51
|
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: 1.8] [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
|
52
|
Zhu JX, Yang HY, Hu WQ, Cheng J, Liu Y, Yi LT, Cheng HY. Active components from Lagotis brachystachya maintain uric acid homeostasis by inhibiting renal TLR4-NLRP3 signaling in hyperuricemic mice. Inflammopharmacology 2021; 29:1187-1200. [PMID: 34244900 DOI: 10.1007/s10787-021-00844-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/26/2021] [Indexed: 01/13/2023]
Abstract
Lagotis brachystachya Maxim is a herb widely used in traditional Tibetan medicine. Our previous study indicated that total extracts from Lagotis brachystachya could lower uric acid levels. This study aimed to further elucidate the active components (luteolin, luteoloside and apigenin) isolated from Lagotis brachystachya and the underlying mechanism in vitro and in vivo. The results showed that treatment with luteolin and luteoloside reversed the reduction of organic anion transporter 1 (OAT1) levels, while apigenin attenuated the elevation of urate transporter 1 (URAT1) and glucose transporter 9 (GLUT9) levels in uric acid-treated HK-2 cells, which was consistent with the finding in the kidneys of potassium oxonate (PO)-induced mice. On the other hand, hepatic xanthine oxidase activity was inhibited by the components. In addition, all of these active components improved the morphology of the kidney in hyperuricemic mice. Moreover, molecular docking showed that luteolin, luteoloside and apigenin could bind Toll-like receptor 4 (TLR4) and NLR family pyrin domain containing 3 (NLRP3). Congruently, western blot analysis showed that the components inhibited TLR4/myeloid differentiation primary response 88 (MyD88)/NLRP3 signaling. In conclusion, these results indicated that luteolin, luteoloside and apigenin could attenuate hyperuricemia by decreasing the production and increasing the excretion of uric acid, which were mediated by inhibiting inflammatory signaling pathways.
Collapse
Affiliation(s)
- Ji-Xiao Zhu
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, People's Republic of China
| | - Hai-Yan Yang
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, People's Republic of China
| | - Wei-Qiong Hu
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, People's Republic of China
| | - Jie Cheng
- Department of Chemical and Pharmaceutical Engineering, Huaqiao University, Fujian province, Xiamen, 361021, People's Republic of China
| | - Yang Liu
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, People's Republic of China
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, Huaqiao University, Fujian province, Xiamen, 361021, People's Republic of China.
| | - Hong-Yu Cheng
- School of Humanities, Jiangxi University of Chinese Medicine, Jiangxi province, Nanchang, 330004, People's Republic of China.
| |
Collapse
|
53
|
Nguyen HT, Vu TY, Dakal TC, Dhabhai B, Nguyen XHQ, Tatipamula VB. Cleroda-4(18),13-dien-15,16-olide as novel xanthine oxidase inhibitors: An integrated in silico and in vitro study. PLoS One 2021; 16:e0253572. [PMID: 34191831 PMCID: PMC8244894 DOI: 10.1371/journal.pone.0253572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/08/2021] [Indexed: 11/18/2022] Open
Abstract
In the present study, in silico predictions and molecular docking were performed on five clerodane diterpenes (1-5) from Polyalthia longifolia seeds to evaluate their potential as xanthine oxidase (XO) inhibitors. The initial screening was conducted by target prediction using TargetNet web server application and only compounds 3 and 4 showed a potential interaction with XO. Compounds 3 and 4 were subsequently subjected to in silico analyses on XO protein structure (PDB: 1N5X) using Schrödinger Release 2020-3 followed by structural modeling & molecular simulation studies to confirm the initial prediction result and identify the binding mode of these compounds to the XO. Molecular docking results revealed that compounds 3 (-37.3 kcal/mol) and 4 (-32.0 kcal/mol) binds more stably to XO than the reference drug allopurinol (-27.0 kcal/mol). Interestingly, two residues Glu 802 and Thr 1010 were observed as the two main H-bond binding sites for both tested compounds and the allopurinol. The center scaffold of allopurinol was positioned by some π-π stacking with Phe 914 and Phe 1009, while that of compounds 3 and 4 were supported by many hydrophobic interactions mainly with Leu 648, Phe 649, Phe 1013, and Leu 1014. Additionally, the docking simulation predicted that the inhibitory effect of compounds 3 and 4 was mediated by creating H-bond with particularly Glu 802, which is a key amino acid for XO enzyme inhibition. Altogether, in vitro studies showed that compounds 3 and 4 had better inhibitory capacity against XO enzyme with IC50 values significantly (p < 0.001) lower than that of allopurinol. In short, the present study identified cleroda-4(18),13-dien-15,16-olide as novel potential XO inhibitors, which can be potentially used for the treatment of gout.
Collapse
Affiliation(s)
- Ha Thi Nguyen
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- Faculty of Medicine, Duy Tan University, Da Nang, Vietnam
| | - Thien-Y Vu
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- * E-mail: (TYV); (VBT)
| | - Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Bhanupriya Dhabhai
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | | | - Vinay Bharadwaj Tatipamula
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- Faculty of Medicine, Duy Tan University, Da Nang, Vietnam
- * E-mail: (TYV); (VBT)
| |
Collapse
|
54
|
Li L, Li Y, Luo J, Jiang Y, Zhao Z, Chen Y, Huang Q, Zhang L, Wu T, Pang J. Resveratrol, a novel inhibitor of GLUT9, ameliorates liver and kidney injuries in a D-galactose-induced ageing mouse model via the regulation of uric acid metabolism. Food Funct 2021; 12:8274-8287. [PMID: 34180933 DOI: 10.1039/d1fo00538c] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Accumulating evidence has shown that chronic injection of d-galactose (d-gal) can mimic natural ageing and induce liver and kidney injury. Previous studies showed that d-gal increased uric acid (UA) levels in mice. The increase in UA levels caused inflammation, accelerated oxidative stress, and aggravated liver and kidney injury. Oxidative stress and inflammation play vital roles in the ageing process. Therefore, reducing the levels of UA in ageing mice improved liver and kidney injury. Glucose transporter 9 (GLUT9) is responsible for the reabsorption of UA in the body, and its inhibition helps downregulate UA levels. The present study investigated the UA-lowering activity of the GLUT9 inhibitor resveratrol (RSV) using the patch clamping technique established in our laboratory in vitro. This research is the first study to demonstrate that RSV effectively inhibits UA uptake via GLUT9 (IC50 = 68.77 μM) in vitro. An in vivo study was also performed to investigate the possible protective effect of RSV on d-gal-induced liver and kidney injury. RSV significantly reduced serum UA levels via the downregulation of GLUT9 mRNA and protein expression and promoted the excretion of excess UA through urine. Biochemical analysis showed that RSV significantly downregulated abnormal increases in serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN) and creatinine (CRE) caused by long-term d-gal treatment, which effectively improved pathological damage, increased superoxide dismutase (SOD) activity and decreased the content of malondialdehyde (MDA) in the liver and kidneys. RSV also downregulated the expression of the inflammatory cytokines, interleukin IL-6, IL-1β and tumor necrosis factor (TNF)-α in the liver and kidneys of ageing mice. Our findings provide new insights into the treatment strategies for ageing-induced liver and kidney injury and reveal a new mechanism of RSV-induced reduction in UA levels in ageing individuals.
Collapse
Affiliation(s)
- Lu Li
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yongmei Li
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Jian Luo
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yanqing Jiang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Zean Zhao
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yanyu Chen
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Qinghua Huang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Leqi Zhang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Ting Wu
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
55
|
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: 2.0] [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
|
56
|
Antihyperuricemic, Anti-Inflammatory and Antihypertensive Effect of a Dry Extract from Solidago virgaurea L. (Asteraceae). Sci Pharm 2021. [DOI: 10.3390/scipharm89020027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Solidago virgaurea L. is a perennial plant used in European traditional medicine as a diuretic or a remedy for inflammatory conditions of the urinary tract but also for gout, especially in the Balkans. The present study was focused on a preclinical, in vivo evaluation of antihyperuricemic, anti-inflammatory, and antihypertensive effects of a dry extract from S. virgaurea L. (ESV). Colorimetric and HPLC–MS techniques were used to identify the main chemical constituents of ESV. Antihyperuricemic effect of ESV was assessed in a rat model of hyperuricemia induced by the administration of potassium oxonate. Antihypertensive effect of ESV was evaluated in hyperuricemic rats by monitoring systolic blood pressure with a non-invasive blood-pressure recording system. The anti-inflammatory effect of ESV was tested using a rat model of paw edema. The main chemical constituents of ESV were rutin and phenolic acids represented by chlorogenic and caffeic acid. ESV demonstrated significant antihyperuricemic effects in rats due to an uricosuric mechanism. Additionally, ESV reduced the progression of arterial hypertension in hyperuricemic rats and also showed anti-inflammatory properties slightly inferior to diclofenac. The results suggest that ESV could be a natural remedy for the treatment of gout and protection against endothelial dysfunction caused by hyperuricemia.
Collapse
|
57
|
Wang YZ, Zhou C, Zhu LJ, He XLS, Li LZ, Zheng X, Xu WF, Dong YJ, Li B, Yu QX, Lv GY, Chen SH. Effects of Macroporous Resin Extract of Dendrobium officinale Leaves in Rats with Hyperuricemia Induced by Fructose and Potassium Oxonate. Comb Chem High Throughput Screen 2021; 25:1294-1303. [PMID: 34053424 DOI: 10.2174/1386207324666210528114345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/09/2021] [Accepted: 04/04/2021] [Indexed: 11/22/2022]
Abstract
AIM AND OBJECTIVE Fructose, as a ubiquitous monosaccharide, can promote ATP consumption and elevate circulating uric acid (UA) levels. Our previous studies confirmed that the macroporous resin extract of Dendrobium officinale leaves (DoMRE) could reduce the UA level of rats with hyperuricemia induced by a high-purine diet. This study aimed to investigate whether DoMRE had a UA-lowering effect on rats with hyperuricemia caused by fructose combined with potassium oxonate, so as to further clarify the UA-lowering effect of DoMRE, and to explore the UA-lowering effect of DoMRE on both UA production and excretion. MATERIALS AND METHODS Rats with hyperuricemia induced by fructose and potassium oxonate were administered with DoMRE and vehicle control, respectively, to compare the effects of the drugs. At the end of the experiment, the serum uric acid (SUA) and creatinine (Cr) levels were measured using an automatic biochemical analyzer, the activities of xanthine oxidase (XOD) were measured using an assay kit, and the protein expression of urate transporter 1 (URAT1), glucose transporter 9 (GLUT9), and ATP-binding cassette superfamily G member 2 (ABCG2) were assessed using immunohistochemical and western blot analyses. Hematoxylin and eosin staining was used to assess the histological changes in the kidney, liver, and intestine. RESULTS Rats with hyperuricemia were induced by fructose and potassiumFructose and potassium induced hyperuricemia in rats. Meanwhile, the activities of XOD were markedly augmented, the expression of URAT1 and GLUT9 was promoted, and the expression of ABCG2 was reduced, which were conducive to the elevation of UA. However, exposure to DoMRE reversed these fructose- and potassium oxonate-induced negative alternations in rats. The activities of XOD were recovered to the normal level, reducing UA formation; the expression of URAT1, ABCG2, and GLUT9 returned to the normal level, resulting in an increase in renal urate excretion. CONCLUSION DoMRE reduces UA levels in rats with hyperuricemia induced by fructose combined with potassium oxonate by inhibiting XOD activity and regulating the expression of ABCG2, URAT1, and GLUT9. DoMRE is a potential therapeutic agent for treating hyperuricemia through inhibiting UA formation and promoting UA excretion.
Collapse
Affiliation(s)
- Yu-Zhi Wang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Cong Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Li-Jie Zhu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xing-Li-Shang He
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Lin-Zi Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xiang Zheng
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Wan-Feng Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ying-Jie Dong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Bo Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qiao-Xian Yu
- Zhejiang Senyu Co., Ltd, Yiwu, Zhejiang, 322099, China
| | - Gui-Yuan Lv
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Su-Hong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
58
|
Novel Quercetin Derivative of 3,7-Dioleylquercetin Shows Less Toxicity and Highly Potent Tyrosinase Inhibition Activity. Int J Mol Sci 2021; 22:ijms22084264. [PMID: 33923988 PMCID: PMC8072539 DOI: 10.3390/ijms22084264] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/17/2021] [Accepted: 04/18/2021] [Indexed: 12/14/2022] Open
Abstract
Quercetin is a well-known plant flavonol and antioxidant; however, there has been some debate regarding the efficacy and safety of native quercetin as a skin-whitening agent via tyrosinase inhibition. Several researchers have synthesized quercetin derivatives as low-toxicity antioxidants and whitening agents. However, no suitable quercetin derivatives have been reported to date. In this study, a novel quercetin derivative was synthesized by the SN2 reaction using quercetin and oleyl bromide. The relationship between the structures and activities of quercetin derivatives as anti-melanogenic agents was assessed using in vitro enzyme kinetics, molecular docking, and quenching studies; cell line experiments; and in vivo zebrafish model studies. Novel 3,7-dioleylquercetin (OQ) exhibited a low cytotoxic concentration level at >100 µg/mL (125 µM), which is five times less toxic than native quercetin. The inhibition mechanism showed that OQ is a competitive inhibitor, similar to native quercetin. Expression of tyrosinase, tyrosinase-related protein 1 (TRP-1) and tyrosinase-related protein 2 (TRP-2), and microphthalmia-associated transcription factor was inhibited in B16F10 melanoma cell lines. mRNA transcription levels of tyrosinase, TRP-1, and TRP-2 decreased in a dose-dependent manner. Melanin formation was confirmed in the zebrafish model using quercetin derivatives. Therefore, OQ might be a valuable asset for the development of novel skin-whitening agents.
Collapse
|
59
|
Xie F, Zhang W, Gong S, Wang Z. Inhibitory effect of lignin from Canna edulis Ker residues on trypsin: kinetics and molecular docking studies. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:2090-2099. [PMID: 32978811 DOI: 10.1002/jsfa.10831] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/19/2020] [Accepted: 09/25/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Lignin extracted from Canna edulis Ker residues shows a strong inhibitory effect on α-glucosidase and a promoting effect on α-amylase. Protease activity inhibition may play a key role in disease processes, such as metastasis, tumor invasion and bacterial colonization. Hence, in the present study, the inhibitory mechanism of lignin on trypsin was examined, including the interaction type, thermodynamic parameters, structure, reaction site and molecular docking. RESULTS The isolated lignin presented an inhibitory effect on trypsin activity with an IC50 value of 1.35 μmol L-1 . This inhibition was a mixed linear type with a constant Ki of 3.92 μmol L-1 . The lignin could bind with the key amino acid residue Ser195 on the active site of the trypsin molecule to inhibit its activity, and the phenolic hydroxyl group and -OH on the β-O-4 structure of the lignin molecule were the major groups bound with trypsin. CONCLUSION These results illustrate the inhibitory effects of Canna edulis residue lignin on protease, which helps with respect to understanding the possible application of lignin in the food industry in functional foods. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Fan Xie
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wei Zhang
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Shengxiang Gong
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengwu Wang
- Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
60
|
Gainche M, Ogeron C, Ripoche I, Senejoux F, Cholet J, Decombat C, Delort L, Berthon JY, Saunier E, Caldefie Chezet F, Chalard P. Xanthine Oxidase Inhibitors from Filipendula ulmaria (L.) Maxim. and Their Efficient Detections by HPTLC and HPLC Analyses. Molecules 2021; 26:1939. [PMID: 33808360 PMCID: PMC8038090 DOI: 10.3390/molecules26071939] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/28/2022] Open
Abstract
Filipendula ulmaria is a plant commonly used for the treatment of several pathologies, such as diarrhoea, ulcers, pain, stomach aches, fevers, and gout. Our study focused on the use of F. ulmaria for the treatment of gout disease. We first studied the chemical composition of a methanolic extract of the aerial parts and demonstrated its xanthine oxidase (XO) inhibitory activity. Then, we performed a fractionation and evaluated the most XO inhibitory active fractions by UV measurement. Purification of some fractions allowed the determination of the inhibitory activity of pure compounds. We demonstrated that spiraeoside, a glycosylated flavonoid, possesses an activity around 25 times higher than allopurinol, used as a reference in the treatment of gout disease. In order to easily and quickly identify potent inhibitors in complex matrix, we developed a complementary strategy based on an HPLC method and an Effect Directed Assay (EDA) method combining HPTLC and biochemical assays. The HPLC method, capable of determining compounds exhibiting interactions with the enzyme, could be an efficient strategy for evaluating potent enzyme inhibitors in a complex mixture. This strategy could be applied for quantitative assays using LC/MS experiments.
Collapse
Affiliation(s)
- Maël Gainche
- Clermont Auvergne INP, Université Clermont Auvergne, CNRS, ICCF, F-63000 Clermont-Ferrand, France; (I.R.); (P.C.)
| | - Clémence Ogeron
- Université Clermont-Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France; (C.O.); (F.S.); (J.C.); (C.D.); (L.D.); (F.C.C.)
| | - Isabelle Ripoche
- Clermont Auvergne INP, Université Clermont Auvergne, CNRS, ICCF, F-63000 Clermont-Ferrand, France; (I.R.); (P.C.)
| | - François Senejoux
- Université Clermont-Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France; (C.O.); (F.S.); (J.C.); (C.D.); (L.D.); (F.C.C.)
| | - Juliette Cholet
- Université Clermont-Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France; (C.O.); (F.S.); (J.C.); (C.D.); (L.D.); (F.C.C.)
| | - Caroline Decombat
- Université Clermont-Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France; (C.O.); (F.S.); (J.C.); (C.D.); (L.D.); (F.C.C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France; (C.O.); (F.S.); (J.C.); (C.D.); (L.D.); (F.C.C.)
| | | | - Etienne Saunier
- Dômes Pharma, 3 Rue André Citroën, 63430 Pont-du-Château, France;
| | - Florence Caldefie Chezet
- Université Clermont-Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France; (C.O.); (F.S.); (J.C.); (C.D.); (L.D.); (F.C.C.)
| | - Pierre Chalard
- Clermont Auvergne INP, Université Clermont Auvergne, CNRS, ICCF, F-63000 Clermont-Ferrand, France; (I.R.); (P.C.)
| |
Collapse
|
61
|
Yu S, He M, Zhai Y, Xie Z, Xu S, Yu S, Xiao H, Song Y. Inhibitory activity and mechanism of trilobatin on tyrosinase: kinetics, interaction mechanism and molecular docking. Food Funct 2021; 12:2569-2579. [PMID: 33625428 DOI: 10.1039/d0fo03264f] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tyrosinase is the rate-limiting enzyme controlling the production of melanin, and tyrosinase inhibitors can regulate the overproduction of melanin by inhibiting tyrosinase activity, which is an effective method to treat pigmentation disorders. In this study, kinetic analysis, multispectroscopic methods and molecular simulation were used to investigate the inhibitory activity and mechanism of trilobatin on tyrosinase. The kinetic analysis showed that trilobatin had significant inhibitory activity on tyrosinase in a reversible and mixed-type manner with IC50 values of (2.24 ± 0.35) × 10-5 mol L-1. The intrinsic fluorescence of tyrosinase was quenched by trilobatin through a static quenching mechanism. Different spectroscopic measurements demonstrated that trilobatin could change the microenvironments and conformation of tyrosinase and molecular docking determined the binding site of quercetin on tyrosinase.
Collapse
Affiliation(s)
- Shuyan Yu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China.
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Wang J, Sun S, Zhao K, Shi H, Fan J, Wang H, Liu Y, Liu X, Wang W. Insights into the inhibitory mechanism of purpurogallin on xanthine oxidase by multiple spectroscopic techniques and molecular docking. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
63
|
Xu L, Zhu L, Dai Y, Gao S, Wang Q, Wang X, Chen X. Impact of yeast fermentation on nutritional and biological properties of defatted adlay (Coix lachryma-jobi L.). Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2020.110396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
64
|
Chen L, Zhao X, He Y, Yang H. Cloning, purification and characterisation of cytosolic fructose-1,6-bisphosphatase from mung bean (Vigna radiata). Food Chem 2021; 347:128973. [PMID: 33444888 DOI: 10.1016/j.foodchem.2020.128973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 11/26/2020] [Accepted: 12/28/2020] [Indexed: 10/22/2022]
Abstract
To improve the crop yield and quality, the cytosolic fructose-1,6-bisphosphatase (cFBPase) from mung bean (Vigna radiata), a rate-limiting enzyme in gluconeogenesis, was cloned, purified, and structurally characterised. To function it required Mg2+ and Mn2+ at 0.01-10 mM. The Michaelis-Menton constant and adenosine monophosphate (AMP) inhibitory constant (Ki) were 7.96 and 111.09 μM, respectively. The functional site residues of AMP binding (Arg30, Asp32, and Phe33) and the active site residues (Asn218 and Met251) were tested via site-directed mutagenesis and molecular docking. Asn218 and Met251 were replaced by Tyr and Leu, respectively. The M251L mutant showed enhanced substrate affinity and activity, resulting from decreased binding energy (-2.58 kcal·mol-1) and molecular distance (4.2 Å). AMP binding site mutations changed the enzyme activities, indicating a connection between the binding and active sites. Furthermore, Ki and docking analysis revealed that Asp32 plays a key role in maintaining the AMP binding conformation.
Collapse
Affiliation(s)
- Lin Chen
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore; National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Suzhou, Jiangsu 215123, PR China
| | - Xue Zhao
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore; National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Suzhou, Jiangsu 215123, PR China
| | - Yun He
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore; National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Suzhou, Jiangsu 215123, PR China
| | - Hongshun Yang
- Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore; National University of Singapore (Suzhou) Research Institute, 377 Lin Quan Street, Suzhou Industrial Park, Suzhou, Jiangsu 215123, PR China.
| |
Collapse
|
65
|
Chen Y, Zhao Z, Li Y, Yang Y, Li L, Jiang Y, Lin C, Cao Y, Zhou P, Tian Y, Wu T, Pang J. Baicalein alleviates hyperuricemia by promoting uric acid excretion and inhibiting xanthine oxidase. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 80:153374. [PMID: 33075645 DOI: 10.1016/j.phymed.2020.153374] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/25/2020] [Accepted: 10/08/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Insufficient renal urate excretion and/or overproduction of uric acid (UA) are the dominant causes of hyperuricemia. Baicalein (BAL) is widely distributed in dietary plants and has extensive biological activities, including antioxidative, anti-inflammatory and antihypertensive activities. PURPOSE To investigate the anti-hyperuricemic effects of BAL and the underlying mechanisms in vitro and in vivo. METHODS We investigated the inhibitory effects of BAL on GLUT9 and URAT1 in vitro through electrophysiological experiments and 14C-urate uptake assays. To evaluate the impact of BAL on serum and urine UA, the expression of GLUT9 and URAT1, and the activity of xanthine oxidase (XOD), we developed a mouse hyperuricemia model by potassium oxonate (PO) injection. Molecular docking analysis based on homology modeling was performed to explain the predominant efficacy of BAL compared with the other test compounds. RESULTS BAL dose-dependently inhibited GLUT9 and URAT1 in a noncompetitive manner with IC50 values of 30.17 ± 8.68 μM and 31.56 ± 1.37 μM, respectively. BAL (200 mg/kg) significantly decreased serum UA and enhanced renal urate excretion in PO-induced hyperuricemic mice. Moreover, the expression of GLUT9 and URAT1 in the kidney was downregulated, and XOD activity in the serum and liver was suppressed. The docking analysis revealed that BAL potently interacted with Trp336, Asp462, Tyr71 and Gln328 of GLUT9 and Ser35 and Phe241 of URAT1. CONCLUSION These results indicated that BAL exerts potent antihyperuricemic efects through renal UA excretal promotion and serum UA production. Thus, we propose that BAL may be a promising treatment for the prevention of hyperuricemia owing to its multitargeted inhibitory activity.
Collapse
Affiliation(s)
- Yanyu Chen
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zean Zhao
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yongmei Li
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yang Yang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lu Li
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yu Jiang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Cuiting Lin
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ying Cao
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Pingzheng Zhou
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yuanxin Tian
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ting Wu
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
66
|
Balaei F, Ansari M, Farhadian N, Moradi S, Shahlaei M. Interactions and effects of food additive dye Allura red on pepsin structure and protease activity; experimental and computational supports. Res Pharm Sci 2020; 16:58-70. [PMID: 33953775 PMCID: PMC8074803 DOI: 10.4103/1735-5362.305189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 06/07/2020] [Accepted: 12/13/2020] [Indexed: 11/18/2022] Open
Abstract
Background and purpose: Today, color additives such as Allura red (AR) are widely used in different kinds of food products. Pepsin is a globular protein that is secreted as a digestive protease from the main cells in the stomach. Because of the important role of pepsin in protein digestion and because of its importance in digestive diseases the study of the interactions of pepsin with chemical food additives is important. Experimental approach: In this study, the interactions between AR and pepsin were investigated by different computational and experimental approaches such as ultraviolet and fluorescence spectroscopy along with computational molecular modeling. Findings/Results: The experimental results of fluorescence indicated that AR can strongly quench the fluorescence of pepsin through a static quenching. Thermodynamic analysis of the binding phenomena suggests that van der Waals forces and hydrogen bonding played a major role in the complex formation. The results of synchronous fluorescence spectra and furrier transformed infra-red (FTIR) experiments showed that there are no significant structural changes in the protein conformation. Also, examined pepsin protease activity revealed that the activity of pepsin was increased upon ligand binding. In agreement with the experimental results, the computational results showed that hydrogen bonding and van der Waals interactions occurred between AR and binding sites. Conclusion and implications: From the pharmaceutical point of view, this interaction can help us to get a deeper understanding of the effect of this synthetic dye on food digestion.
Collapse
Affiliation(s)
- Fatemeh Balaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| | - Mohabbat Ansari
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| | - Negin Farhadian
- Substance Abuse Prevention Research Center, Health Institute Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| | - Mohsen Shahlaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| |
Collapse
|
67
|
Xie F, Gong S, Zhang W, Wang Z. Kinetics and Molecular Docking Studies of Activating Effect of
Canna edulis
Ker Residue Lignin on the Activity of Lipase. STARCH-STARKE 2020. [DOI: 10.1002/star.202000154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Fan Xie
- Shanghai Engineering Research Center of Food Microbiology School of Medical Instrument and Food Engineering University of Shanghai for Science and Technology Shanghai 200093 P. R. China
| | - Shengxiang Gong
- Department of Food Science and Technology School of Agriculture and Biology Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Wei Zhang
- Department of Food Science and Technology School of Agriculture and Biology Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| | - Zhengwu Wang
- Department of Food Science and Technology School of Agriculture and Biology Shanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 China
| |
Collapse
|
68
|
An MF, Wang MY, Shen C, Sun ZR, Zhao YL, Wang XJ, Sheng J. Isoorientin exerts a urate-lowering effect through inhibition of xanthine oxidase and regulation of the TLR4-NLRP3 inflammasome signaling pathway. J Nat Med 2020; 75:129-141. [PMID: 33188510 DOI: 10.1007/s11418-020-01464-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/29/2020] [Indexed: 12/21/2022]
Abstract
Isoorientin (ISO), a natural flavonoid compound, has been identified in several plants and its biological activity is determined and the study on lowering uric acid has not been reported. In view of the current status of treatment of hyperuricemia, we evaluated the hypouricemic effects of ISO in vivo and in vitro, and explored the underlying mechanisms. Yeast extract-induced hyperuricemia animal model as well as hypoxanthine and xanthine oxidase (XOD) co-induced high uric acid L-O2 cell model and enzymatic experiments in vitro were selected. The XOD activity and uric acid (UA) level were inhibited after the treatment of ISO in vitro and in vivo. Furthermore, serum creatinine (CRE) and blood urea nitrogen (BUN) levels were also significantly reduced and liver damage was recovered in pathological histology after the ISO administration in hyperuricemia animal model. The results of mechanism illustrated that protein expressions such as XOD, toll-like receptor 4 (TLR4), cathepsin B (CTSB), NLRP3, and its downstream caspase-1 as well as interleukin-18 (IL-18) were markedly downregulated by ISO intervention in vitro and in vivo. Our results suggest that ISO exerts a urate-lowering effect through inhibiting XOD activity and regulating TLR4-NLRP3 inflammasome signal pathway, thus representing a promising candidate therapeutic agent for hyperuricemia. Both animal models and in vitro experiments suggested that ISO may effectively lower uric acid produce. The mechanism might be the inhibition of XOD activity and NLRP3 inflammasome of upregulation.
Collapse
Affiliation(s)
- Meng-Fei An
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
| | - Ming-Yue Wang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
| | - Chang Shen
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
| | - Ze-Rui Sun
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650224, People's Republic of China
| | - Yun-Li Zhao
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650224, People's Republic of China.
- College of Science, Yunnan Agricultural University, Kunming, 650224, People's Republic of China.
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China.
| | - Xuan-Jun Wang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650224, People's Republic of China.
- College of Science, Yunnan Agricultural University, Kunming, 650224, People's Republic of China.
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, 650224, People's Republic of China.
| | - Jun Sheng
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, 650224, People's Republic of China.
- College of Science, Yunnan Agricultural University, Kunming, 650224, People's Republic of China.
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, 650224, People's Republic of China.
| |
Collapse
|
69
|
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: 2.4] [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]
|
70
|
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: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
71
|
Liu P, Xu H, Shi Y, Deng L, Chen X. Potential Molecular Mechanisms of Plantain in the Treatment of Gout and Hyperuricemia Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:3023127. [PMID: 33149752 PMCID: PMC7603577 DOI: 10.1155/2020/3023127] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/13/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND The incidence of gout and hyperuricemia is increasing year by year in the world. Plantain is a traditional natural medicine commonly used in the treatment of gout and hyperuricemia, but the molecular mechanism of its active compounds is still unclear. Based on network pharmacology, this article predicts the targets and pathways of effective components of plantain for gout and hyperuricemia and provides effective reference for clinical medication. METHOD Traditional Chinese medicine systems pharmacology database and analysis platform (TCMSP) and SymMap databases were used to screen out the active compounds and their targets in plantain. GeneCards, Therapeutic Target Database (TTD), and Online Mendelian Inheritance in Man (OMIM) databases were used to find the targets corresponding to gout and hyperuricemia. Venn diagram was used to obtain the intersection targets of plantain and diseases. The interaction network of the plantain active compounds-targets-pathways-diseases was constructed by using Cytoscape 3.7.2 software. Finally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were carried out. RESULT Seven active compounds were identified by network pharmacological analysis, including dinatin, baicalein, baicalin, sitosterol, 6-OH-luteolin, stigmasterol, and luteolin. Plantain plays a role in gout and hyperuricemia diseases by regulating various biological processes, cellular components, and molecular functions. The core targets of plantain for treating gout are MAPK1, RELA, TNF, NFKBIA, and IFNG, and the key pathways are pathways in cancer, hypoxia-inducible factor-1 (HIF-1) signaling pathway, interleukin (IL)-17 signaling pathway, Chagas disease (American trypanosomiasis), and relaxin signaling pathway. The core targets of plantain for hyperuricemia are RELA, MAPK1, NFKBIA, CASP3, CASP8, and TNF, and the main pathways are pathways in cancer, apoptosis, hepatitis B, IL-17 signaling pathway, and toxoplasmosis. CONCLUSION This study explored the related targets and mechanisms of plantain for the treatment of gout and hyperuricemia from the perspective of network pharmacological analysis, reflecting the characteristics of multiple components, multiple targets, and multiple pathways, and it provides a good theoretical basis for the clinical application of plantain.
Collapse
Affiliation(s)
- Pei Liu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Huachong Xu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yucong Shi
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaoyin Chen
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
72
|
Grigore-Gurgu L, Crăciunescu O, Aprodu I, Bolea CA, Iosăgeanu A, Petre BA, Bahrim GE, Oancea A, Stănciuc N. Tailoring the Health-Promoting Potential of Protein Hydrolysate Derived from Fish Wastes and Flavonoids from Yellow Onion Skins: From Binding Mechanisms to Microencapsulated Functional Ingredients. Biomolecules 2020; 10:biom10101416. [PMID: 33036339 PMCID: PMC7600352 DOI: 10.3390/biom10101416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023] Open
Abstract
This study focuses on combining different bioprocessing tools in order to develop an in-depth engineering approach for enhancing the biological properties of two valuable food by-products, namely fish waste and yellow onion skins, in a single new bioactive formulation. Bone tissue from phytophagous carp (Hypophthalmichthys molitrix) was used to obtain bioactive peptides through papain-assisted hydrolysis. The peptides with molecular weight lower than 3 kDa were characterized through MALDI-ToF/ToF mass spectrometry and bioinformatics tools. As a prerequisite for microencapsulation, the ability of these peptides to bind the flavonoids extracted from yellow onion skins was further tested through fluorescence quenching measurements. The results obtained demonstrate a considerable binding potency with a binding value of 106 and also the presence of one single or one class of binding site during the interaction process of flavonoids with peptides, in which the main forces involved are hydrogen bonds and van der Waals interactions. In the freeze-drying microencapsulation process, an efficiency for total flavonoids of 88.68 ± 2.37% was obtained, considering the total flavonoids and total polyphenols from the powder of 75.72 ± 2.58 quercetin equivalents/g dry weight (DW) and 97.32 ± 2.80 gallic acid equivalents/g DW, respectively. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test on the L929 cell line cultivated in the presence of different concentrations of microencapsulated samples (0.05–1.5 mg/mL) proved no sign of cytotoxicity, the cell viability being over 80% for all the samples.
Collapse
Affiliation(s)
- Leontina Grigore-Gurgu
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 111 Domnească Street, 800201 Galați, Romania; (L.G.-G.); (I.A.); (C.A.B.); (G.E.B.)
| | - Oana Crăciunescu
- National Institute of Research and & Development for Biological Sciences, 296 Splaiul Independentei, 060031 Bucharest, Romania; (O.C.); (A.I.); (A.O.)
| | - Iuliana Aprodu
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 111 Domnească Street, 800201 Galați, Romania; (L.G.-G.); (I.A.); (C.A.B.); (G.E.B.)
| | - Carmen Alina Bolea
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 111 Domnească Street, 800201 Galați, Romania; (L.G.-G.); (I.A.); (C.A.B.); (G.E.B.)
| | - Andreea Iosăgeanu
- National Institute of Research and & Development for Biological Sciences, 296 Splaiul Independentei, 060031 Bucharest, Romania; (O.C.); (A.I.); (A.O.)
| | - Brîndușa Alina Petre
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iasi, 700506 Iaşi, Romania;
- Center for Fundamental Research and Experimental Development in Translation Medicine–TRANSCEND, Regional Institute of Oncology, 700483 Iaşi, Romania
| | - Gabriela Elena Bahrim
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 111 Domnească Street, 800201 Galați, Romania; (L.G.-G.); (I.A.); (C.A.B.); (G.E.B.)
| | - Anca Oancea
- National Institute of Research and & Development for Biological Sciences, 296 Splaiul Independentei, 060031 Bucharest, Romania; (O.C.); (A.I.); (A.O.)
| | - Nicoleta Stănciuc
- Faculty of Food Science and Engineering, Dunarea de Jos University of Galati, 111 Domnească Street, 800201 Galați, Romania; (L.G.-G.); (I.A.); (C.A.B.); (G.E.B.)
- Correspondence: ; Tel.: +40-0336-130-183
| |
Collapse
|
73
|
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: 6.0] [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
|
74
|
Chen J, Li Q, Ye Y, Ran M, Ruan Z, Jin N. Inhibition of xanthine oxidase by theaflavin: Possible mechanism for anti-hyperuricaemia effect in mice. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.06.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
75
|
Yang Y, Zhang ZC, Zhou Q, Yan JX, Zhang JL, Su GH. Hypouricemic effect in hyperuricemic mice and xanthine oxidase inhibitory mechanism of dietary anthocyanins from purple sweet potato (Ipomoea batatas L.). J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104151] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
76
|
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: 53] [Impact Index Per Article: 10.6] [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
|
77
|
Kim JY, Wang Y, Li ZP, Baiseitova A, Ban YJ, Park KH. Xanthine Oxidase Inhibition and Anti-LDL Oxidation by Prenylated Isoflavones from Flemingia philippinensis Root. Molecules 2020; 25:molecules25133074. [PMID: 32640700 PMCID: PMC7411605 DOI: 10.3390/molecules25133074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 01/18/2023] Open
Abstract
Xanthine oxidase is a frontier enzyme to produce oxidants, which leads to inflammation in the blood. Prenylated isoflavones from Flemingia philippinensis were found to display potent inhibition against xanthine oxidase (XO). All isolates (1–9) inhibited XO enzyme with IC50 ranging 7.8~36.4 μM. The most active isoflavones (2–5, IC50 = 7.8~14.8 μM) have the structural feature of a catechol motif in B-ring. Inhibitory behaviors were disclosed as a mixed type I mode of inhibition with KI < KIS. Binding affinities to XO enzyme were evaluated. Fluorescence quenching effects agreed with inhibitory potencies (IC50s). The compounds (2–5) also showed potent anti-LDL oxidation effects in the thiobarbituric acid-reactive substances (TBARS) assay, the lag time of conjugated diene formation, relative electrophoretic mobility (REM), and fragmentation of apoB-100 on copper-mediated LDL oxidation. The compound 4 protected LDL oxidation with 0.7 μM in TBARS assay, which was 40-fold more active than genistein (IC50 = 30.4 μM).
Collapse
Affiliation(s)
- Jeong Yoon Kim
- Division of Applied Life Science (BK21 Plus), IALS, Gyeongsang National University, Jinju 52828, Korea; (J.Y.K.); (Z.P.L.); (A.B.); (Y.J.B.)
| | - Yan Wang
- College of Food and Biological Engineering, Qiqihar University, Qiqihar 161006, China;
| | - Zuo Peng Li
- Division of Applied Life Science (BK21 Plus), IALS, Gyeongsang National University, Jinju 52828, Korea; (J.Y.K.); (Z.P.L.); (A.B.); (Y.J.B.)
| | - Aizhamal Baiseitova
- Division of Applied Life Science (BK21 Plus), IALS, Gyeongsang National University, Jinju 52828, Korea; (J.Y.K.); (Z.P.L.); (A.B.); (Y.J.B.)
| | - Yeong Jun Ban
- Division of Applied Life Science (BK21 Plus), IALS, Gyeongsang National University, Jinju 52828, Korea; (J.Y.K.); (Z.P.L.); (A.B.); (Y.J.B.)
| | - Ki Hun Park
- Division of Applied Life Science (BK21 Plus), IALS, Gyeongsang National University, Jinju 52828, Korea; (J.Y.K.); (Z.P.L.); (A.B.); (Y.J.B.)
- Correspondence: ; Tel.: +82-55-772-1965; Fax: +82-55-772-1969
| |
Collapse
|
78
|
Liu S, Yu J, Guo S, Fang H, Chang X. Inhibition of pancreatic α-amylase by Lonicera caerulea berry polyphenols in vitro and their potential as hyperglycemic agents. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2020.109288] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
79
|
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: 1.0] [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
|
80
|
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: 81] [Impact Index Per Article: 16.2] [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
|
81
|
From Xanthine Oxidase Inhibition to In Vivo Hypouricemic Effect: An Integrated Overview of In Vitro and In Vivo Studies with Focus on Natural Molecules and Analogues. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9531725. [PMID: 32184901 PMCID: PMC7060854 DOI: 10.1155/2020/9531725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/10/2019] [Accepted: 12/24/2019] [Indexed: 01/05/2023]
Abstract
Hyperuricemia is characterized by elevated uric acid (UA) levels on blood, which can lead to gout, a common pathology. These high UA levels are associated with increased purine ingestion and metabolization and/or its decreased excretion. In this field, xanthine oxidase (XO), by converting hypoxanthine and xanthine to UA, plays an important role in hyperuricemia control. Based on limitations and adverse effects associated with the use of allopurinol and febuxostat, the most known approved drugs with XO inhibitory effect, the search for new molecules with XO activity is growing. However, despite the high number of studies, it was found that the majority of tested products with relevant XO inhibition were left out, and no further pharmacological evaluation was performed. Thus, in the present review, available information published in the past six years concerning isolated molecules with in vitro XO inhibition complemented with cytotoxicity evaluation as well as other relevant studies, including in vivo hypouricemic effect, and pharmacokinetic/pharmacodynamic profile was compiled. Interestingly, the analysis of data collected demonstrated that molecules from natural sources or their mimetics and semisynthetic derivatives constitute the majority of compounds being explored at the moment by means of in vitro and in vivo animal studies. Therefore, several of these molecules can be useful as lead compounds and some of them can even have the potential to be considered in the future clinical candidates for the treatment of hyperuricemia.
Collapse
|
82
|
Wang J, Zhao J, Yan Y, Liu D, Wang C, Wang H. Inhibition of glycosidase by ursolic acid: in vitro, in vivo and in silico study. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:986-994. [PMID: 31650545 DOI: 10.1002/jsfa.10098] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Controlling the blood glucose level is an effective method to reduce type 2 diabetes and prevent diabetes-related complications. Ursolic acid is a plant extract that can reduce postprandial hyperglycemia effectively. This study aimed to explore the inhibitory effect and interaction mechanism of ursolic acid against α-amylase and α-glucosidase. RESULTS In this study, the effect of ursolic acid on glycosidase was studied in vitro, in vivo, and in silico. The half-maximal inhibitory concentration (IC50 ) of ursolic acid on α-amylase and α-glucosidase was 0.482 ± 0.12 mg mL-1 and 0.213 ± 0.042 mg mL-1 , respectively. The results of enzymatic kinetics showed that ursolic acid inhibited α-amylase and α-glucosidase activity in a non-competitive manner. The fluorescence spectrum showed that the combination of ursolic acid and glycosidase caused the intrinsic fluorescence quenching of glycosidase. The observation of starch granules revealed that the activity of α-amylase was inhibited and the hydrolysis of starch granules was prevented in the presence of ursolic acid. Molecular docking results showed that ursolic acid bound to the inactive site of α-amylase and α-glucosidase through the formation of ursolic acid-glucosidase complex. Ursolic acid interacted with α-amylase and α-glucosidase mainly through hydrogen bonding. The postprandial hypoglycemic effect of ursolic acid in C57BL/6J mice showed that the high concentration of ursolic acid could quickly reduce postprandial blood glucose level. CONCLUSION Ursolic acid can be considered as a natural ingredient in functional foods to control postprandial blood glucose levels and prevent diabetes by delaying the digestion of starch in foods. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing, China
| | - Jiang Zhao
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, China
| | - Yong Yan
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, China
| | - Dong Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, China
| | - Chengtao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing, China
- Beijing Laboratory for Food Quality and Safety, Beijing Technology and Business University, Beijing, China
| | - Hao Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology (TUST), Tianjin, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing, China
| |
Collapse
|
83
|
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: 3.8] [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
|
84
|
Experimental investigation and molecular dynamics simulation of the binding of ellagic acid to bovine liver catalase: Activation study and interaction mechanism. Int J Biol Macromol 2020; 143:850-861. [DOI: 10.1016/j.ijbiomac.2019.09.146] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/15/2019] [Accepted: 09/19/2019] [Indexed: 01/14/2023]
|
85
|
Liu Y, Chen H, Xiang H, Lei H, Zhang D, Qiu Y, Xu L. Inhibition and molecular mechanism of diosmetin against xanthine oxidase by multiple spectroscopies and molecular docking. NEW J CHEM 2020. [DOI: 10.1039/d0nj00679c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Studying the inhibition and molecular mechanism of diosmetin against xanthine oxidase helps to develop natural product xanthine oxidase inhibitors.
Collapse
Affiliation(s)
- Yongjie Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science
- Wuhan Polytechnic University
- Wuhan 430023
- China
- School of Biology and Pharmaceutical Engineering
| | - Hao Chen
- School of Chemistry and Environmental Engineering
- Wuhan Polytechnic University
- Wuhan
- China
| | - Huilong Xiang
- Hubei Key Laboratory of Animal Nutrition and Feed Science
- Wuhan Polytechnic University
- Wuhan 430023
- China
- School of Biology and Pharmaceutical Engineering
| | - Huan Lei
- School of Biology and Pharmaceutical Engineering
- Wuhan Polytechnic University
- Wuhan 430023
- China
| | - Di Zhang
- School of Biology and Pharmaceutical Engineering
- Wuhan Polytechnic University
- Wuhan 430023
- China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science
- Wuhan Polytechnic University
- Wuhan 430023
- China
| | - Lingyun Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science
- Wuhan Polytechnic University
- Wuhan 430023
- China
- School of Biology and Pharmaceutical Engineering
| |
Collapse
|
86
|
Kikiowo B, Ogunleye AJ, Inyang OK, Adelakun NS, Omotuyi OI, Metibemu DS, David TI, Oludoyi OO, Ijatuyi TT. Flavones scaffold of Chromolaena odorata as a potential xanthine oxidase inhibitor: Induced Fit Docking and ADME studies. ACTA ACUST UNITED AC 2019; 10:227-234. [PMID: 32983938 PMCID: PMC7502905 DOI: 10.34172/bi.2020.29] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/02/2019] [Accepted: 10/13/2019] [Indexed: 12/12/2022]
Abstract
Introduction: Gout is a type of painful inflammation initiated by the interactions between monosodium urate crystals and connective tissue. Xanthine oxidase (XO) catalyzes the oxidation of hypoxanthine to xanthine, then to uric acid. The primary treatments for gout include XO inhibitors. At present, allopurinol is the most used XO inhibitor for the treatment of gout. However, it can cause adverse effects commonly known as allopurinol hypersensitivity syndrome, thereby limiting its usage. Consequently, it is necessary to develop potent and less toxic inhibitors of XO. Chromolaena odorata is one of such plants under investigation for its diverse health benefits. Methods: Phytochemicals of C. odorata were screened against XO receptor, using molecular docking. The top five hit compounds of glide docking yield flavones scaffold which were subjected to induced fit docking (IFD) and absorption, distribution, metabolism, and excretion (ADME) studies. Results: The result showed that flavones scaffold of C. odorata can bind with higher affinity and lower free energy values when compared to that of the standard, allopurinol. The IFD scores of the flavones scaffold range from -1525.25 to -1527.99 kcal/mol. Conclusion: Our results have shown that flavones scaffold might have the potential to act as an effective drug candidate when compared to allopurinol in treating and/or preventing gout and some inflammatory condition.
Collapse
Affiliation(s)
- Babatomiwa Kikiowo
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.,Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Adewale Joseph Ogunleye
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Olumide Kayode Inyang
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.,Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Niyi Samuel Adelakun
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.,Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Olaposi Idowu Omotuyi
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.,Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Damilohun Samuel Metibemu
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.,Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Temitope Israel David
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.,Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Oluwatoyin Olajide Oludoyi
- Centre for Biocomputing and Drug Development, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria.,Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | - Taiwo Tolulope Ijatuyi
- Department of Biochemistry, Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| |
Collapse
|
87
|
Interaction mechanism of carnosic acid against glycosidase (α-amylase and α-glucosidase). Int J Biol Macromol 2019; 138:846-853. [DOI: 10.1016/j.ijbiomac.2019.07.179] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 11/18/2022]
|
88
|
Cao X, He Y, Kong Y, Mei X, Huo Y, He Y, Liu J. Elucidating the interaction mechanism of eriocitrin with β-casein by multi-spectroscopic and molecular simulation methods. Food Hydrocoll 2019. [DOI: 10.1016/j.foodhyd.2019.03.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
89
|
Yu Q, Fan L, Duan Z. Five individual polyphenols as tyrosinase inhibitors: Inhibitory activity, synergistic effect, action mechanism, and molecular docking. Food Chem 2019; 297:124910. [PMID: 31253292 DOI: 10.1016/j.foodchem.2019.05.184] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/07/2019] [Accepted: 05/27/2019] [Indexed: 01/01/2023]
Abstract
Polyphenols can inhibit the enzymatic browning in food, but their indistinct synergistic effect and conformational change have limited their applications. In this paper, the mixture of quercetin, cinnamic acid and ferulic acid (Group 11, KI = 0.239 mM) possessed a higher inhibition ability than quercetin (KI = 0.361 mM), which could promote the spontaneous binding process. The final Group 11-tyrosinase complex is more stable, and the hydrophobic effect is the major driving force during the binding process. Moreover, there is not a direct relationship between the destruction of secondary structures and catalytic activity of tyrosinase. The interaction between ferulic acid and tyrosinase could destroy the secondary structures of enzyme but it had little impact on the tyrosinase activity. Molecular docking suggested that three polyphenols from Group 11 have synergistic effect on tyrosinase. This study provides new perspectives about the development of tyrosinase inhibitors in food products.
Collapse
Affiliation(s)
- Qun Yu
- State Key Laboratory of Food Science & Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Liuping Fan
- State Key Laboratory of Food Science & Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China.
| | - Zhenhua Duan
- Institute of Food Research, Hezhou University, Guangxi 542899, China
| |
Collapse
|
90
|
An Improved Method for the Synthesis of Butein Using SOCl 2/EtOH as Catalyst and Deciphering Its Inhibition Mechanism on Xanthine Oxidase. Molecules 2019; 24:molecules24101948. [PMID: 31117192 PMCID: PMC6572126 DOI: 10.3390/molecules24101948] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/18/2019] [Accepted: 05/19/2019] [Indexed: 12/24/2022] Open
Abstract
Butein (3,4,2',4'-tetrahydroxychalcone) belongs to the chalcone family of flavonoids and possesses various biological activities. In this study, butein was synthesized through aldol condensation catalyzed by thionyl chloride (SOCl2)/ethyl alcohol (EtOH) for the first time. The optimal reaction conditions including the molar ratio of reactants, the dosage of catalyst, and the reaction time on the yield of product were investigated, and the straightforward strategy assembles the yield of butein up to 88%. Butein has been found to inhibit xanthine oxidase (XO) activity. Herein, the inhibitory mechanism of butein against XO was discussed in aspects of inhibition kinetic, fluorescence titration, synchronous fluorescence spectroscopy, and molecular docking. The inhibition kinetic analysis showed that butein possessed a stronger inhibition on XO in an irreversible competitive manner with IC50 value of 2.93 × 10-6 mol L-1. The results of fluorescence titrations and synchronous fluorescence spectroscopy indicated that butein was able to interact with XO at one binding site, and the fluorophores of XO were placed in a more hydrophobic environment with the addition of butein. Subsequently, the result of molecular docking between butein and XO protein revealed that butein formed hydrogen bonding with the amino acid residues located in the hydrophobic cavity of XO. All the results suggested that the inhibitory mechanism of butein on XO may be the insertion of butein into the active site occupying the catalytic center of XO to avoid the entrance of xanthine and inducing conformational changes in XO.
Collapse
|
91
|
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: 4.2] [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
|
92
|
Malik N, Dhiman P, Khatkar A. In Silico and 3D QSAR Studies of Natural Based Derivatives as Xanthine Oxidase Inhibitors. Curr Top Med Chem 2019; 19:123-138. [PMID: 30727896 DOI: 10.2174/1568026619666190206122640] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/23/2018] [Accepted: 01/27/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND A large number of disorders and their symptoms emerge from deficiency or overproduction of specific metabolites has drawn the attention for the discovery of new therapeutic agents for the treatment of disorders. Various approaches such as computational drug design have provided the new methodology for the selection and evaluation of target protein and the lead compound mechanistically. For instance, the overproduction of xanthine oxidase causes the accumulation of uric acid which can prompt gout. OBJECTIVE In the present study we critically discussed the various techniques such as 3-D QSAR and molecular docking for the study of the natural based xanthine oxidase inhibitors with their mechanistic insight into the interaction of xanthine oxidase and various natural leads. CONCLUSION The computational studies of deferent natural compounds were discussed as a result the flavonoids, anthraquinones, xanthones shown the remarkable inhibitory potential for xanthine oxidase inhibition moreover the flavonoids such as hesperidin and rutin were found as promising candidates for further exploration.
Collapse
Affiliation(s)
- Neelam Malik
- Department of Pharmaceutical sciences, Maharshi Dayanand University Rohtak, Haryana, India
| | - Priyanka Dhiman
- Department of Pharmaceutical sciences, Maharshi Dayanand University Rohtak, Haryana, India
| | - Anurag Khatkar
- Laboratory for Preservation Technology and Enzyme Inhibition Studies, Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
93
|
Ren J, Liao L, Shang S, Zheng Y, Sha W, Yuan E. Purification, Characterization, and Bioactivities of Polyphenols from Platycladus orientalis (L.) Franco. J Food Sci 2019; 84:667-677. [PMID: 30779137 DOI: 10.1111/1750-3841.14483] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 01/12/2023]
Abstract
The polyphenols (PF) from Platycladus Orientalis (L.) Franco leaves were purified by using 10 different macroporous adsorption resins. HPD-722 resin showed the best adsorption and desorption capacities. The static and dynamic adsorption and desorption of PF on HPD-722 resin were studied and the total polyphenols were separated into two fractions, PF-A and PF-B. PF-A and PF-B demonstrated similar scavenging activity of free radical (DPPH, ABTS, hydroxyl radical, superoxide anion). The scavenging activity of PF-A and PF-B on hydroxyl radical and superoxide anion radical reached the equal levels of vitamin C and gallic acid. The IC50 value of PF-A for hydroxyl radical scavenging activity and superoxide anion radical scavenging activity were 0.50 and 0.56 mg/mL, while those of PF-B were 0.61 and 0.64 mg/mL. PF-A and PF-B could reduce the overproduction of inflammatory cytokines (TNF-α, Pro-IL-1β, and IL-6) induced by lipopolysaccharide and their protein expression in THP-1 cells. PF-B exhibited better anti-inflammatory effect than PF-A in the dosage range of 1.0-4.0 µg/mL. Structural identification of PF-A and PF-B were conducted by HPLC-MS/MS. Ten polyphenol compounds were identified in PF-A and PF-B, respectively, by HPLC-MS/MS, including quercetin, apigenin, myricetin, and so on. Molecular docking studies indicated that apigenin, myricetin, luteolin, kaempferol, and quercetin effectively inhibit xanthine oxidase by forming hydrogen bonds with the amino acid residues and binding to the active site of the enzyme. The results might supply useful information for better understanding the chemical structure, antioxidant, and anti-inflammatory activities of Platycladuso (L.) Franco leaves polyphenols. PRACTICAL APPLICATION: This study demonstrated that polyphenols from P. orientalis (L.) Franco leaves have the potential applications as functional food ingredient for the prevention and treatment of gout and inflammation, hyperuricemia and gout.
Collapse
Affiliation(s)
- Jiaoyan Ren
- School of Food Science and Engineering, South China Univ. of Technology, Guangzhou, China
- Sino-Singapore International Joint Research Institute, Guangzhou Knowledge City, China
| | - Linfeng Liao
- School of Food Science and Engineering, South China Univ. of Technology, Guangzhou, China
| | - Shuaiming Shang
- School of Food Science and Engineering, South China Univ. of Technology, Guangzhou, China
| | - Yamei Zheng
- School of Food Science and Engineering, South China Univ. of Technology, Guangzhou, China
| | - Wanqian Sha
- School of Food Science and Engineering, South China Univ. of Technology, Guangzhou, China
| | - Erdong Yuan
- School of Food Science and Engineering, South China Univ. of Technology, Guangzhou, China
| |
Collapse
|
94
|
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: 85] [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
|
95
|
Pharmacological urate-lowering approaches in chronic kidney disease. Eur J Med Chem 2019; 166:186-196. [PMID: 30769179 DOI: 10.1016/j.ejmech.2019.01.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/20/2019] [Accepted: 01/20/2019] [Indexed: 02/05/2023]
Abstract
Chronic kidney disease (CKD) has become a global public health issue and uric acid (UA) remains a major risk factor of CKD. As the main organ for the elimination of UA, kidney owned a group of urate transporters in tubular epithelium. Kidney disease hampered the UA excretion, and the accumulation of serum UA in return harmed the renal function. Commercially, there are three kinds of agents targeting at urate-lowering, xanthine oxidoreductase inhibitor which prevents the production of UA, uricosuric which increases the concentration of UA in urine thus decreasing serum UA level, and uricase which converts UA to allantoin resulting in the dramatic decrement of serum UA. Of note, in patients with CKD, administration of above-mentioned agents, alone or combined, needs special attention. New evidence is emerging for the efficacy of several urate-lowering drugs for the treatment of hyperuricemia in patients with CKD. Besides, loads of novel and promising drug candidates and phytochemicals are in the different phases of research and development. As of today, there is insufficient evidence to recommend the widespread use of UA-lowering therapy to prevent or slow down the progression of CKD. The review summarized the evidence and perspectives about the treatment of hyperuricemia with CKD for medicinal chemist and nephrologist.
Collapse
|
96
|
Wu H, Zeng W, Chen G, Guo Y, Yao C, Li J, Liang Z. Spectroscopic techniques investigation on the interaction of glucoamylase with 1-deoxynojirimycin: Mechanistic and conformational study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 206:613-621. [PMID: 30098884 DOI: 10.1016/j.saa.2018.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/22/2018] [Accepted: 08/05/2018] [Indexed: 06/08/2023]
Abstract
1-Deoxynojirimycin (DNJ), a representative polyhydroxylated alkaloids, is widely used in the field of antidiabetic, antitumor, and anti-HIV. The present study tried to clarify the interaction mechanism of DNJ with glucoamylase by multi-spectroscopic techniques, dynamic light scattering in combination with molecular modeling strategies from biophysics point of view. Fluorescence and UV-vis data indicated that fluorescence quenching mechanism of glucoamylase and DNJ was a dynamic manner. The association constant, binding site and thermodynamic parameters were also obtained from fluorescence spectrum at different temperatures. Synchronous fluorescence, circular dichroism and dynamic light scattering methods demonstrated that their interaction induced microenvironment changes around tryptophan residue and protein conformational alteration. The main driving force was hydrophobic interaction and hydrogen bonding. In addition, molecular docking study indicated that 1-deoxynojirimycin could bind in the catalytic domain of glucoamylase and interact with amino acid residues Arg78, Asp79, Glu203 and Glu424 by forming hydrogen bonds. Molecular dynamics simulation demonstrated that profiles of atomic fluctuation remained the rigidity of ligand binding site. This study elucidated the detailed interaction mechanism of DNJ with glucoamylase, which will be helpful for pharmaceutical companies to design new α-glucosidase inhibitor drugs based on polyhydroxylated alkaloids compound like DNJ.
Collapse
Affiliation(s)
- Hao Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, Guangxi, China
| | - Wei Zeng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, Guangxi, China
| | - Guiguang Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, Guangxi, China
| | - Ye Guo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, Guangxi, China
| | - Chengzhen Yao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, Guangxi, China
| | - Juan Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, Guangxi, China
| | - Zhiqun Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
97
|
Mehmood A, Zhao L, Ishaq M, Safdar B, Wang C, Nadeem M. Optimization of total phenolic contents, antioxidant, and in-vitro xanthine oxidase inhibitory activity of sunflower head. CYTA - JOURNAL OF FOOD 2018. [DOI: 10.1080/19476337.2018.1504121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
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 Science and Chemical Engineering, 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 Science and Chemical Engineering, 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 Science and Chemical Engineering, 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 Science and Chemical Engineering, Beijing Technology and Business University, Beijing, 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 Science and Chemical Engineering, Beijing Technology and Business University, Beijing, China
| | - Muhammad Nadeem
- Institute of Food Science and Nutrition, University of Sargodha, Sargodha, Pakistan
| |
Collapse
|
98
|
An in-depth view of potential dual effect of thymol in inhibiting xanthine oxidase activity: Electrochemical measurements in combination with four way PARAFAC analysis and molecular docking insights. Int J Biol Macromol 2018; 119:1298-1310. [DOI: 10.1016/j.ijbiomac.2018.08.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/25/2018] [Accepted: 08/05/2018] [Indexed: 02/01/2023]
|
99
|
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: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
100
|
Wang RD, Su GH, Wang L, Xia Q, Liu R, Lu Q, Zhang JL. Identification and mechanism of effective components from rape (Brassica napus L.) bee pollen on serum uric acid level and xanthine oxidase activity. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.05.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|