1
|
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
|
2
|
Yang M, Xu Y, Yu Q, Li M, Yang L, Yang Y. Spectroscopic Relationship between XOD and TAOZHI Total Polyphenols Based on Chemometrics and Molecular Docking Techniques. Molecules 2024; 29:4288. [PMID: 39339283 PMCID: PMC11433701 DOI: 10.3390/molecules29184288] [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: 08/05/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Xanthine oxidase (XOD) is a key enzyme that promotes the oxidation of xanthine/hypoxanthine to form uric acid, and the accumulation of uric acid leads to hyperuricaemia. The prevalence of gout caused by hyperuricaemia is increasing year by year. TAOZHI (TZ) can be used for the treatment of rheumatic arthralgia due to qi stagnation and blood stasis and contains a large number of polyphenolic components. The aim of this study was to investigate the relationship between chromatograms and XOD inhibition of 21 batches of TZ total polyphenol extract samples. Chemometric methods such as grey correlation analysis, bivariate correlation analysis, and partial least squares regression were used to identify the active ingredient groups in the total polyphenol extracts of TZ, which were validated using molecular docking techniques. The total polyphenol content contained in the 21 batches did not differ significantly, and all batches showed inhibitory effects on XOD. Spectroeffect correlation analysis showed that the inhibitory effect of TZ on XOD activity was the result of the synergistic effect of multiple components, and the active component groups screened to inhibit XOD were F2 (4-O-Caffeoylquinic acid), F4, and F10 (naringenin). The molecular docking results showed that the binding energies of all nine dockings were lower than -7.5 kcal/mol, and the binding modes included hydrogen bonding, hydrophobic forces, salt bridges, and π-staking, and the small molecules might exert their pharmacological effects by binding to XOD through the residue sites of the amino acids, such as threonine, arginine, and leucine. This study provides some theoretical basis for the development and utilisation of TZ total polyphenols.
Collapse
Affiliation(s)
- Mingyu Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
- Guizhou Key Laboratory for Raw Material of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Yitang Xu
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
- Guizhou Key Laboratory for Raw Material of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Qihua Yu
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
- Guizhou Key Laboratory for Raw Material of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Mengyu Li
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
- Guizhou Key Laboratory for Raw Material of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Liyong Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Ye Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
- Guizhou Key Laboratory for Raw Material of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| |
Collapse
|
3
|
Lian Y, Fu G, Liang X, He X, Xu J, Fan H, Wan Y. Combination of Artemisia selengensis Turcz leaves polysaccharides and dicaffeoylquinic acids could be a potential inhibitor for hyperuricemia. Int J Biol Macromol 2024; 271:132687. [PMID: 38806079 DOI: 10.1016/j.ijbiomac.2024.132687] [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: 08/02/2023] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
Caffeioyl quinic acids and polysaccharides from Artemisia selengensis Turcz are considered potential bioactive substances for hyperuricemia (HUA) treatment. While the mechanism of multi-component combined intervention of polysaccharides and dicaffeoylquinic acids (diCQAs) is not yet clear. In this study, we investigated the effect of A. selengensis Turcz leaves polysaccharides (APS) on the HUA treatment with diCQAs in vitro by direct inhibition of XOD activities and in vivo by using animal model. The results showed that APS had almost no inhibitory effect on XOD activities in vitro, but the inhibitory activity of diCQAs on XOD was affected by changes in inhibition type and inhibition constant. Compared to APS and diCQAs alone, high-dose APS and diCQAs in combination (ADPSh) could significantly reduce the production of uric acid (16.38 % reduction compared to diCQAs group) and oxidative stress damage. Additionally, this combined therapy showed promise in restoring the gut microbiota balance and increasing the short-chain fatty acids levels. The results suggested that APS and diCQAs in combination could be a potential inhibitor for HUA treatment.
Collapse
Affiliation(s)
- Yingzhu Lian
- State Key Laboratory of Food Science and Resources, College of Food Science & Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Guiming Fu
- State Key Laboratory of Food Science and Resources, College of Food Science & Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Xinmei Liang
- State Key Laboratory of Food Science and Resources, College of Food Science & Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Xinchao He
- State Key Laboratory of Food Science and Resources, College of Food Science & Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Jin Xu
- State Key Laboratory of Food Science and Resources, College of Food Science & Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Haowei Fan
- State Key Laboratory of Food Science and Resources, College of Food Science & Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Yin Wan
- State Key Laboratory of Food Science and Resources, College of Food Science & Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| |
Collapse
|
4
|
Lou Y, Gao Q, Fan M, Waleed AA, Wang L, Li Y, Qian H. Ferulic acid ameliorates hyperuricemia by regulating xanthine oxidase. Int J Biol Macromol 2023; 253:126542. [PMID: 37634782 DOI: 10.1016/j.ijbiomac.2023.126542] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Hyperuricemia is characterized by elevated uric acid (UA) level in the body. The xanthine oxidase (XO) inhibitory ability is an important way to evaluate the anti-hyperuricemia effect of natural products. Ferulic acid (FA) is a phenolic acid compound, and it is a free radical scavenger with many physiological functions. The aim of this study was to investigate the structure-activity relationship, potential mechanism and interaction of FA as XO's inhibitor. In the cell experiment, using 1.25 mM adenosine to incubate for 24 h under the optimal conditions (37 °C, pH = 7.2) can increase the UA production by 1.34 folds. PCR analysis showed that FA could reduce the mRNA expression level of XO. FA inhibited XO in a mixed mode (IC50 = 13.25 μM). The fluorescence quenching of XO by FA occurs through a static mechanism, with an inhibition constant of Ki = 9.527 × 10-5 mol L-1 and an apparent coefficient of α = 1.768. The enthalpy and entropy changes were found as -267.79 KJ mol-1 and - 860.85 KJ mol-1, indicating that both hydrogen binding and hydrophobic are involved in the interaction of this polyphenolic natural compound with XO. Thus, FA supplementation may be a potential therapeutic strategy to improve hyperuricemia by reducing UA production.
Collapse
Affiliation(s)
- Ye Lou
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Qiang Gao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Mingcong Fan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Al-Ansi Waleed
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
| | - Haifeng Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China.
| |
Collapse
|
5
|
Chen J, Zhang Z, Li H, Tang H. Exploring the effect of a series of flavonoids on tyrosinase using integrated enzyme kinetics, multispectroscopic, and molecular modelling analyses. Int J Biol Macromol 2023; 252:126451. [PMID: 37619686 DOI: 10.1016/j.ijbiomac.2023.126451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/07/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
The control of food browning can be achieved by inhibiting tyrosinase (TY) activity, but current studies on the interaction of flavonoids as potent inhibitors with TY are inadequate. Herein, the effect of a library of flavonoids on TY was investigated using enzyme kinetics, multispectroscopic methods, and molecular modelling. Some flavonoids including 4, 8, 10, 17, 18, 28, 30, 33, and 34 exhibited potent TY inhibitory activity, with compound 10 demonstrating reversible inhibition in a mixed-competitive manner. Ultraviolet-visible spectral changes confirmed the formation of flavonoid-TY complexes. Fluorescence quenching analysis suggested effective intrinsic fluorescence quenching by flavonoids through static quenching with the ground-state complex formation. Synchronous fluorescence spectra showed the microenvironment change around the fluorophores induced by flavonoids. ANS-binding fluorescence assay indicated TY's surface hydrophobicity change by flavonoids and highlighted the change in secondary structure conformation, which was further confirmed by Fourier-transform infrared spectra. Molecular modelling results helped visualize the preferred binding conformation at the active site of TY, and demonstrated the important role of hydrophobic interaction and hydrogen bonding in stabilizing the flavonoid-TY complexes. These findings prove that diverse flavonoid structures distinctly impact their binding behavior on TY and contribute to understanding flavonoids' potential as TY inhibitors in controlling food browning.
Collapse
Affiliation(s)
- Jin Chen
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Zhuangwei Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Huihui Li
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Hongjin Tang
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| |
Collapse
|
6
|
Wang Y, Zhou L, Chen M, Liu Y, Yang Y, Lu T, Ban F, Hu X, Qian Z, Hong P, Zhang Y. Mining Xanthine Oxidase Inhibitors from an Edible Seaweed Pterocladiella capillacea by Using In Vitro Bioassays, Affinity Ultrafiltration LC-MS/MS, Metabolomics Tools, and In Silico Prediction. Mar Drugs 2023; 21:502. [PMID: 37888437 PMCID: PMC10608504 DOI: 10.3390/md21100502] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
The prevalence of gout and the adverse effects of current synthetic anti-gout drugs call for new natural and effective xanthine oxidase (XOD) inhibitors to target this disease. Based on our previous finding that an edible seaweed Pterocladiella capillacea extract inhibits XOD, XOD-inhibitory and anti-inflammatory activities were used to evaluate the anti-gout potential of different P. capillacea extract fractions. Through affinity ultrafiltration coupled with liquid chromatography tandem mass spectrometry (LC-MS/MS), feature-based molecular networking (FBMN), and database mining of multiple natural products, the extract's bioactive components were traced and annotated. Through molecular docking and ADMET analysis, the possibility and drug-likeness of the annotated XOD inhibitors were predicted. The results showed that fractions F4, F6, F4-2, and F4-3 exhibited strong XOD inhibition activity, among which F4-3 reached an inhibition ratio of 77.96% ± 4.91% to XOD at a concentration of 0.14 mg/mL. In addition, the P. capillacea extract and fractions also displayed anti-inflammatory activity. Affinity ultrafiltration LC-MS/MS analysis and molecular networking showed that out of the 20 annotated compounds, 8 compounds have been previously directly or indirectly reported from seaweeds, and 4 compounds have been reported to exhibit anti-gout activity. Molecular docking and ADMET showed that six seaweed-derived compounds can dock with the XOD activity pocket and follow the Lipinski drug-like rule. These results support the value of further investigating P. capillacea as part of the development of anti-gout drugs or related functional foods.
Collapse
Affiliation(s)
- Yawen Wang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Longjian Zhou
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Minqi Chen
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Yayue Liu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yu Yang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Tiantian Lu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Fangfang Ban
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Xueqiong Hu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
| | - Zhongji Qian
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| | - Pengzhi Hong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yi Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (Y.W.); (L.Z.); (M.C.); (Y.L.); (Y.Y.); (T.L.); (F.B.); (X.H.); (Z.Q.); (P.H.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
7
|
Wang X, Dong L, Dong Y, Bao Z, Lin S. Corn Silk Flavonoids Ameliorate Hyperuricemia via PI3K/AKT/NF-κB Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37294890 DOI: 10.1021/acs.jafc.3c03422] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hyperuricemia (HUA) is a widespread metabolic disease marked by an elevated level of uric acid, and is a risk factor for premature death. The protective effect of corn silk flavonoids (CSF) against HUA and its potential mechanisms were explored. Five important apoptosis and inflammation-related signaling pathways were identified by network pharmacological analysis. The CSF exhibited significant uric acid (UA)-lowering activity in vitro by decreasing xanthine oxidase (XOD) and increasing hypoxanthine-guanine phosphoribosyl transferase levels. In a potassium oxonate-induced HUA in vivo, CSF treatment effectively inhibited XOD activity and promoted UA excretion. Furthermore, it decreased the levels of TNF-α and IL-6 and restored pathological damage. In summary, CSF is a functional food component to improve HUA by reducing inflammation and apoptosis through the down-regulating PI3K/AKT/NF-κB pathway.
Collapse
Affiliation(s)
- Xizhu Wang
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- Liaoning Engineering Research Center of Special Dietary Food, Dalian polytechnic University, Dalian 116034, P. R. China
| | - Liu Dong
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- Liaoning Engineering Research Center of Special Dietary Food, Dalian polytechnic University, Dalian 116034, P. R. China
| | - Yifei Dong
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- Liaoning Engineering Research Center of Special Dietary Food, Dalian polytechnic University, Dalian 116034, P. R. China
| | - Zhijie Bao
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- Liaoning Engineering Research Center of Special Dietary Food, Dalian polytechnic University, Dalian 116034, P. R. China
| | - Songyi Lin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- Liaoning Engineering Research Center of Special Dietary Food, Dalian polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
8
|
Saleem H, Yaqub A, Rafique R, Ali Chohan T, Malik DES, Tousif MI, Khurshid U, Ahemad N, Ramasubburayan R, Rengasamy KR. Nutritional and medicinal plants as potential sources of enzyme inhibitors toward the bioactive functional foods: an updated review. Crit Rev Food Sci Nutr 2023; 64:9805-9828. [PMID: 37255100 DOI: 10.1080/10408398.2023.2217264] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Enzymes are biologically active complex protein molecules that catalyze most chemical reactions in living organisms, and their inhibitors accelerate biological processes. This review emphasizes medicinal food plants and their isolated chemicals inhibiting clinically important enzymes in common diseases. A mechanistic overview was investigated to explain the mechanism of these food bases enzyme inhibitors. The enzyme inhibition potential of medicinal food plants and their isolated substances was searched in Ovid, PubMed, Science Direct, Scopus, and Google Scholar. Cholinesterase, amylase, glucosidase, xanthine oxidase, tyrosinase, urease, lipoxygenase, and others were inhibited by crude extracts, solvent fractions, or isolated pure chemicals from medicinal food plants. Several natural compounds have shown tyrosinase inhibition potential, including quercetin, glabridin, phloretin-4-O-β-D-glucopyranoside, lupinalbin, and others. Some of these compounds' inhibitory kinetics and molecular mechanisms are also discussed. Phenolics and flavonoids inhibit enzyme activity best among the secondary metabolites investigated. Several studies showed flavonoids' significant antioxidant and anti-inflammatory activities, highlighting their medicinal potential. Overall, many medicinal food plants, their crude extracts/fractions, and isolated compounds have been studied, and some promising compounds depending on the enzyme have been found. Still, more studies are recommended to derive potential pharmacologically active functional foods.
Collapse
Affiliation(s)
- Hammad Saleem
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| | - Anam Yaqub
- Fatima Memorial Medical and Dental College, Lahore, Pakistan
| | | | - Tahir Ali Chohan
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| | - Durr-E-Shahwar Malik
- Institute of Pharmaceutical Sciences, Peoples University of Medical and Health Sciences, NawabShah, Pakistan
| | - Muhammad Imran Tousif
- Department of Chemistry, Division of Science and Technology, University of Education Lahore, Pakistan
| | - Umair Khurshid
- Department of Pharmaceutical Chemistry, The Islamia University of Bahawalpur, Pakistan
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ramasamy Ramasubburayan
- Marine Biomedical Research Lab & Environmental Toxicology Unit, Department of Prosthodotics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Kannan Rr Rengasamy
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| |
Collapse
|
9
|
Song J, Wang Z, Chi Y, Zhang Y, Fang C, Shu Y, Cui J, Bai H, Wang J. Anti-gout activity and the interaction mechanisms between Sanghuangporus vaninii active components and xanthine oxidase. Bioorg Chem 2023; 133:106394. [PMID: 36801789 DOI: 10.1016/j.bioorg.2023.106394] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/13/2023]
Abstract
Xanthine oxidase (XO) plays a critical role in the progression of gout. We showed in a previous study that Sanghuangporus vaninii (S. vaninii), a perennial, medicinal, and edible fungus traditionally used to treat various symptoms, contains XO inhibitors. In the current study, we isolated an active component of S. vaninii using high performance countercurrent chromatography and identified it as davallialactone using mass spectrometry with 97.726 % purity. A microplate reader showed that davallialactone had mixed inhibition of XO activity with a half-inhibitory concentration value of 90.07 ± 2.12 μM. In addition, the collision between davallialactone and XO led to fluorescence quenching and conformational changes in XO, which were mainly driven by hydrophobicity and hydrogen bonding. Molecular simulations further showed that davallialactone was located at the center of the molybdopterin (Mo-Pt) of XO and interacted with amino acid residues Phe798, Arg912, Met1038, Ala1078, Ala1079, Gln1194, and Gly1260, suggesting that entering the enzyme-catalyzed reaction was unfavorable for the substrate. We also observed face-to-face π-π interactions between the aryl ring of davallialactone and Phe914. Cell biology experiments indicated that davallialactone reduced the expression of the inflammatory factors, tumor necrosis factor alpha and interleukin-1 beta (P < 0.05), can effectively alleviate cellular oxidative stress. This study showed that davallialactone significantly inhibits XO and has the potential to be developed into a novel medicine to prevent hyperuricemia and treat gout.
Collapse
Affiliation(s)
- Jiling Song
- The College of Chemistry, Changchun Normal University, Changchun 130032, China
| | - Zhanwei Wang
- The College of Chemistry, Changchun Normal University, Changchun 130032, China
| | - Yu Chi
- The College of Chemistry, Changchun Normal University, Changchun 130032, China
| | - Yong Zhang
- The Central Laboratory, Changchun Normal University, Changchun 130032, China; Nanguan Middle School, Honghua Gang District, Zunyi 563000, China
| | - Chenyi Fang
- The College of Chemistry, Changchun Normal University, Changchun 130032, China
| | - Yuting Shu
- The College of Chemistry, Changchun Normal University, Changchun 130032, China
| | - Jing Cui
- The Central Laboratory, Changchun Normal University, Changchun 130032, China
| | - Helong Bai
- The College of Chemistry, Changchun Normal University, Changchun 130032, China
| | - Jing Wang
- The College of Chemistry, Changchun Normal University, Changchun 130032, China; The Central Laboratory, Changchun Normal University, Changchun 130032, China.
| |
Collapse
|
10
|
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: 1.0] [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
|
11
|
Crosstalk between xanthine oxidase (XO) inhibiting and cancer chemotherapeutic properties of comestible flavonoids- a comprehensive update. J Nutr Biochem 2022; 110:109147. [PMID: 36049673 DOI: 10.1016/j.jnutbio.2022.109147] [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: 06/03/2021] [Revised: 12/17/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Gout is an inflammatory disease caused by metabolic disorder or genetic inheritance. People throughout the world are strongly dependent on ethnomedicine for the treatment of gout and some receive satisfactory curative treatment. The natural remedies as well as established drugs derived from natural sources or synthetically made exert their action by mechanisms that are closely associated with anticancer treatment mechanisms regarding inhibition of xanthine oxidase, feedback inhibition of de novo purine synthesis, depolymerization and disappearance of microtubule, inhibition of NF-ĸB activation, induction of TRAIL, promotion of apoptosis, and caspase activation and proteasome inhibition. Some anti-gout and anticancer novel compounds interact with same receptors for their action, e.g., colchicine and colchicine analogues. Dietary flavonoids, i.e., chrysin, kaempferol, quercetin, fisetin, pelargonidin, apigenin, luteolin, myricetin, isorhamnetin, phloretinetc etc. have comparable IC50 values with established anti-gout drug and effective against both cancer and gout. Moreover, a noticeable number of newer anticancer compounds have already been isolated from plants that have been using by local traditional healers and herbal practitioners to treat gout. Therefore, the anti-gout plants might have greater potentiality to become selective candidates for screening of newer anticancer leads.
Collapse
|
12
|
Iron-doped cerium/nucleotide coordination polymer as highly efficient peroxidase mimic for colorimetric detection of fluoride ion. Mikrochim Acta 2022; 189:346. [PMID: 36001171 DOI: 10.1007/s00604-022-05410-6] [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: 05/07/2022] [Accepted: 07/06/2022] [Indexed: 10/15/2022]
Abstract
A new coordination polymer (Ce-Fe-GMP) with excellent catalytic activity was prepared by a facile route, which was further applied to the detection of F- with high sensitivity and selectivity. The simple doping of Fe3+ into the coordination network can easily modulate the mixing ratio of Ce3+ and Ce4+ in the presence of H2O2, which can extremely improve the catalytic ability of Ce-Fe-GMP. Based on the synergistic effect, the Ce-Fe-GMP with dual-active sites shows better peroxidase activity than that of Ce-GMP. In addition, we found that F- can inhibit the peroxidase activity of Ce-Fe-GMP because of the coordination structure fragmentation and the regulation of Ce3+/Ce4+ ratio. Therefore, different concentrations of F- can be detected by the colorimetric reaction based on this mechanism. The absorption at 652 nm displays a good linear relationship versus the concentration of F- over the range 2.0 to 100.0 μM. Furthermore, F- in real mineral-mixed samples can be measured with satisfactory results. The colorimetric strategy based on the peroxidase activity of Ce-Fe-GMP is simple and low-cost, which shows the potential applications in the field of on-site environment measurement.
Collapse
|
13
|
Vijeesh V, Vysakh A, Jisha N, Latha MS. In vitro enzyme inhibition and in vivo anti-hyperuricemic potential of eugenol: An experimental approach. Drug Dev Ind Pharm 2022; 47:1998-2003. [PMID: 35616220 DOI: 10.1080/03639045.2022.2083156] [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] [Indexed: 10/18/2022]
Abstract
Xanthine oxidase (XO) was accountable for the uric acid synthesis in the body and is considered as a prominent therapeutic target in urate lowering treatment. Eugenol is a natural compound commonly found in the clove, cinnamon etc. and have various biological activities. This study was designed to examine the anti-hyperuricemic effect of eugenol by in vitro and in vivo studies. Potassium oxonate (PO) was used to induce hyperuricemia in Wistar rats. Different doses of eugenol (1.25, 2.5 and 5mg/kg bwt orally) were used for the treatment and various biological function markers (renal, hepatic and hematological) were analyzed. The IC50 value obtained for eugenol was 3.51 ± 0.002 μM. The kinetic studies revealed that the eugenol exhibited a mixed type of inhibition. Abnormality in the levels of various biological function markers were observed in the PO treated rats. Upon the eugenol treatment, those biological function markers were retained near to its normal values. The study proved the anti-hyperuricemic potential of eugenol against the PO induced hyperuricemia model.
Collapse
Affiliation(s)
- V Vijeesh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - A Vysakh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - Ninan Jisha
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - M S Latha
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| |
Collapse
|
14
|
Feng S, Wu S, Xie F, Yang CS, Shao P. Natural compounds lower uric acid levels and hyperuricemia: Molecular mechanisms and prospective. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Liu Y, Peng B. A Novel Hyaluronic Acid-Black Rice Anthocyanins Nanocomposite: Preparation, Characterization, and Its Xanthine Oxidase (XO)-Inhibiting Properties. Front Nutr 2022; 9:879354. [PMID: 35495941 PMCID: PMC9048741 DOI: 10.3389/fnut.2022.879354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/25/2022] [Indexed: 12/05/2022] Open
Abstract
To promote the normal metabolism of human uric acid, high-performance hyaluronic acid-black rice anthocyanins (HAA) nanocomposite particles were successfully prepared by a simple crosslinking method as a novel xanthine oxidase inhibitor. Its structure and properties were characterized by scanning electron microscopy (SEM), transmission electron microscopy (TEM), Fourier transform infrared spectrometry (FT-IR), and X-ray diffraction (XRD). SEM and TEM electron microscopy showed an obvious double-layer spherical structure with a particle size of ~298 nm. FT-IR and XRD analysis confirmed that black rice anthocyanins (ATC) had been successfully loaded into the hyaluronic acid (HA) structure. Nanocomposite particles (embedded form) showed higher stability in different environments than free black rice ATC (unembedded form). In addition, the preliminary study showed that the inhibition rate of the nanocomposite particles on Xanthine oxidase (XO) was increased by 40.08%. These results indicate that HAA nanocomposite particles can effectively improve black rice ATC's stability and activity, creating an ideal new material for inhibiting XO activity that has a broad application prospect.
Collapse
|
16
|
Du H, Li SJ. Inhibition of porphyra polysaccharide on xanthine oxidase activity and its inhibition mechanism. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 266:120446. [PMID: 34628362 DOI: 10.1016/j.saa.2021.120446] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/18/2021] [Accepted: 09/24/2021] [Indexed: 06/13/2023]
Abstract
Xanthine oxidase (XO) is a purine catabolic enzyme related to hyperuricemia and gout. Porphyra polysaccharide (PP) is a kind of sulfated polysaccharide with potent biological activity. Herein, the interaction mechanism between PP and XO was studied by enzyme kinetics and multi-spectroscopy methods for the first time. Inhibition kinetics assay showed that PP reversibly inhibited XO activity in a mixed competitive manner with an IC50 of 10.53 ± 0.69 mg/ml. Fluorescence titration studies and thermodynamic parameter calculations revealed that PP could spontaneously bind to XO through hydrophobic interactions, with a class of binding site. Circular dichroism analysis demonstrated that PP induced secondary structure rearrangement and conformational change of XO. Molecular docking further revealed that PP inserted into the hydrophobic cavity of XO, occupying the catalytic center, leading to the inhibition of XO activity. This study may provide new insights into the inhibitory mechanism of PP as a promising XO inhibitor.
Collapse
Affiliation(s)
- Hongyan Du
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, PR China
| | - Shu Jie Li
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, PR China; Qilu Institute of Technology, Shandong 250200, PR China.
| |
Collapse
|
17
|
Screening of uric acid-lowering active components of corn silk polysaccharide and its targeted improvement on renal excretory dysfunction in hyperuricemia mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
18
|
Vijeesh V, Jisha N, Vysakh A, Latha MS. Interaction of eugenol with xanthine oxidase: Multi spectroscopic and in silico modelling approach. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 258:119843. [PMID: 33933941 DOI: 10.1016/j.saa.2021.119843] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/04/2021] [Accepted: 04/12/2021] [Indexed: 06/12/2023]
Abstract
Eugenol, a major component in clove has various biological activities. The current study focused to the binding potential of eugenol with Xanthine oxidase (XO) were evaluated using multi spectroscopic techniques and in silico docking studies. Xanthine oxidase, a superoxide generating enzyme, catalyses hypoxanthine and xanthine to uric acid. An excessive uric acid and superoxide anion radical in our body causes many serious clinical complications. The activity and the structural alterations can be a significant method to reduce this kind of risk factors. The results obtained from the fluorescence titration exhibited the interactions initiated by a static quenching mechanism. The ultraviolet (UV), fourier-transform infrared (FTIR), circular dichroism (CD) spectroscopic analysis of eugenol bind with XO indicated the secondary structural alteration in XO. Docking studies showed molecular level interaction of eugenol with the amino acid residues of Thr 1010, Phe 914, Phe 1009, Leu 1014, Phe 1009, Val 1011, Arg 880, Ala 1078, Glu 802, Leu 648and Leu 873 which residing at the catalytic active site of the XO. These results inferred that the eugenol can interact with XO in a remarkable manner and these findings provide a supporting data for the XO inhibition studies to propose a new lead compound.
Collapse
Affiliation(s)
- V Vijeesh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - Ninan Jisha
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - A Vysakh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - M S Latha
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India.
| |
Collapse
|
19
|
Wan Y, Qian J, Li Y, Shen Y, Chen Y, Fu G, Xie M. Inhibitory mechanism of xanthine oxidase activity by caffeoylquinic acids in vitro. Int J Biol Macromol 2021; 184:843-856. [PMID: 34146563 DOI: 10.1016/j.ijbiomac.2021.06.075] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/14/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Abstract
In this study, the inhibitory activities of eight caffeoylquinic acids (CQAs) against xanthine oxidase (XOD) in vitro were investigated, and the interaction mechanisms between each compound and XOD were studied. HPLC and fluorescence spectra showed that the inhibitory activities of dicaffeoylquinic acids (diCQAs) were higher than that of monocaffeoylquinic acids (monoCQAs), due to the main roles of hydrophobic interaction and hydrogen bond between XOD and diCQAs. Both the binding constant and the lowest binding energy data indicated that the affinities of diCQAs to XOD were stronger than that of monoCQAs. Circular dichroism showed that the structure of XOD was compacted with the increased of α-helix content, resulting in decreased enzyme catalytic activity. Molecular docking revealed that CQAs preferentially bind to the flavin adenine dinucleotide region in XOD. These results provided the mechanisms of CQAs on inhibiting XOD and the further utilization of CQAs as XOD inhibitors to prevent hyperuricemia.
Collapse
Affiliation(s)
- Yin Wan
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Jin Qian
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Yizhen Li
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Yuefeng Shen
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Yanru Chen
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Guiming Fu
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China
| |
Collapse
|
20
|
Mehmood A, Zhao L, Ishaq M, Xin W, Zhao L, Wang C, Hossen I, Zhang H, Lian Y, Xu M. Anti-hyperuricemic potential of stevia (Stevia rebaudiana Bertoni) residue extract in hyperuricemic mice. Food Funct 2021; 11:6387-6406. [PMID: 32613954 DOI: 10.1039/c9fo02246e] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Hyperuricemia (HUA) is considered a potent risk factor for the development of gout, renal failure, and cardiovascular disease. The current project was designed to use stevia (Stevia rebaudiana Bertoni) byproduct, named stevia residue extract (STVRE), for the treatment of HUA. Male Kunming mice were divided into six groups: normal control, model control, positive control (allopurinol, 5 mg per kg body weight [bw]), STVRE-1 (75 mg per kg bw), STVRE-2 (150 mg per kg bw), and STVRE-3 (300 mg per kg bw). HUA was induced by the administration of potassium oxonate (100 mg per kg bw), fructose (10% w/v), and yeast extract (100 mg per kg bw) for 8 weeks. STVRE significantly (p < 0.05) decreased uric acid (UA) production and ameliorated UA excretion by interacting with urate transporters. The STVRE remarkably attenuated oxidative stress mediated by UA and downregulated inflammatory-related response markers such as COX-2, NF-κB, PGE2, IL-1β, and TNF-α. Furthermore, STVRE also reversed HUA-induced abnormalities in kidneys compared with the MC group. The results of our study suggest that STVRE has potential to attenuate hyperuricemia and renal protective effects, and may be used as a natural supplement for the possible treatment of UA-related disorders.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives School of Food and Chemical Technology, Beijing Technology and Business University, Beijing 100048, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Action mechanisms and interaction of two key xanthine oxidase inhibitors in galangal: Combination of in vitro and in silico molecular docking studies. Int J Biol Macromol 2020; 162:1526-1535. [DOI: 10.1016/j.ijbiomac.2020.07.297] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/09/2020] [Accepted: 07/27/2020] [Indexed: 11/18/2022]
|
22
|
Insights from multispectral and molecular docking investigation on the xanthine oxidase inhibition by 1,4-dicaffeoylquinic acid. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
23
|
Tang H, Huang L, Zhao D, Sun C, Song P. Interaction mechanism of flavonoids on bovine serum albumin: Insights from molecular property-binding affinity relationship. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 239:118519. [PMID: 32480277 DOI: 10.1016/j.saa.2020.118519] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 06/11/2023]
Abstract
The molecular structure properties-binding affinity relationship of a series of flavonoids and bovine serum albumin (BSA) was investigated in vitro from comparing the binding constants determined through the fluorescence method. As a result, the binding process was greatly influenced by different structural elements or substituents of flavonoids under analysis. The hydroxylation at the positions C3, C6, C4', C5' (for type I) and C5, C3' (for type II) were in favor of forming hydrogen bonds with the amino acids of BSA, which was of great importance in the binding and interaction between flavonoids and the protein. The decreased affinity could be realized by the methoxylation (C8, C3' and C4') and glycosylation (C3 and C7) of flavonoid type I. However, the adverse trend on binding affinity was observed when the methoxylation and glycosylation appeared at the sites C4' and C7, C4' of structure type II, respectively. Meanwhile, glycosylation at C7 mainly induced the decline in the affinity of flavonoids (type III), and the hydrogenation of the C2C3 double bond for type I was beneficial to increase the affinity on BSA. Moreover, part of flavonoids could mediate the conformational alteration of secondary structures of the protein during the interaction process, which was inferred by means of the synchronous fluorescence spectra. The determinations of ANS fluorescence probe suggested that hydrophobic interaction played an important role in the binding of a majority of flavonoids to BSA. Further evidences from the site-specific experiments revealed that the location of flavonoids 19, 29 and 34 binding on BSA mainly belonged to site I, while compound 3 bound to both sites I and II. Additionally, molecular modelling studies further confirmed the indispensable character of hydrophobic interaction and hydrogen bonds, and illustrated the preferred complex binding behaviors.
Collapse
Affiliation(s)
- Hongjin Tang
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| | - Lin Huang
- Blood Purification Center, Affiliated Yijishan Hospital of Wannan Medical College, Wuhu 241001, PR China
| | - Dongsheng Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Chunyong Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Ping Song
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| |
Collapse
|
24
|
Singh JV, Bedi PMS, Singh H, Sharma S. Xanthine oxidase inhibitors: patent landscape and clinical development (2015–2020). Expert Opin Ther Pat 2020; 30:769-780. [DOI: 10.1080/13543776.2020.1811233] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jatinder Vir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | | | - Harbinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
25
|
Jiang LL, Gong X, Ji MY, Wang CC, Wang JH, Li MH. Bioactive Compounds from Plant-Based Functional Foods: A Promising Choice for the Prevention and Management of Hyperuricemia. Foods 2020; 9:foods9080973. [PMID: 32717824 PMCID: PMC7466221 DOI: 10.3390/foods9080973] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Hyperuricemia is a common metabolic disease that is caused by high serum uric acid levels. It is considered to be closely associated with the development of many chronic diseases, such as obesity, hypertension, hyperlipemia, diabetes, and cardiovascular disorders. While pharmaceutical drugs have been shown to exhibit serious side effects, and bioactive compounds from plant-based functional foods have been demonstrated to be active in the treatment of hyperuricemia with only minimal side effects. Indeed, previous reports have revealed the significant impact of bioactive compounds from plant-based functional foods on hyperuricemia. This review focuses on plant-based functional foods that exhibit a hypouricemic function and discusses the different bioactive compounds and their pharmacological effects. More specifically, the bioactive compounds of plant-based functional foods are divided into six categories, namely flavonoids, phenolic acids, alkaloids, saponins, polysaccharides, and others. In addition, the mechanism by which these bioactive compounds exhibit a hypouricemic effect is summarized into three classes, namely the inhibition of uric acid production, improved renal uric acid elimination, and improved intestinal uric acid secretion. Overall, this current and comprehensive review examines the use of bioactive compounds from plant-based functional foods as natural remedies for the management of hyperuricemia.
Collapse
Affiliation(s)
- Lin-Lin Jiang
- Department of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China;
| | - Xue Gong
- Department of Pharmacy, Baotou Medical College, Baotou 014060, China; (X.G.); (M.-Y.J.); (C.-C.W.)
| | - Ming-Yue Ji
- Department of Pharmacy, Baotou Medical College, Baotou 014060, China; (X.G.); (M.-Y.J.); (C.-C.W.)
| | - Cong-Cong Wang
- Department of Pharmacy, Baotou Medical College, Baotou 014060, China; (X.G.); (M.-Y.J.); (C.-C.W.)
| | - Jian-Hua Wang
- Department of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China;
- Correspondence: (J.-H.W.); (M.-H.L.); Tel.: +86-472-716-7795 (M.-H.L.)
| | - Min-Hui Li
- Department of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China;
- Department of Pharmacy, Baotou Medical College, Baotou 014060, China; (X.G.); (M.-Y.J.); (C.-C.W.)
- Department of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
- Pharmaceutical Laboratory, Inner Mongolia Institute of Traditional Chinese Medicine, Hohhot 010020, China
- Inner Mongolia Key Laboratory of Characteristic Geoherbs Resources Protection and Utilization, Baotou Medical College, Baotou 014060, China
- Correspondence: (J.-H.W.); (M.-H.L.); Tel.: +86-472-716-7795 (M.-H.L.)
| |
Collapse
|
26
|
Liu L, Zhang L, Ren L, Xie Y. Advances in structures required of polyphenols for xanthine oxidase inhibition. FOOD FRONTIERS 2020. [DOI: 10.1002/fft2.27] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Liangliang Liu
- Institute of Bast Fiber Crops Chinese Academy of Agricultural Sciences Changsha 410205 China
| | - Li Zhang
- College of Chemistry and Materials Engineering Huaihua University Huaihua 418000 China
| | - Licheng Ren
- Institute of Bast Fiber Crops Chinese Academy of Agricultural Sciences Changsha 410205 China
- Department of Plastic and Cosmetic Surgery Shenzhen University General Hospital Shenzhen 518055 China
| | - Yixi Xie
- Institute of Bast Fiber Crops Chinese Academy of Agricultural Sciences Changsha 410205 China
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province Xiangtan University Xiangtan 411105 China
| |
Collapse
|
27
|
Mehmood A, Rehman AU, Ishaq M, Zhao L, Li J, Usman M, Zhao L, Rehman A, Zad OD, Wang C. In vitro and in silico Xanthine Oxidase Inhibitory Activity of Selected Phytochemicals Widely Present in Various Edible Plants. Comb Chem High Throughput Screen 2020; 23:917-930. [PMID: 32342806 DOI: 10.2174/1386207323666200428075224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 11/22/2022]
Abstract
AIM AND OBJECTIVE The present study was designed to evaluate the xanthine oxidase (XO) inhibitory and antioxidant activities of 30 bioactive compounds present in edible food plants for the possible treatment of hyperuricemia. MATERIALS AND METHODS The XO inhibitory, SO and DPPH radical scavenging activities of selected dietary polyphenols were determined by using colorimetric assays. The molecular docking analysis was performed to evaluate the insight into inhibitory mode of action of bioactive compounds against XO. RESULTS The results show that apigenin, galangin, kaempferol, quercetin, genistein and resveratrol potently inhibit XO enzyme among all tested compounds. Flavonoids exhibit higher, anthocyanins and hydroxycinnamic acids moderate, maslinic acid, ellagic acid, salicylic acid, [6]-gingerol and flavan-3-ols showed weak XO inhibitory activity. The results of molecular docking study revealed that these bioactive compounds bind with the active site of XO and occupy the active site which further prevents the entrance of substrate and results in the inhibition of XO. CONCLUSION Inhibition of XO gives a robust biochemical basis for management of hyperuricemia, gout and other associated diseases via controlling uric acid synthesis.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advance Innovation Center for Food Nutrition and Human Health, School of Food and Chemical Technology, China-Canada Joint Laboratory for Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Ashfaq Ur Rehman
- Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Science and Biotechnology, College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Muhammad Ishaq
- Beijing Advance Innovation Center for Food Nutrition and Human Health, School of Food and Chemical Technology, China-Canada Joint Laboratory for Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Liang Zhao
- Beijing Advance Innovation Center for Food Nutrition and Human Health, School of Food and Chemical Technology, China-Canada Joint Laboratory for Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Jiayi Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Muhammad Usman
- Beijing Advance Innovation Center for Food Nutrition and Human Health, School of Food and Chemical Technology, China-Canada Joint Laboratory for Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Lei Zhao
- Beijing Advance Innovation Center for Food Nutrition and Human Health, School of Food and Chemical Technology, China-Canada Joint Laboratory for Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Abdur Rehman
- State Key Laboratory of Food Science, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Oumeddour D Zad
- Beijing Advance Innovation Center for Food Nutrition and Human Health, School of Food and Chemical Technology, China-Canada Joint Laboratory for Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Chengtao Wang
- Beijing Advance Innovation Center for Food Nutrition and Human Health, School of Food and Chemical Technology, China-Canada Joint Laboratory for Food Nutrition and Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| |
Collapse
|
28
|
Zhao J, Huang L, Sun C, Zhao D, Tang H. Studies on the structure-activity relationship and interaction mechanism of flavonoids and xanthine oxidase through enzyme kinetics, spectroscopy methods and molecular simulations. Food Chem 2020; 323:126807. [PMID: 32330646 DOI: 10.1016/j.foodchem.2020.126807] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
In this study, some flavonoids were screened as potent xanthine oxidase (XO) inhibitors in vitro. Flavonoid 9 was demonstrated to exhibit the inhibitory activity through a ping-pong mechanism. Further structure-activity relationship revealed that different structural elements had greatly influenced the inhibition effect on XO and underlined the requirement of hydroxyl groups at C5 and C4' of flavonoid type I. Moreover, some bioactive flavonoids could efficiently quench the intrinsic fluorescence of XO by either static or static-dynamic mixed mechanism. The synchronous fluorescence, ANS-binding fluorescence, Fourier transform infrared spectra and circular dichroism suggested that active flavonoids could bind to the active center of XO, prevent the entrance of substrate, and induce the rearrangement and conformation change of its secondary structures, ultimately resulting in the significant inhibition effect. Additionally, molecular docking further confirmed these conclusions and highlighted the great importance of hydrophobic interactions and hydrogen bonds for the formation of stable complex conformation.
Collapse
Affiliation(s)
- Jie Zhao
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Lin Huang
- Blood Purification Center, Affiliated Yijishan Hospital of Wannan Medical College, Wuhu 241001, PR China
| | - Chunyong Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Dongsheng Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Hongjin Tang
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| |
Collapse
|
29
|
Tang H, Huang L, Sun C, Zhao D. Exploring the structure–activity relationship and interaction mechanism of flavonoids and α-glucosidase based on experimental analysis and molecular docking studies. Food Funct 2020; 11:3332-3350. [DOI: 10.1039/c9fo02806d] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An integrated method was explored to investigate the structure–activity relationship and interaction mechanism between a library of natural flavonoids and α-glucosidase.
Collapse
Affiliation(s)
- Hongjin Tang
- College of Biological and Chemical Engineering
- Anhui Polytechnic University
- Wuhu 241000
- P. R. China
| | - Lin Huang
- Blood Purification Center
- Affiliated Yijishan Hospital of Wannan Medical College
- Wuhu 241001
- P. R. China
| | - Chunyong Sun
- College of Pharmacy
- Shandong University of Traditional Chinese Medicine
- Jinan 250355
- P. R. China
| | - Dongsheng Zhao
- College of Pharmacy
- Shandong University of Traditional Chinese Medicine
- Jinan 250355
- P. R. China
| |
Collapse
|
30
|
Ishaq M, Mehmood A, Ur Rehman A, Dounya Zad O, Li J, Zhao L, Wang C, Hossen I, Naveed M, Lian Y. Antihyperuricemic effect of dietary polyphenol sinapic acid commonly present in various edible food plants. J Food Biochem 2019; 44:e13111. [PMID: 31849075 DOI: 10.1111/jfbc.13111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 11/28/2022]
Abstract
The present study was conducted to evaluate the antihyperuricemic effect of sinapic acid (SA). The results showed that SA potently inhibited xanthine oxidase (XOD) in a dose-dependent manner by entering the enzyme active site and thwarting the entrance of the substrate. These results were further confirmed by the quantum chemical descriptors analysis and 1 H NMR titration analysis. The in vivo results indicated that SA not only has the potential to inhibit serum and hepatic XOD (p < .05), but also remarkably lowered serum and urine uric acid levels at 50 and 100 mg/kg bw. Furthermore, SA regulated serum creatinine and blood urea nitrogen levels to normal and lowered inflammation in the renal tubules. Thus, the utilization of SA as an antihyperuricemic agent may have considerable potential for the development of functional foods for the possible treatment of hyperuricemia. PRACTICAL APPLICATIONS: Plant-derived bioactive compounds have multiple health benefits. The present study assesses the effects of sinapic acid against hyperuricemia. The results suggested that sinapic acid may have a strong protective effect against uric acid-related complications and may be used for the formulation of functional foods. However, further mechanistic studies are required to verify this hypothesis.
Collapse
Affiliation(s)
- Muhammad Ishaq
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, China
| | - Arshad Mehmood
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, China
| | - Ashfaq Ur Rehman
- Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Science and Biotechnology, College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Oumeddour Dounya Zad
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, China
| | - Jiayi Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Lei 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 (BTBU), Beijing, China
| | - Chengtao Wang
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, China
| | - Imam Hossen
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, China
| | - Muhammad Naveed
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University (BTBU), Beijing, China
| | - Yunhe Lian
- Chenguang Biotech Group Co., Ltd., Quzhou, China
| |
Collapse
|
31
|
Tang H, Ma F, Zhao D. Integrated multi-spectroscopic and molecular modelling techniques to probe the interaction mechanism between salvianolic acid A and α‑glucosidase. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 218:51-61. [PMID: 30954797 DOI: 10.1016/j.saa.2019.03.109] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 06/09/2023]
Abstract
α-Glucosidase (AG) is an important drug target for the treatment of type 2 diabetes mellitus in humans due to the potential effect of down regulating glucose absorption in patients. In our previous study, salvianolic acid A (SAA) was found to exhibit potent AG inhibitory activity, whereas the interaction mechanism was still ambiguous. Herein, the interaction mechanism of SAA and AG was investigated by multi-spectroscopic methods along with molecular docking. As a result, it was found that SAA reversibly inhibited AG in a competitive manner with IC50 of 16.44 ± 0.18 μM, and the inhibition belonged to a multi-phase kinetics process with a first-order reaction. The intrinsic fluorescence of AG could be strongly quenched by SAA through a static quenching mechanism. The negative Gibbs free energy change and positive values of enthalpy and entropy change revealed that the binding of SAA to AG was spontaneous and dominated mainly by hydrophobic interactions, and only a single binding site was determined for them. Analysis of synchronous fluorescence, ANS-binding fluorescence, circular dichroism and Fourier transform infrared spectra suggested that the binding of SAA to AG induced rearrangement and conformational changes of the enzyme. Besides, further molecular modelling validated that SAA could bind to the active domain and prevent the entrance of substrate, resulting in the inhibition of AG activity. These findings provide new insights into understanding the interaction mechanism of SAA on AG.
Collapse
Affiliation(s)
- Hongjin Tang
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| | - Fei Ma
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, PR China
| | - Dongsheng Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| |
Collapse
|
32
|
Effect of Salvia miltiorrhiza on acetylcholinesterase: Enzyme kinetics and interaction mechanism merging with molecular docking analysis. Int J Biol Macromol 2019; 135:303-313. [PMID: 31128195 DOI: 10.1016/j.ijbiomac.2019.05.132] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 11/22/2022]
Abstract
Acetylcholinesterase (AchE) serves as an important target for Alzheimer's disease. Salvia miltiorrhiza has been used to treat cardiovascular disease for hundreds of years. However, the interaction between S. miltiorrhiza and AchE is still inadequate. Herein, an integrated method including molecular docking and experimental studies was employed to investigate the interaction. Consequently, some components were screened as potent AchE inhibitors by in silico and in vitro. Among them, miltirone (MT) and salvianolic acid A (SAA) reversibly inhibited AchE in a mixed-competitive manner. Fluorescence data revealed that SAA and salvianolic acid C (SAC) strongly quenched the intrinsic fluorescence of AchE through a static quenching mechanism, and the binding was spontaneous and dominated by hydrophobic interaction inferred by the thermodynamic parameters. The synchronous and ANS-binding fluorescence spectra suggested that SAA and SAC could bind to the enzyme and induce its conformation changes of secondary structures, which was further confirmed by Fourier transform infrared spectra. Meanwhile, molecular docking presented the probable binding modes of inhibitors to AchE and highlighted the key role of hydrophobic interaction and hydrogen bonds for the stability of docking complex. These findings put more insights into understanding the interaction of S. miltiorrhiza chemicals and AchE, as well as Alzheimer's disease.
Collapse
|
33
|
Tang H, Zhao D. Investigation of the interaction between salvianolic acid C and xanthine oxidase: Insights from experimental studies merging with molecular docking methods. Bioorg Chem 2019; 88:102981. [PMID: 31085372 DOI: 10.1016/j.bioorg.2019.102981] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/26/2022]
Abstract
Xanthine oxidase (XO) has emerged as an important target for gout. In our previous study, salvianolic acid C (SAC) was found to show potent XO inhibitory activity, whereas the interaction mechanism was still not clear. Herein, an integrated approach consisting of enzyme kinetics, multi-spectroscopic methods and molecular docking was employed to investigate the interaction between SAC and XO. Consequently, SAC exhibited a rapid and mixed-type inhibition of XO with IC50 of 5.84 ± 0.18 μM. The fluorescence data confirmed that SAC presented a strong fluorescence quenching effect through a static quenching procedure. The values of enthalpy change, entropy change and Gibbs free energy change indicated that their binding was spontaneous and driven mainly by hydrophobic interactions. Analysis of synchronous fluorescence, circular dichroism and fourier transform infrared spectra demonstrated that SAC induced conformational changes of the enzyme. Besides, further molecular docking revealed that SAC occupied the catalytic center resulting in the inhibition of XO activity. This study provides a comprehensive understanding on the interaction mechanism of SAC on XO.
Collapse
Affiliation(s)
- Hongjin Tang
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| | - Dongsheng Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| |
Collapse
|
34
|
Tang H, Ma F, Zhao D, Xue Z. Exploring the effect of salvianolic acid C on α-glucosidase: Inhibition kinetics, interaction mechanism and molecular modelling methods. Process Biochem 2019. [DOI: 10.1016/j.procbio.2019.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Mehmood A, Ishaq M, Zhao L, Safdar B, Rehman AU, Munir M, Raza A, Nadeem M, Iqbal W, Wang C. Natural compounds with xanthine oxidase inhibitory activity: A review. Chem Biol Drug Des 2019; 93:387-418. [PMID: 30403440 DOI: 10.1111/cbdd.13437] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/10/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023]
Abstract
Hyperuricemia (HUA), a disease due to an elevation of body uric acid level and responsible for various diseases such as gout, cardiovascular disorders, and renal failure, is a major ground debate for the medical science these days. Considering the risk factors linked with allopathic drugs for the treatment of this disease, the debate has now become a special issue. Previously, we critically discussed the role of dietary polyphenols in the treatment of HUA. Besides dietary food plants, many researchers figure out the tremendous effects of medicinal plants-derived phytochemicals against HUA. Keeping in mind all these aspects, we reviewed all possible managerial studies related to HUA through medicinal plants (isolated compounds). In the current review article, we comprehensively discussed various bioactive compounds, chemical structures, and structure-activity relationship with responsible key enzyme xanthine oxidase.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| | - Muhammad Ishaq
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| | - Lei Zhao
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| | - Bushra Safdar
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| | - Ashfaq-Ur Rehman
- Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Science and Biotechnology, College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Masooma Munir
- Food Science Research Institute, National Agricultural Research Centre, Islamabad, Pakistan.,Institute of Food Science and Nutrition, University of Sargodha, Sargodha, Pakistan
| | - Ali Raza
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| | - Muhammad Nadeem
- Institute of Food Science and Nutrition, University of Sargodha, Sargodha, Pakistan
| | - Waheed Iqbal
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
| | - Chengtao Wang
- Beijing Advance Innovation center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, School of Food and Chemical Technology, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
36
|
Screening of xanthine oxidase inhibitor from selected edible plants and hypouricemic effect of Rhizoma Alpiniae Officinarum extract on hyperuricemic rats. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.09.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
37
|
Inhibition mechanism of baicalein and baicalin on xanthine oxidase and their synergistic effect with allopurinol. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.10.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
Identification of the free phenolic profile of Adlay bran by UPLC-QTOF-MS/MS and inhibitory mechanisms of phenolic acids against xanthine oxidase. Food Chem 2018; 253:108-118. [DOI: 10.1016/j.foodchem.2018.01.139] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/23/2017] [Accepted: 01/22/2018] [Indexed: 12/11/2022]
|
39
|
Li M, Yu Y, Liu J, Chen Z, Cao S. Investigation of the interaction between benzaldehyde thiosemicarbazone compounds and xanthine oxidase. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2018.01.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
40
|
Ding H, Hu X, Xu X, Zhang G, Gong D. Inhibitory mechanism of two allosteric inhibitors, oleanolic acid and ursolic acid on α-glucosidase. Int J Biol Macromol 2018; 107:1844-1855. [PMID: 29030193 DOI: 10.1016/j.ijbiomac.2017.10.040] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/19/2017] [Accepted: 10/09/2017] [Indexed: 11/28/2022]
Abstract
Glycemic control which can be efficaciously regulated by inhibiting α-glucosidase activity is an effective therapy for diabetes mellitus. This work is to investigate the kinetics and inhibition mechanism of oleanolic acid and ursolic acid on α-glucosidase. Oleanolic acid and ursolic acid exhibited potent inhibitory activities with IC50 values of (6.35±0.02)×10-6 and (1.69±0.03)×10-5molL-1 respectively in a reversible and non-competitive manner. Both of them binding to α-glucosidase induced the conformational change and intrinsic fluorescence quenching of α-glucosidase. The binding constants of oleanolic acid and ursolic acid with α-glucosidase at 298K were (2.04±0.02)×103 and (1.87±0.02)×103Lmol-1, respectively. Docking results showed that oleanolic acid and ursolic acid bound in different allosteric sites of cavity 2 and cavity 4 on α-glucosidase, respectively, which triggered allosteric regulation to perturb conformational dynamics of α-glucosidase, eventually leading to a decrease of catalytic activity of the enzyme. The substrate was not catalyzed by α-glucosidase to generate further products due to formation of a nonreactive ternary complex of oleanolic acid- or ursolic acid-α-glucosidase-substrate. The combination of oleanolic acid and ursolic acid displayed a significant synergistic inhibition on α-glucosidase.
Collapse
Affiliation(s)
- Huafang Ding
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xing Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Ximing Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Info- rmation Engineering, Jiangsu University of Technology, Changzhou 213001, China
| | - Guowen Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| | - Deming Gong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; New Zealand Institute of Natural Medicine Research, 8 Ha Crescent, Auckland 2104, New Zealand
| |
Collapse
|
41
|
Mehmood A, Zhao L, Wang C, Nadeem M, Raza A, Ali N, Shah AA. Management of hyperuricemia through dietary polyphenols as a natural medicament: A comprehensive review. Crit Rev Food Sci Nutr 2017; 59:1433-1455. [PMID: 29278921 DOI: 10.1080/10408398.2017.1412939] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hyperuricemia, a condition due to high serum uric acid level and is notorious to health. It is considered to be a potent risk factor for gout and dramatically associated in the development of many chronic diseases such as malignant tumor, cardiovascular disorders and renal failure. Modern innovative medicinal and therapeutic interventions are underlying these days to combat hyperuricemia. Previously reported studies revealed the significant impact of dietary polyphenols (e.g. anthocyanins, phenolic acids, flavonoids etc.) against hyperurecemia disorder. Dietary plant polyphenols, unlike anti- hyperuricemic agents, are not reported to have any side effects in curing hyperuricemia. The current comprehensive review figure outs the use of dietary polyphenols as a natural remedy for the management of hyperuricemia. The sources, affiliated pathways, mode of actions and factors affecting their efficiency to prevent hyperuricemia are deeply discussed in this article. Additionally, limitations and suggestions regarding previously reported studies are also highlighted.
Collapse
Affiliation(s)
- Arshad Mehmood
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China.,c Institute of Food Science and Nutrition, University of Sargodha , Sargodha , Pakistan
| | - Lei Zhao
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Chengtao Wang
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Muhammad Nadeem
- c Institute of Food Science and Nutrition, University of Sargodha , Sargodha , Pakistan
| | - Ali Raza
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Nawazish Ali
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Amjad Abbas Shah
- c Institute of Food Science and Nutrition, University of Sargodha , Sargodha , Pakistan
| |
Collapse
|