levels, and the comorbidities of HUA. FA treatment downregulated the expression of urate absorption transporter genes and repressed the toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) pathway in UA-stimulated HK-2 cells. To examine these effects in vivo, FA or allopurinol (positive control) was given to rats with HUA induced by a high-fructose/fat diet (HFFD) for 20 weeks. FA markedly decreased the serum UA, blood urea nitrogen, and creatinine levels. The expression of urate absorption transporters was downregulated, whereas the expression of secretion transporters was upregulated in the kidneys and intestines of FA-treated HUA rats. Additionally, FA mitigated renal oxidative stress, and suppressed the activation of the TLR4/NF-κB pathway and the downstream inflammatory response-related markers in the kidneys. Moreover, FA remodeled the composition of the gut microbiota, characterized by an increase in beneficial bacteria (e.g., Lactobacillus and Ruminococcus) and a decrease in pathogenic bacteria (e.g., Bacteroides). In conclusion, our study validated FA as an effective nutrient to ameliorate HFFD-induced HUA, suggesting its potential to mitigate the HUA-associated renal impairment and intestinal microbiota disturbance.
Collapse
Affiliation(s)
- Nanhai Zhang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, People's Republic of China.
| | - Jingxuan Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, People's Republic of China.
| | - Liang Zhao
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Zhen Zhao
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, People's Republic of China.
| | - Shiran Wang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, People's Republic of China.
| | - Liebing Zhang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, People's Republic of China.
| | - Feng Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, People's Republic of China.
| |
Collapse
|
32
|
He P, Xu H, Yang C, Yu D, Liu Y, Du J, Li Y. Unveiling the inhibitory mechanism of aureusidin targeting xanthine oxidase by multi-spectroscopic methods and molecular simulations. RSC Adv 2023; 13:1606-1616. [PMID: 36688063 PMCID: PMC9827282 DOI: 10.1039/d2ra06997k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/27/2022] [Indexed: 01/10/2023] Open
Abstract
Xanthine oxidase (XO) is a key target for gout treatment. Great efforts have been made towards the discovery and development of new XO inhibitors. Aureusidin (AUR), a natural compound, emerges as the second reported XO inhibitor with an aurone skeleton with an IC50 value of 7.617 ± 0.401 μM in vitro. The inhibitory mechanism of AUR against XO was explored through enzyme kinetic studies, multi-spectroscopic methods, computer simulation techniques, and ADME prediction. The results showed that AUR acts as a rapid reversible and mixed-type XO inhibitor and its binding to XO was driven by hydrogen bonding and hydrophobic interaction. Moreover, AUR presented a strong fluorescence quenching effect through a static quenching process and induced a conformation change of XO. Its binding pattern with XO was revealed through molecular docking, and its affinity toward XO was enhanced through interactions with key amino acid residues in the active pocket of XO. Further, AUR demonstrated good stability and pharmacokinetic behavior properties in molecular dynamics simulation and ADME prediction. In short, the current work clarified in depth the inhibitory mechanism of AUR on XO firstly and then provided fresh insights into its further development as a natural potent XO inhibitor with aurone skeleton.
Collapse
Affiliation(s)
- Pei He
- School of Chemical Engineering, Sichuan UniversityChengdu610065China+86 28 8540 5220
| | - Haiqi Xu
- School of Chemical Engineering, Sichuan UniversityChengdu610065China+86 28 8540 5220
| | - Can Yang
- School of Chemical Engineering, Sichuan UniversityChengdu610065China+86 28 8540 5220
| | - Dehong Yu
- School of Chemical Engineering, Sichuan UniversityChengdu610065China+86 28 8540 5220
| | - Yi Liu
- School of Chemical Engineering, Sichuan UniversityChengdu610065China+86 28 8540 5220
| | - Jiana Du
- School of Chemical Engineering, Sichuan UniversityChengdu610065China+86 28 8540 5220
| | - Yanfang Li
- School of Chemical Engineering, Sichuan UniversityChengdu610065China+86 28 8540 5220
| |
Collapse
|
33
|
Zhang H, Liu W, Qi SM, Chi JF, Gao Q, Lin XH, Ren S, Wang Z, Lei XJ, Li W. Improved effect of fresh ginseng paste (radix ginseng-ziziphus jujube) on hyperuricemia based on network pharmacology and molecular docking. Front Pharmacol 2022; 13:955219. [PMID: 36386218 PMCID: PMC9641371 DOI: 10.3389/fphar.2022.955219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 10/10/2022] [Indexed: 08/02/2024] Open
Abstract
Background: Hyperuricemia (HUA) is a metabolic disease caused by reduced excretion or increased production of uric acid. This research aims to study the practical components, active targets, and potential mechanism of the "Radix ginseng (RG)-Ziziphus jujube (ZJ)" herb pair through molecular docking, network pharmacology, and animal experiments. Methods: The potential targets of "Radix ginseng (RG)-Ziziphus jujube (ZJ)" herb pair were obtained from the TCMSP database. The therapeutic targets of HUA were acquired from the GendCards, OMIM, PharmGkb, and TTD databases. Protein-protein interaction network (PPI) was constructed in the STRING 11.0 database. The David database was used for enrichment analysis. Molecular Docking was finished by the AutoDock Vina. And we employed Radix ginseng and Ziziphus jujube as raw materials, which would develop a new functional food fresh ginseng paste (FGP) after boiling. In addition, benzbromarone (Ben) (7.8 mg/kg) and allopurinol (All) (5 mg/kg) were used as positive drugs to evaluate the hyperuricemia induced by FGP (400 and 800 mg/kg) potassium oxazine (PO) (100 mg/kg) and hypoxanthine (HX) (500 mg/kg) on mice. Results: The results showed that 25 targets in the "RG-ZJ" herb pair interacted with hyperuricemia. Then protein-protein interaction (PPI) analysis showed that TNF, IL-1β, and VEGFA were core genes. KEGG enrichment analysis showed that the Toll-like receptor signaling pathway and IL-17 signaling pathway were mainly involved. Meantime, animal experiments showed that FGP could improve the HUA status of mice by reducing serum UA BUN, XO, and liver XO levels (p < 0.05, p < 0.01). Furthermore, we analyzed the main ingredients of FGP by HPLC. We found that the main ingredients of FGP had solid binding activity to the core target of HUA by molecular docking. Conclusion: This study explored the active ingredients and targets of the "RG-ZJ" herb pair on HUA through network pharmacology, molecular docking, and animal experiments. It revealed the improvement of FGP in mice with HUA.
Collapse
Affiliation(s)
- Hao Zhang
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| | - Wei Liu
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| | - Si-Min Qi
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| | - Jian-Feng Chi
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Qiang Gao
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Xiang-Hui Lin
- Liaoning Xifeng Pharmaceutical Group Co., Ltd., Huanren, China
| | - Shen Ren
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Zi Wang
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Xiu-juan Lei
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
| | - Wei Li
- College of Chinese Medicinal Materials, National and Local Joint Engineering Research Center for Ginseng Breeding and Development, Jilin Agricultural University, Changchun, China
- College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| |
Collapse
|
34
|
Sun L, Ni C, Zhao J, Wang G, Chen W. Probiotics, bioactive compounds and dietary patterns for the effective management of hyperuricemia: a review. Crit Rev Food Sci Nutr 2022; 64:2016-2031. [PMID: 36073759 DOI: 10.1080/10408398.2022.2119934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Hyperuricemia is closely linked with an increased risk of developing hypertension, diabetes, renal failure and other metabolic syndromes. Probiotics, bioactive compounds and dietary patterns are safe cost-efficient ways to control hyperuricemia, whereas comprehensive reviews of their anti-hyperuricemic mechanisms are limited. This review summarizes the roles of probiotics, bioactive compounds and dietary patterns in treating hyperuricemia and critically reviews the possible mechanisms by which these interventions exert their activities. The dietary patterns are closely related to the occurrence of hyperuricemia through the indirect action of gut microbiota or the direct effects of host purine metabolism. The Mediterranean and Dietary Approaches to Stop Hypertension diets help reduce serum uric acid concentrations and thus prevent hyperuricemia. Meanwhile, probiotics alleviate hyperuricemia by ways of absorbing purine, restoring gut microbiota dysbiosis and inhibiting xanthine oxidase (XO) activity. Bioactive compounds such as polyphenols, peptides and alkaloids exert various anti-hyperuricemic effects, by regulating urate transporters, blocking the active sites of XO and inhibiting the toll-like receptor 4/nuclear factor kappa B signaling pathway and NOD-, LRR- and pyrin domain-containing protein 3 signaling pathway. This review will assist people with hyperuricemia to adopt a healthy diet and contribute to the application of natural products with anti-hyperuricemic activity.
Collapse
Affiliation(s)
- Lei Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Caixin Ni
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|