1
|
Bencsik T, Balázs O, Vida RG, Zsidó BZ, Hetényi C, Valentová K, Poór M. Effects of catechins, resveratrol, silymarin components and some of their conjugates on xanthine oxidase-catalyzed xanthine and 6-mercaptopurine oxidation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024. [PMID: 39606799 DOI: 10.1002/jsfa.14045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Over the past two decades, the global incidence of gout has markedly increased, affecting people worldwide. Considering the side effects of xanthine oxidase (XO) inhibitor drugs (e.g. allopurinol and febuxostat) used in the treatment of hyperuricemia and gout, the potential application of phytochemicals has been widely studied. In addition, XO also takes part in the elimination of certain drugs, including 6-mercaptopurine. In the current explorative study, we aimed to examine the potential effects of tea catechins, resveratrol, silymarin flavonolignans and some of their conjugated metabolites on XO-catalyzed xanthine and 6-mercaptopurine oxidation, applying in vitro assays and modeling studies. RESULTS Catechins, resveratrol and resveratrol conjugates exerted no or only weak inhibitory effects on XO. Silybin A, silybin B and isosilybin A were weak, silychristin was a moderate, while 2,3-dehydrosilychristin was a potent inhibitor of the enzyme. Sulfate metabolites of silybin A, silybin B and isosilybin A were considerably stronger inhibitors compared to the parent flavonolignans, and the sulfation of 2,3-dehydrosilychristin slightly increased its inhibitory potency. Silychristin was the sole flavonolignan tested, where sulfate conjugation decreased its inhibitory effect. CONCLUSION 2,3-Dehydrosilychristin seems to be a promising candidate for examining its in vivo antihyperuricemic effects, because both the parent compound and its sulfate conjugate are highly potent inhibitors of XO. © 2024 The Author(s). Journal of the Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Tímea Bencsik
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Orsolya Balázs
- Department of Pharmaceutics and Central Clinical Pharmacy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Róbert G Vida
- Department of Pharmaceutics and Central Clinical Pharmacy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Balázs Z Zsidó
- Pharmacoinformatics Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Csaba Hetényi
- Pharmacoinformatics Unit, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Miklós Poór
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
- Molecular Medicine Research Group, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
2
|
Liang S, Xu D, Wu J, Jiang Q, Zeng Y. Phyllanthi Fructus ameliorates hyperuricemia and kidney injure via inhibiting uric acid synthesis, modulating urate transporters, and alleviating inflammation. Sci Rep 2024; 14:27605. [PMID: 39528682 PMCID: PMC11555318 DOI: 10.1038/s41598-024-79350-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Phyllanthi Fructus, known as Yuganzi (YGZ), is a unique medicine and food homologous fruit with both medicinal and nutritional properties. Its historical use in treating hyperuricemia (HUA) and gout is well-documented. However, the precise therapeutic effects and potential molecular mechanisms remain unclear. In this study, an experimental rat modelling by a high-fat/high-sugar diet and potassium oxonate/adenine oral administration was used to evaluate the pharmacodynamic effects of YGZ. Network pharmacology, molecular docking and molecular dynamics simulation were utilized to elucidate the potential mechanisms. Supplementation with YGZ effectively ameliorated HUA by inhibiting xanthine oxidase activity, and enhancing uric acid excretion through up-regulating of OAT1 and ABCG2, while down-regulating of URAT1. Furthermore, YGZ supplementation enhanced superoxide dismutase activity, reduced malondialdehyde content, and inhibited the secretion of IL1B, IL6, TNFα, ICAM1, VCAM1, TGFβ1, and NF-κB protein expression. Network pharmacology analysis indicated that YGZ influences 138 targets, modulating the disease network via lipid and atherosclerosis, insulin resistance, HIF-1, TNF, IL-17, TLRS, and NF-κB signaling pathways. Molecular docking analysis suggested that organic acids (e.g. ellagic acid, gallic acid) and flavonoids (e.g. quercetin, delphinidin, luteolin, epigallocatechin gallate) exhibited superior binding abilities to key targets (e.g. XDH, ABCG2, URAT1, OAT1, IRS1, PTGS2, TLR4). Noteworthy, molecular dynamics simulations confirmed that epigallocatechin gallate binds to URAT1 with the greatest stability. These results provide substantial evidence for the therapeutic efficacy of YGZ and establish a theoretical foundation for the development of natural products in treating hyperuricemia.
Collapse
Affiliation(s)
- Shaoyu Liang
- The First Affiliated Hospital of Shenzhen University, Shenzhen, 518020, China
| | - Dandan Xu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Junhong Wu
- The First Affiliated Hospital of Shenzhen University, Shenzhen, 518020, China
| | - Qianqian Jiang
- The First Affiliated Hospital of Shenzhen University, Shenzhen, 518020, China.
| | - Yongchang Zeng
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, 510000, China.
| |
Collapse
|
3
|
Ullah Z, Yue P, Mao G, Zhang M, Liu P, Wu X, Zhao T, Yang L. A comprehensive review on recent xanthine oxidase inhibitors of dietary based bioactive substances for the treatment of hyperuricemia and gout: Molecular mechanisms and perspective. Int J Biol Macromol 2024; 278:134832. [PMID: 39168219 DOI: 10.1016/j.ijbiomac.2024.134832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024]
Abstract
Hyperuricemia (HUA) has attained a considerable global health concern, related to the development of other metabolic syndromes. Xanthine oxidase (XO), the main enzyme that catalyzes xanthine and hypoxanthine into uric acid (UA), is a key target for drug development against HUA and gout. Available XO inhibitors are effective, but they come with side effects. Recent, research has identified new XO inhibitors from dietary sources such as flavonoids, phenolic acids, stilbenes, alkaloids, polysaccharides, and polypeptides, effectively reducing UA levels. Structural activity studies revealed that -OH groups and their substitutions on the benzene ring of flavonoids, polyphenols, and stilbenes, cyclic rings in alkaloids, and the helical structure of polysaccharides are crucial for XO inhibition. Polypeptide molecular weight, amino acid sequence, hydrophobicity, and binding mode, also play a significant role in XO inhibition. Molecular docking studies show these bioactive components prevent UA formation by interacting with XO substrates via hydrophobic, hydrogen bonds, and π-π interactions. This review explores the potential bioactive substances from dietary resources with XO inhibitory, and UA lowering potentials detailing the molecular mechanisms involved. It also discusses strategies for designing XO inhibitors and assisting pharmaceutical companies in developing safe and effective treatments for HUA and gout.
Collapse
Affiliation(s)
- Zain Ullah
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Panpan Yue
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Guanghua Mao
- School of the Environment and Safety Engineering, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Min Zhang
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Peng Liu
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China
| | - Xiangyang Wu
- School of the Environment and Safety Engineering, Jiangsu University, Xuefu Rd. 301, Zhenjiang 212013, Jiangsu, China
| | - Ting Zhao
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China.
| | - Liuqing Yang
- School of Chemistry and Chemical Engineering, Jiangsu University, Xuefu Road 301, Zhenjiang 212013, China.
| |
Collapse
|
4
|
Li K, Wang Y, Liu W, Zhang C, Xi Y, Zhou Y, Li H, Liu X. Structure-Activity Relationships and Changes in the Inhibition of Xanthine Oxidase by Polyphenols: A Review. Foods 2024; 13:2365. [PMID: 39123556 PMCID: PMC11312107 DOI: 10.3390/foods13152365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Hyperuricemia (HUA), or elevated uric acid in the blood, has become more prevalent in recent years. Polyphenols, which are known to have good inhibitory activity on xanthine oxidoreductase (XOR), are effective in uric acid reduction. In this review, we address the structure-activity relationship of flavonoids that inhibit XOR activity from two perspectives: the key residues of XOR and the structural properties of flavonoids. Flavonoids' inhibitory effect is enhanced by their hydroxyl, methoxy, and planar structures, whereas glycosylation dramatically reduces their activity. The flavonoid structure-activity relationship informed subsequent discussions of the changes that occur in polyphenols' XOR inhibitory activity during their extraction, processing, gastrointestinal digestion, absorption, and interactions. Furthermore, gastrointestinal digestion and heat treatment during processing can boost the inhibition of XOR. Polyphenols with comparable structures may have a synergistic effect, and their synergy with allopurinol thus provides a promising future research direction.
Collapse
Affiliation(s)
- Kexin Li
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Yumei Wang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Wanlu Liu
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | | | - Yu Xi
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Yanv Zhou
- The Product Makers Co., Ltd., Shanghai 200444, China
| | - He Li
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| | - Xinqi Liu
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China; (K.L.); (Y.W.); (W.L.)
| |
Collapse
|
5
|
Du J, Wang N, Yu D, He P, Gao Y, Tu Y, Li Y. Data mining-guided alleviation of hyperuricemia by Paeonia veitchii Lynch through inhibition of xanthine oxidase and regulation of renal urate transporters. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 124:155305. [PMID: 38176275 DOI: 10.1016/j.phymed.2023.155305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Hyperuricemia (HUA) is a metabolic disease characterized by a high level of uric acid (UA). The extensive historical application of traditional Chinese medicine (TCM) offers a range of herbs and prescriptions used for the treatment of HUA-related disorders. However, the core herbs in the prescriptions and their mechanisms have not been sufficiently explained. PURPOSE Our current investigation aimed to estimate the anti-HUA effect and mechanisms of Paeonia veitchii Lynch, an herb with high use frequency identified from data mining of TCM prescriptions. METHODS Prescriptions for HUA/gout treatment were statistically analyzed through a data mining approach to determine the common nature and use frequency of their composition herbs. The chemical constituents of Paeonia veitchii extract (PVE) were analyzed by UPLC-QTOF-MS/MS, while its UA-lowering effect was further evaluated in adenosine-induced liver cells and potassium oxonate (PO) and hypoxanthine (HX)-induced HUA mice. RESULTS A total of 225 prescriptions involving 246 herbs were sorted out. The properties, flavors and meridians of the appearing herbs were mainly cold, bitter and liver, respectively, while their efficacy was primarily concentrated on clearing heat and dispelling wind. Further usage frequency analysis yielded the top 20 most commonly used herbs, in which PVE presented significant inhibitory activity (IC50 = 131.33 µg/ml) against xanthine oxidase (XOD), and its constituents showed strong binding with XOD in a molecular docking study and further were experimentally validated through XOD enzymatic inhibition and surface plasmon resonance (SPR). PVE (50 to 200 μg/ml) dose-dependently decreased UA levels by inhibiting XOD expression and activity in BRL 3A liver cells. In HUA mice, oral administration of PVE exhibited a significant UA-lowering effect, which was attributed to the reduction of UA production by inhibiting XOD activity and expression, as well as the enhancement of UA excretion by regulating renal urate transporters (URAT1, GLUT9, OAT1 and ABCG2). Noticeably, all doses of PVE treatment did not cause any liver injury, and displayed a renal protective effect. CONCLUSIONS Our results first comprehensively clarified the therapeutic effect and mechanisms of PVE against HUA through suppressing UA production and promoting UA excretion with hepatic and renal protection, suggesting that PVE could be a promising UA-lowering candidate with a desirable safety profile for the treatment of HUA and prevention of gout.
Collapse
Affiliation(s)
- Jiana Du
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Na Wang
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Dehong Yu
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Pei He
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yu Gao
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yanbei Tu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Yanfang Li
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| |
Collapse
|
6
|
Balázs O, Dombi Á, Zsidó BZ, Hetényi C, Valentová K, Vida RG, Poór M. Inhibition of xanthine oxidase-catalyzed xanthine and 6-mercaptopurine oxidation by luteolin, naringenin, myricetin, ampelopsin and their conjugated metabolites. Biomed Pharmacother 2023; 167:115548. [PMID: 37734263 DOI: 10.1016/j.biopha.2023.115548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023] Open
Abstract
Luteolin, naringenin, myricetin, and ampelopsin are abundant flavonoids in nature, and several dietary supplements also contain them at very high doses. After the peroral intake, flavonoids go through extensive presystemic biotransformation; therefore, typically their sulfate/glucuronic acid conjugates reach high concentrations in the circulation. Xanthine oxidase (XO) enzyme is involved in uric acid production, and it also takes part in the elimination of certain drugs (e.g., 6-mercaptopurine). The inhibitory effects of flavonoid aglycones on XO have been widely studied; however, only limited data are available regarding their sulfate and glucuronic acid conjugates. In this study, we examined the impacts of luteolin, naringenin, myricetin, ampelopsin, and their sulfate/glucuronide derivatives on XO-catalyzed xanthine and 6-mercaptopurine oxidations employing in vitro enzyme incubation assays and molecular modeling studies. Our major results/conclusions are the following: (1) Sulfate metabolites were stronger while glucuronic acid derivatives were weaker inhibitors of XO compared to the parent flavonoids. (2) Naringenin, ampelopsin, and their metabolites were weak inhibitors of the enzyme. (3) Luteolin, myricetin, and their sulfates were highly potent inhibitors of XO, and the glucuronides of luteolin showed moderate inhibitory impacts. (4) Conjugated metabolites of luteolin and myricetin can be involved in the inhibitory effects of these flavonoids on XO enzyme.
Collapse
Affiliation(s)
- Orsolya Balázs
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary; Department of Pharmaceutics and Central Clinical Pharmacy, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary
| | - Ágnes Dombi
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary
| | - Balázs Z Zsidó
- Unit of Pharmacoinformatics, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Csaba Hetényi
- Unit of Pharmacoinformatics, Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-142 00 Prague, Czech Republic
| | - Róbert G Vida
- Department of Pharmaceutics and Central Clinical Pharmacy, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, H-7624 Pécs, Hungary.
| |
Collapse
|
7
|
Wang R, Yue X, Shan G, Qiu X, Wang L, Yang L, Li J, Yang B. A novel multi-hyphenated approach to screen and character the xanthine oxidase inhibitors from saffron floral bio-residues. Int J Biol Macromol 2023; 248:125990. [PMID: 37499709 DOI: 10.1016/j.ijbiomac.2023.125990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Recently, the incidence of hyperuricemia increased with patient rejuvenation, searching for new xanthine oxidase (XOD) inhibitors from natural products becomes important. In our previous work, a flavonoid extract of saffron floral bio-residues (SFB) was found to alleviate hyperuricemia via inhibiting XOD. In this study, an integrated approach combining two-dimensional liquid chromatography, surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC) was developed to online screen and character the potential XOD inhibitors from SFB. The two-dimensional liquid chromatography consisted of affinity chromatography and reverse phase chromatography (2D-AR), in which an XOD column, an inactive XOD column, and a control column were used in the first dimensional liquid chromatography to avoid phenomena of "false positive" and "missing screen of compounds with weak affinity to XOD" that often occur in the screening process, and a C18 column was used in the second dimensional liquid chromatography to separate the mixed XOD binders. Four flavonoid glycosides, i.e., quercetin-3-O-sophoroside (QS), kaempferol-3-O-sophoroside (KS), kaempferol-3-O-rutinoside (KR), and kaempferol-3-O-glucoside (KG), were thus successfully screened and identified from SFB extract by the 2D-AR method. The affinity of QS, KS, KR, KG, kaempferol (aglycone of KS, KR and KG), and quercetin (aglycone of QS) binding to XOD was investigated using SPR method, with KD ranged from 4.8 μM to 47.6 μM. The inhibitor constant (KI) of KS, KR, KG, quercetin and kaempferol were 4.92 mM, 1.11 mM, 0.294 mM, 4.93 μM and 3.27 μM, respectively, determined using ITC method. Finally, the anti-XOD activities of KS, the most abundant flavonoid in SFB extract, and kaempferol in hyperuricemia mice were verified, which suggested that the multi-hyphenated approach established herein can be applied for screen and character the XOD inhibitors in natural products.
Collapse
Affiliation(s)
- Ran Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Xingnan Yue
- Shanxi University of Chinese Medicine, College of Basic Medical Sciences, Jinzhong 030619, PR China
| | - Guangzhi Shan
- Institute of Medicinal Biotechnology, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, PR China
| | - Xiaodan Qiu
- Institute of Medicinal Biotechnology, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, PR China
| | - Lan Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Li Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Jiaqi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Bin Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China.
| |
Collapse
|
8
|
Zhang ZX, Mo RM, Liu DB, Liu YS, Liu CH, Li YS, Liu ZH, Qin D. Research on the Efficacy of Ganpu Vine Tea in Inhibiting Uric Acid Production. Metabolites 2023; 13:704. [PMID: 37367863 DOI: 10.3390/metabo13060704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Ganpu vine tea is a new type of health care citrus fruit tea made from citrus shell, Pu-er tea, and vine tea baked as raw materials. In this study, the in vitro uric acid synthase inhibition system and hyperuric acid cell model were constructed to appraise the uric acid lowering efficacy of Ganpu vine tea, traditional Ganpu tea, and vine tea. Results showed that in the uric acid synthase inhibition system, the aqueous extract can inhibite the puric metabolically related enzymes, such as adenosine deaminase (ADA), purine nucleoside phosphorylase (PNP), and xanthine oxidase (XOD). The ability of the aqueous extract to inhibit the above enzyme was as follows: vine tea > Ganpu vine tea > Ganpu tea; all teas had a strong effect on XOD inhibition. The hyperuric acid cell model test showed that the aqueous extract inhibited uric acid production through accumulating inosine and hypoxanthine and hindering xanthine synthesis. The uric acid reductive ability was as follows: Vine tea > Ganpu vine tea > Ganpu tea. The inhibition of enzymes related to uric acid synthesis and the inhibition of uric acid production were significantly enhanced through adding vine tea to Ganpu tea. It also shows that flavonoids are the main factor driving this ability because they are the main active ingredients in these botanical drinks.
Collapse
Affiliation(s)
- Zhi-Xu Zhang
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Horticulture College, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering Technology for Utilization Ingredients from Botanicals, Changsha 410128, China
- State Key Laboratory of Subhealth Intervention Technology, Changsha 410128, China
| | - Run-Ming Mo
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Dong-Bo Liu
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Horticulture College, Hunan Agricultural University, Changsha 410128, China
- State Key Laboratory of Subhealth Intervention Technology, Changsha 410128, China
| | - Yi-Song Liu
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Cong-Hui Liu
- Hunan Kangqi 100 Biotechnology Ltd., Zhuzhou 412205, China
| | - Yong-Shen Li
- Hunan Kangqi 100 Biotechnology Ltd., Zhuzhou 412205, China
| | - Zhong-Hua Liu
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- Horticulture College, Hunan Agricultural University, Changsha 410128, China
- National Research Center of Engineering Technology for Utilization Ingredients from Botanicals, Changsha 410128, China
| | - Dan Qin
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
9
|
Vijeesh V, Vysakh A, Jisha N, Latha M. Multispectroscopic binding studies and in silico docking analysis of interactions of malic acid with xanthine oxidase. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
10
|
Mehmood A, Li J, Rehman AU, Kobun R, Llah IU, Khan I, Althobaiti F, Albogami S, Usman M, Alharthi F, Soliman MM, Yaqoob S, Awan KA, Zhao L, Zhao L. Xanthine oxidase inhibitory study of eight structurally diverse phenolic compounds. Front Nutr 2022; 9:966557. [PMID: 36204384 PMCID: PMC9531272 DOI: 10.3389/fnut.2022.966557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/05/2022] [Indexed: 12/03/2022] Open
Abstract
This project was designed to explore the xanthine oxidase (XO) inhibitory mechanism of eight structurally diverse phenolic compounds [quercetin: C1, quercetin-3-rhamnoside: C2, 4, 5-O-dicaffeoylquinic acid: C3, 3, 5-O-dicaffeoylquinic acid: C4, 3, 4-O-di-caffeoylquinic acid: C5, 4-O-caffeoylquinic acid (C6), 3-O-caffeoylquinic acid: C7, and caffeic acid: C8]. For this purpose, in-vitro and different computational methods were applied to determine the xanthine oxidase (XO) inhibitory potential of eight structurally diverse phenolic compounds. The results revealed that phenolic compounds (C1–C8) possess strong to weak XO inhibitory activity. These results were further confirmed by atomic force microscopy (AFM) and 1H NMR analysis. Furthermore, computational study results revealed that phenolic compounds (C1–C8) bind with the surrounding amino acids of XO at the molybdenum (MO) site. These in-vitro and in-silico results divulge that phenolic compounds have a strong potential to lower uric acid levels via interacting with the XO enzyme and can be used to combat hyperuricemia.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, China
- Department of Food Science and Technology, University of Haripur, Haripur, Pakistan
| | - Jiayi Li
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ashfaq Ur Rehman
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Rovina Kobun
- Faculty of Food Science and Nutrition, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| | - Inam U Llah
- Department of Food Science and Technology, University of Haripur, Haripur, Pakistan
| | - Imran Khan
- Department of Food Science and Technology, University of Haripur, Haripur, Pakistan
| | - Fayez Althobaiti
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Muhammad Usman
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Fahad Alharthi
- Department of Biology, College of Science, Taif University, Taif, Saudi Arabia
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Taif, Saudi Arabia
| | - Sanabil Yaqoob
- Department of Food Science and Technology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Kanza Aziz Awan
- Department of Food Science and Technology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Liang Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, China
- *Correspondence: Liang Zhao
| | - Lei Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, China
- Lei Zhao
| |
Collapse
|
11
|
Zhao J, Huang L, Li R, Zhang Z, Chen J, Tang H. Insights from multi-spectroscopic analysis and molecular modeling to understand the structure-affinity relationship and the interaction mechanism of flavonoids with gliadin. Food Funct 2022; 13:5061-5074. [PMID: 35404372 DOI: 10.1039/d1fo03816h] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gliadin, as a main component of wheat storage protein, is used as a drug encapsulation and delivery system owing to its specific characteristics. Flavonoids are regarded as active natural products with a variety of pharmacological effects. In this study, an integrated method including UV-vis, fluorescence, and FT-IR spectroscopy and molecular modelling was applied to explore the structure-affinity relationship and the interaction nature between a library of flavonoids and gliadin. The characteristic UV-vis spectral changes of gliadin mediated by flavonoids with absorption bands at 218 and 278 nm demonstrated the existence of an interaction depending on generating the ground-state complexes. Fluorescence quenching results showed that the intrinsic fluorescence of gliadin could be effectively quenched by flavonoids coupled with the formation of flavonoid-gliadin complexes through the static quenching mechanism. The structure-affinity relationship revealed the critical structural elements associated with the binding affinity on gliadin and underlined the favorable substituents at the specific positions of flavonoid skeletons leading to a stronger binding potency. From the analysis of synchronous fluorescence spectra, flavonoids could cause the conformation change of gliadin and impact the microenvironment around TYR and TRP residues. Moreover, the ANS fluorescent probe assay suggested that these flavonoids also influenced the surface hydrophobicity of glaidin based on the further exposure or blocking of hydrophobic domains. Molecular modelling was subsequently performed and illustrated the proposed binding conformation of flavonoids on gliadin. Combined with the FT-IR spectra, these results further confirmed the important role of hydrophobic interactions and hydrogen bonds in their binding process.
Collapse
Affiliation(s)
- Jie Zhao
- College of Biological and Food 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
| | - Renjie Li
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, P. R. China.
| | - Zhuangwei Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jin Chen
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, P. R. China.
| | - Hongjin Tang
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, P. R. China.
| |
Collapse
|
12
|
Feng S, Wu S, Xie F, Yang CS, Shao P. Natural compounds lower uric acid levels and hyperuricemia: Molecular mechanisms and prospective. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2022.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
13
|
Goyal A, Tanwar B, Kumar Sihag M, Sharma V. Sacha inchi (Plukenetia volubilis L.): An emerging source of nutrients, omega-3 fatty acid and phytochemicals. Food Chem 2021; 373:131459. [PMID: 34731811 DOI: 10.1016/j.foodchem.2021.131459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/20/2021] [Accepted: 10/20/2021] [Indexed: 12/30/2022]
Abstract
Sacha inchi (Plukenetia volubilis) (SI) is an oleaginous plant producing oil and protein-rich seeds. It has been cultivated for centuries and is native to the tropical rainforest of the Amazon region of South America including parts of Peru and northwestern Brazil. At present, SI seeds are emerging as a potential source of macro- and micronutrients, α-linolenic acid and phytochemicals. This review attempts to elucidate the nutrients, phytonutrients, safety, toxicity, health benefits and food applications of SI seed. Recent scientific studies have associated the consumption of SI seed/oil with reduced risk of chronic inflammatory diseases. However, lack of awareness and in-depth understanding has resulted in it being neglected both at the consumer and industrial level. In all, SI is an underutilized and undervalued oleaginous crop which not only has the potential to mitigate food and nutritional insecurity but also offers humongous opportunities for the development of novel value-added food products.
Collapse
Affiliation(s)
- Ankit Goyal
- Department of Dairy Chemistry, Mansinhbhai Institute of Dairy and Food Technology, Mehsana 384002, Gujarat, India.
| | - Beenu Tanwar
- Department of Dairy Technology, Mansinhbhai Institute of Dairy and Food Technology, Mehsana 384002, Gujarat, India.
| | - Manvesh Kumar Sihag
- Department of Dairy Chemistry, College of Dairy Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana 141001, Punjab, India.
| | - Vivek Sharma
- Dairy Chemistry Division, National Dairy Research Institute (ICAR-NDRI), Karnal, Haryana, India.
| |
Collapse
|
14
|
Zhu M, Pan J, Hu X, Zhang G. Epicatechin Gallate as Xanthine Oxidase Inhibitor: Inhibitory Kinetics, Binding Characteristics, Synergistic Inhibition, and Action Mechanism. Foods 2021; 10:2191. [PMID: 34574301 PMCID: PMC8464939 DOI: 10.3390/foods10092191] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023] Open
Abstract
Epicatechin gallate (ECG) is one of the main components of catechins and has multiple bioactivities. In this work, the inhibitory ability and molecular mechanism of ECG on XO were investigated systematically. ECG was determined as a mixed xanthine oxidase (XO) inhibitor with an IC50 value of 19.33 ± 0.45 μM. The promotion of reduced XO and the inhibition of the formation of uric acid by ECG led to a decrease in O2- radical. The stable ECG-XO complex was formed by hydrogen bonds and van der Waals forces, with the binding constant of the magnitude of 104 L mol-1, and ECG influenced the stability of the polypeptide skeleton and resulted in a more compact conformation of XO. Computational simulations further characterized the binding characteristics and revealed that the inhibitory mechanism of ECG on XO was likely that ECG bound to the vicinity of flavin adenine dinucleotide (FAD) and altered the conformation of XO, hindering the entry of substrate and the diffusion of catalytic products. ECG and allopurinol bound to different active sites of XO and exerted a synergistic inhibitory effect through enhancing their binding stability with XO and changing the target amino acid residues of XO. These findings may provide a theoretical basis for the further application of ECG in the fields of food nutrition and functional foods.
Collapse
Affiliation(s)
| | | | | | - Guowen Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (M.Z.); (J.P.); (X.H.)
| |
Collapse
|
15
|
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
|
16
|
Zhang L, Liu L, Xiao A, Huang S, Li D. Screening and analysis of xanthine oxidase inhibitors in jute leaves and their protective effects against hydrogen peroxide-induced oxidative stress in cells. OPEN CHEM 2020. [DOI: 10.1515/chem-2020-0178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
AbstractJute (Corchorus capsularis L.) is an annual herb of the bast fiber plant and has great potentials in food and medicinal usages because of its various bioactivities. In this study, ultrafiltration coupled with high-performance liquid chromatography-mass spectrometry was established for screening xanthine oxidase inhibitors from the jute leaves extract. Under the optimum screening conditions, three inhibitors were successfully screened and identified as chlorogenic acid, echinacoside, and isorhamnetin-rutinoside with UV and MS data. The fluorescent quenching analysis showed that three inhibitors quenched the fluorescence intensities of enzyme with different binding capacities. For further exploring the bioactivity of three inhibitors, the protective effects on hydrogen peroxide-induced oxidative stress was investigated using human normal liver cell (LO2), human gastric mucosal epithelial cell (GES-1), and human umbilical vein endothelial cell (HUVEC). As a result, they exhibited protective effects on three injured cells in dose-dependent manners without cytotoxicity. To evaluate the difference among different jute species obtained in our laboratories, the amounts of three compounds in ten samples were assessed and analyzed. The results showed that it could be divided into three groups. The jute leaves showed nutrient and medical potentials and deserved further research on pharmaceutical and biochemical utilization in future.
Collapse
Affiliation(s)
- Lang Zhang
- Characteristic Fruit and Vegetable Research Office, Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, People's Republic of China
| | - Liangliang Liu
- Characteristic Fruit and Vegetable Research Office, Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, People's Republic of China
| | - Aiping Xiao
- Characteristic Fruit and Vegetable Research Office, Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, People's Republic of China
| | - Siqi Huang
- Characteristic Fruit and Vegetable Research Office, Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, People's Republic of China
| | - Defang Li
- Characteristic Fruit and Vegetable Research Office, Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, People's Republic of China
| |
Collapse
|
17
|
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]
|
18
|
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
|
19
|
Manninen H, Durandin N, Hopia A, Vuorimaa-Laukkanen E, Laaksonen T. Taste compound – Nanocellulose interaction assessment by fluorescence indicator displacement assay. Food Chem 2020; 318:126511. [DOI: 10.1016/j.foodchem.2020.126511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 02/01/2023]
|
20
|
Liu L, Zhang L, Ren L, Xie Y. Advances in structures required of polyphenols for xanthine oxidase inhibition. FOOD FRONTIERS 2020. [DOI: 10.1002/fft2.27] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Liangliang Liu
- Institute of Bast Fiber Crops Chinese Academy of Agricultural Sciences Changsha 410205 China
| | - Li Zhang
- College of Chemistry and Materials Engineering Huaihua University Huaihua 418000 China
| | - Licheng Ren
- Institute of Bast Fiber Crops Chinese Academy of Agricultural Sciences Changsha 410205 China
- Department of Plastic and Cosmetic Surgery Shenzhen University General Hospital Shenzhen 518055 China
| | - Yixi Xie
- Institute of Bast Fiber Crops Chinese Academy of Agricultural Sciences Changsha 410205 China
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province Xiangtan University Xiangtan 411105 China
| |
Collapse
|
21
|
Kim DS, Lim SB. Semi-Continuous Subcritical Water Extraction of Flavonoids from Citrus unshiu Peel: Their Antioxidant and Enzyme Inhibitory Activities. Antioxidants (Basel) 2020; 9:E360. [PMID: 32344942 PMCID: PMC7278842 DOI: 10.3390/antiox9050360] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/26/2022] Open
Abstract
We extracted and hydrolyzed bioactive flavonoids from C. unshiu peel using subcritical water (SW) in a semi-continuous mode. The individual flavonoid yields, antioxidant and enzyme inhibitory activities of the SW extracts were analyzed. The extraction yields of hesperidin and narirutin increased with increasing temperature from 145 °C to 165 °C. Hydrothermal hydrolysis products (HHP), such as monoglucosides (hesperetin-7-O-glucoside and prunin) and aglycones (hesperetin and naringenin) were obtained in the SW extracts at temperatures above 160 °C. The sum of hesperidin and its HHP in the SW extracts was strongly correlated with antioxidant activities, whereas the contents of hesperetin and naringenin were strongly correlated with enzyme inhibitory activities. Hesperetin exhibited the highest antioxidant activities (2,2-diphenyl-1-picrylhydrazyl radical scavenging activity, ferric-reducing antioxidant power, and oxygen radical absorbance capacity), whereas hesperetin-7-O-glucoside exhibited the highest enzyme inhibitory activities (angiotensin-І converting enzyme (ACE) and pancreatic lipase (PL)). Naringenin exhibited the highest enzyme inhibitory activities (xanthine oxidase and α-glucosidase). PMFs (sinensetin, nobiletin, and tangeretin) also exhibited relatively high inhibitory activities against ACE and PL. This study confirms the potential of SW for extracting and hydrolyzing bioactive flavonoids from C. unshiu peel using an environmentally friendly solvent (water) and a shorter extraction time.
Collapse
Affiliation(s)
| | - Sang-Bin Lim
- Department of Food Bioengineering, Jeju National University, Jeju 63243, Korea;
| |
Collapse
|
22
|
Dai T, Li T, He X, Li X, Liu C, Chen J, McClements DJ. Analysis of inhibitory interaction between epigallocatechin gallate and alpha-glucosidase: A spectroscopy and molecular simulation study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 230:118023. [PMID: 31927512 DOI: 10.1016/j.saa.2019.118023] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/23/2019] [Accepted: 12/30/2019] [Indexed: 06/10/2023]
Abstract
Alpha-glucosidase is one of the main enzymes responsible for digesting starch. Inhibiting its activity is therefore being targeted as a strategy for tackling diabetes. Certain food components have the potential to act as natural α-glucosidase (SCG) inhibitors, such as the polyphenols found in tea. In this study, epigallocatechin gallate (EGCG) was shown to strongly inhibit SCG activity (IC50 value = 3.7 × 10-5 M). Multi-spectroscopic binding molecular simulations indicated that EGCG spontaneously bound to SCG through a combination of hydrogen bonding and hydrophobic interactions. The hypothesis was supported by the results from intrinsic fluorescence quenching, conformational change, surface hydrophobicity decrease, and molecular docking analysis of the SCG after binding. Molecular docking provided powerful visual insights into the nature of the molecular interactions involved. This research provides important new information about the interaction mechanism of EGCG and SCG, which may be beneficial to the development of functional foods to prevent diabetes.
Collapse
Affiliation(s)
- Taotao Dai
- State Key Laboratory of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Ti Li
- State Key Laboratory of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Xiaohong He
- State Key Laboratory of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Chengmei Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Jun Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China.
| | | |
Collapse
|