1
|
Zhou HB, Feng LJ, Weng XH, Wang T, Lu H, Bian YB, Huang ZY, Zhang JL. Inhibition mechanism of cordycepin and ergosterol from Cordyceps militaris Link. against xanthine oxidase and cyclooxygenase-2. Int J Biol Macromol 2024; 258:128898. [PMID: 38141695 DOI: 10.1016/j.ijbiomac.2023.128898] [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: 10/20/2023] [Revised: 12/06/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Cordyceps militaris Link. (C. militaris) is an entomopathogenic fungus that parasitizes the pupa or cocoon of lepidopteran insect larvae, with various bioactive compounds. Cordycepin and ergosterol are the two active components in C. militaris. This study aimed to evaluate the inhibitory activity of cordycepin and ergosterol against xanthine oxidase (XO) and cyclooxygenase-2 (COX-2), as well as investigate the inhibition mechanism. Cordycepin could better inhibit XO (IC50 = 0.014 mg/mL) and COX-2 (IC50 = 0.055 mg/mL) than ergosterol. Additionally, surface hydrophobicity and circular dichroism (CD) spectra results confirmed the conformational changes in enzymes induced by cordycepin and ergosterol. Finally, cordycepin and ergosterol significantly decreased uric acid (UA) and inflammatory factors to normal level in mice with gouty nephropathy (GN). This study could provide theoretical evidence for utilization of C. militaris in hyperuricemia-management functional foods.
Collapse
Affiliation(s)
- H B Zhou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - L J Feng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - X H Weng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - T Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - H Lu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Y B Bian
- College of Plant Science & Technology, Huazhong Agricultural University, Wuhan 430070, China; Wuhan HUAYU XINMEI Mushroom industry Company Limited, Wuhan 430070, China
| | - Z Y Huang
- Wuhan HUAYU XINMEI Mushroom industry Company Limited, Wuhan 430070, China
| | - J L Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, Hubei 430070, China.
| |
Collapse
|
2
|
Zeng X, Liu Y, Fan Y, Wu D, Meng Y, Qin M. Agents for the Treatment of Gout: Current Advances and Future Perspectives. J Med Chem 2023; 66:14474-14493. [PMID: 37908076 DOI: 10.1021/acs.jmedchem.3c01710] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Gout is characterized by hyperuricemia and the deposition of monosodium urate (MSU) crystals around joints. Despite the availability of several drugs on the market, its treatment remains challenging owing to the notable side effects, such as hepatorenal toxicity and cardiovascular complications, that are associated with most existing agents. This perspective aims to summarize the current research progress in the development of antigout agents, particularly focusing on xanthine oxidase (XO) and urate anion transporter 1 (URAT1) inhibitors from a medicinal chemistry viewpoint and their preliminary structure-activity relationships (SARs). This perspective provides valuable insights and theoretical guidance to medicinal chemists for the discovery of antigout agents with novel chemical structures, better efficiency, and lower toxicity.
Collapse
Affiliation(s)
- Xiaoyi Zeng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yajing Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuxin Fan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Di Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yangyang Meng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Mingze Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
3
|
Saleem H, Yaqub A, Rafique R, Ali Chohan T, Malik DES, Tousif MI, Khurshid U, Ahemad N, Ramasubburayan R, Rengasamy KR. Nutritional and medicinal plants as potential sources of enzyme inhibitors toward the bioactive functional foods: an updated review. Crit Rev Food Sci Nutr 2023; 64:9805-9828. [PMID: 37255100 DOI: 10.1080/10408398.2023.2217264] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Enzymes are biologically active complex protein molecules that catalyze most chemical reactions in living organisms, and their inhibitors accelerate biological processes. This review emphasizes medicinal food plants and their isolated chemicals inhibiting clinically important enzymes in common diseases. A mechanistic overview was investigated to explain the mechanism of these food bases enzyme inhibitors. The enzyme inhibition potential of medicinal food plants and their isolated substances was searched in Ovid, PubMed, Science Direct, Scopus, and Google Scholar. Cholinesterase, amylase, glucosidase, xanthine oxidase, tyrosinase, urease, lipoxygenase, and others were inhibited by crude extracts, solvent fractions, or isolated pure chemicals from medicinal food plants. Several natural compounds have shown tyrosinase inhibition potential, including quercetin, glabridin, phloretin-4-O-β-D-glucopyranoside, lupinalbin, and others. Some of these compounds' inhibitory kinetics and molecular mechanisms are also discussed. Phenolics and flavonoids inhibit enzyme activity best among the secondary metabolites investigated. Several studies showed flavonoids' significant antioxidant and anti-inflammatory activities, highlighting their medicinal potential. Overall, many medicinal food plants, their crude extracts/fractions, and isolated compounds have been studied, and some promising compounds depending on the enzyme have been found. Still, more studies are recommended to derive potential pharmacologically active functional foods.
Collapse
Affiliation(s)
- Hammad Saleem
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| | - Anam Yaqub
- Fatima Memorial Medical and Dental College, Lahore, Pakistan
| | | | - Tahir Ali Chohan
- Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
| | - Durr-E-Shahwar Malik
- Institute of Pharmaceutical Sciences, Peoples University of Medical and Health Sciences, NawabShah, Pakistan
| | - Muhammad Imran Tousif
- Department of Chemistry, Division of Science and Technology, University of Education Lahore, Pakistan
| | - Umair Khurshid
- Department of Pharmaceutical Chemistry, The Islamia University of Bahawalpur, Pakistan
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Ramasamy Ramasubburayan
- Marine Biomedical Research Lab & Environmental Toxicology Unit, Department of Prosthodotics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Kannan Rr Rengasamy
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| |
Collapse
|
4
|
Peng X, Liu K, Hu X, Gong D, Zhang G. Hesperetin-Cu(II) complex as potential α-amylase and α-glucosidase inhibitor: Inhibition mechanism and molecular docking. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 290:122301. [PMID: 36603279 DOI: 10.1016/j.saa.2022.122301] [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: 09/28/2022] [Revised: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
Inhibition of α-amylase and α-glucosidase activity is an effective way for controlling postprandial blood glucose-related diabetes. The study found that hesperetin-Cu(II) complex (Hsp-Cu(II)) exhibited a stronger inhibitory ability on α-amylase and α-glucosidase compared to hesperetin (Hsp), with smaller IC50 values of Hsp-Cu(II) (60.3 ± 0.9 µM for α-amylase; 1.25 ± 0.03 µM for α-glucosidase) than Hsp (115.6 ± 1.1 µM for α-amylase; 55.2 ± 0.1 µM for α-glucosidase). Interestingly, Hsp-Cu(II) and acarbose exerted a synergistic effect on inhibition of α-glucosidase. The binding affinities of Hsp-Cu(II) to α-amylase and α-glucosidase were strong with the Ka values (binding constant) in the magnitude order of 105, which was 9 times larger than Hsp. After interacting, Hsp-Cu(II) reduced α-helix contents of α-amylase and α-glucosidase, resulting in a looser conformation of these two enzymes. Molecular simulations manifested that Hsp-Cu(II) bound to the active center of enzymes driven by hydrogen bonds and interacted with the key catalytic amino acids (α-amylase: Gln63, Asp300 and His305; α-glucosidase: Tyr158, Asp215, Glu277 and Glu411), altering the conformation of enzymes, blocking the entrance of substrates, ultimately reducing the activities of α-glucosidase and α-amylase. This study has demonstrated that Hsp-Cu(II) may be a promising candidate of functional nutritional additive and medicine for the prevention of diabetes.
Collapse
Affiliation(s)
- Xi Peng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; Jiangxi Biotech Vocational College, Nanchang 330200, China
| | - Kai Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xing Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Deming Gong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Guowen Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
5
|
Song D, Zhao H, Wang L, Wang F, Fang L, Zhao X. Ethanol extract of Sophora japonica flower bud, an effective potential dietary supplement for the treatment of hyperuricemia. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
6
|
Xanthine oxidase inhibitory potentials of flavonoid aglycones of Tribulus terrestris: in vivo, in silico and in vitro studies. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-022-00448-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Despite the ongoing safety-driven spate of flavonoid xanthine oxidase (XOD) inhibition investigations, there is a lack of flavonoid-based uricostatic antihyperuricemic agents in clinical medicine. The poor pharmacokinetic profiles of glycosides (the natural form of existence of most flavonoids) relative to their aglycones could be largely responsible for this paradox. This investigation was aimed at providing both functional and molecular bases for the possible discovery of XOD inhibitory (or uricostatic) anti-hyperuricemic flavonoid aglycones from the leaves of a flavonoid-rich medicinal plant, Tribulus terrestris. To this end, the flavonoid aglycone fraction of T. terrestris leaf extract (FATT) was evaluated in vivo for antihyperuricemic activity in ethanol-induced hyperuricemic mice, monitoring serum and liver uric acid levels. Molecular docking and molecular dynamics simulation studies were carried out on the three major flavonoid aglycones of T. terrestris (isorhamnetin, quercetin and kaempferol) against an inhibitor conformation XOD model. The three flavonoids were also subjected to in vitro XOD activity assay, comparing their IC50 to that of allopurinol, a standard uricostatic antihyperuricemic drug.
Results
FATT significantly lowered serum uric acid (p < 0.0001) and liver uric acid (p < 0.05) levels of the experimental animals, implying anti-hyperuricemic activity with uricostatic action mechanism allusions. Molecular docking studies revealed high binding affinity values (− 7.8, − 8.1, − 8.2 kcal/mol) for the aglycones (isorhamnetin, quercetin and kaempferol, respectively). Radius of gyration and RMSD analyses of the molecular dynamics simulation trajectories of the three aglycone–XOD complexes revealed substantial stability, the highest stability being demonstrated by the kaempferol–XOD complex. In vitro XOD activity assay showed kaempferol (IC50: 8.2 ± 0.9 μg/ml), quercetin (IC50: 20.4 ± 1.3 μg/ml) and isorhamnetin (IC50: 22.2 ± 2.1 μg/ml) to be more potent than allopurinol (IC50: 30.1 ± 3.0 μg/ml).
Conclusion
This work provides a scientific basis for the use of T. terrestris in the treatment of hyperuricemia-related (e.g. kidney stone and gout) disorders. It also provides the molecular basis for a focussed screening of the flavonoid aglycones chemical space for the possible discovery of flavonoid-based uricostatic anti-hyperuricemic drugs or drug templates.
Collapse
|
7
|
Yang Y, Chen Q, Ruan S, Ao J, Liao SG. Insights into the Inhibitory Mechanism of Viniferifuran on Xanthine Oxidase by Multiple Spectroscopic Techniques and Molecular Docking. Molecules 2022; 27:molecules27227730. [PMID: 36431832 PMCID: PMC9694772 DOI: 10.3390/molecules27227730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022] Open
Abstract
Viniferifuran was investigated for its potential to inhibit the activity of xanthine oxidase (XO), a key enzyme catalyzing xanthine to uric acid. An enzyme kinetics analysis showed that viniferifuran possessed a strong inhibition on XO in a typical anti-competitive manner with an IC50 value of 12.32 μM (IC50 for the first-line clinical drug allopurinol: 29.72 μM). FT-IR and CD data analyses showed that viniferifuran could induce a conformational change of XO with a decrease in the α-helix and increases in the β-sheet, β-turn, and random coil structures. A molecular docking analysis revealed that viniferifuran bound to the amino acid residues located within the activity cavity of XO by a strong hydrophobic interaction (for Ser1214, Val1011, Phe914, Phe1009, Leu1014, and Phe649) and hydrogen bonding (for Asn768, Ser876, and Tyr735). These findings suggested that viniferifuran might be a promising XO inhibitor with a favorable mechanism of action.
Collapse
Affiliation(s)
- Yaxin Yang
- School of Basic Medicine, Guizhou Medical University, Guian New District, Guizhou 550025, China
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, Guizhou 550025, China
| | - Qian Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, Guizhou 550025, China
| | - Shiyang Ruan
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, Guizhou 550025, China
| | - Junli Ao
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, Guizhou 550025, China
| | - Shang-Gao Liao
- School of Basic Medicine, Guizhou Medical University, Guian New District, Guizhou 550025, China
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, Guizhou 550025, China
- Correspondence: or
| |
Collapse
|
8
|
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]
|
9
|
Vijeesh V, Vysakh A, Jisha N, Latha MS. In vitro enzyme inhibition and in vivo anti-hyperuricemic potential of eugenol: An experimental approach. Drug Dev Ind Pharm 2022; 47:1998-2003. [PMID: 35616220 DOI: 10.1080/03639045.2022.2083156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Xanthine oxidase (XO) was accountable for the uric acid synthesis in the body and is considered as a prominent therapeutic target in urate lowering treatment. Eugenol is a natural compound commonly found in the clove, cinnamon etc. and have various biological activities. This study was designed to examine the anti-hyperuricemic effect of eugenol by in vitro and in vivo studies. Potassium oxonate (PO) was used to induce hyperuricemia in Wistar rats. Different doses of eugenol (1.25, 2.5 and 5mg/kg bwt orally) were used for the treatment and various biological function markers (renal, hepatic and hematological) were analyzed. The IC50 value obtained for eugenol was 3.51 ± 0.002 μM. The kinetic studies revealed that the eugenol exhibited a mixed type of inhibition. Abnormality in the levels of various biological function markers were observed in the PO treated rats. Upon the eugenol treatment, those biological function markers were retained near to its normal values. The study proved the anti-hyperuricemic potential of eugenol against the PO induced hyperuricemia model.
Collapse
Affiliation(s)
- V Vijeesh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - A Vysakh
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - Ninan Jisha
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| | - M S Latha
- School of Biosciences, Mahatma Gandhi University, Priyadarshini Hills, Kottayam, Kerala, India
| |
Collapse
|
10
|
Du H, Li SJ. Inhibition of porphyra polysaccharide on xanthine oxidase activity and its inhibition mechanism. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 266:120446. [PMID: 34628362 DOI: 10.1016/j.saa.2021.120446] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/18/2021] [Accepted: 09/24/2021] [Indexed: 06/13/2023]
Abstract
Xanthine oxidase (XO) is a purine catabolic enzyme related to hyperuricemia and gout. Porphyra polysaccharide (PP) is a kind of sulfated polysaccharide with potent biological activity. Herein, the interaction mechanism between PP and XO was studied by enzyme kinetics and multi-spectroscopy methods for the first time. Inhibition kinetics assay showed that PP reversibly inhibited XO activity in a mixed competitive manner with an IC50 of 10.53 ± 0.69 mg/ml. Fluorescence titration studies and thermodynamic parameter calculations revealed that PP could spontaneously bind to XO through hydrophobic interactions, with a class of binding site. Circular dichroism analysis demonstrated that PP induced secondary structure rearrangement and conformational change of XO. Molecular docking further revealed that PP inserted into the hydrophobic cavity of XO, occupying the catalytic center, leading to the inhibition of XO activity. This study may provide new insights into the inhibitory mechanism of PP as a promising XO inhibitor.
Collapse
Affiliation(s)
- Hongyan Du
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, PR China
| | - Shu Jie Li
- Department of Biophysics, School of Physical Science, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, PR China; Qilu Institute of Technology, Shandong 250200, PR China.
| |
Collapse
|
11
|
Luo LS, Wang Y, Dai LJ, He FX, Zhang JL, Zhou Q. Triterpenoid acids from medicinal mushroom Inonotus obliquus (Chaga) alleviate hyperuricemia and inflammation in hyperuricemic mice: Possible inhibitory effects on xanthine oxidase activity. J Food Biochem 2021; 46:e13932. [PMID: 34528276 DOI: 10.1111/jfbc.13932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/11/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022]
Abstract
The purpose of this study was to explore the hypouricemic effect in hyperuricemia mice of triterpenoid acids from Inonotus obliquus (TAIO), and decipher of the underlying xanthine oxidase inhibitory mechanism. Measurement of xanthine oxidase (XO) inhibitory activity was assayed. Organ indexes and serum biochemical indicators were measured in potassium oxonate-induced hyperuricemia mice. Studies showed that TAIO had the strong inhibitory effect on XO activity, and its inhibition type was mixed and reversible. In vivo, TAIO decreased efficiently uric acid level, hepatic XO, serum blood urea nitrogen activities in hyperuricemia mice. Indicating that TAIO may ameliorate kidney damage and relieve inflammation in hyperuricemic mice, and had the inhibitory effect on XO activity. Furthermore, eight triterpenoids were identified by Ultra performance liquid chromatography electrospray quadrupole time of flight mass spectrometry. These findings proved that triterpenoids from Inonotus obliquus would have potential biological characteristics and effect on controlling hyperuricemia and gout as an active supplement. PRACTICAL APPLICATIONS: There are a large amount of evidence indicating that hyperuricemia and gout are related to the hypertension and obesity. And gout and hyperuricemia are also possible connection with cardiovascular disease and metabolic syndrome. Currently, xanthine oxidase is the target of many kinds of chemical drugs at present, but the therapeutic drugs used in clinical medicine will produce more or less side effects. Therefore, the aim of this study was to explore the material basis of effective substances for reducing uric acid in Inonotus obliquus and to evaluate its effect. This study can provide a promising application of Inonotus obliquus in the fields of functional foods or medicines for gout and hyperuricemia.
Collapse
Affiliation(s)
- Lin-Song Luo
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yu Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Li-Jun Dai
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Fang-Xia He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jiu-Liang Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, China
| | - Qing Zhou
- Department of Pharmacy, Wuhan City Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Mehmood A, Zhao L, Ishaq M, Xin W, Zhao L, Wang C, Hossen I, Zhang H, Lian Y, Xu M. Anti-hyperuricemic potential of stevia (Stevia rebaudiana Bertoni) residue extract in hyperuricemic mice. Food Funct 2021; 11:6387-6406. [PMID: 32613954 DOI: 10.1039/c9fo02246e] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Hyperuricemia (HUA) is considered a potent risk factor for the development of gout, renal failure, and cardiovascular disease. The current project was designed to use stevia (Stevia rebaudiana Bertoni) byproduct, named stevia residue extract (STVRE), for the treatment of HUA. Male Kunming mice were divided into six groups: normal control, model control, positive control (allopurinol, 5 mg per kg body weight [bw]), STVRE-1 (75 mg per kg bw), STVRE-2 (150 mg per kg bw), and STVRE-3 (300 mg per kg bw). HUA was induced by the administration of potassium oxonate (100 mg per kg bw), fructose (10% w/v), and yeast extract (100 mg per kg bw) for 8 weeks. STVRE significantly (p < 0.05) decreased uric acid (UA) production and ameliorated UA excretion by interacting with urate transporters. The STVRE remarkably attenuated oxidative stress mediated by UA and downregulated inflammatory-related response markers such as COX-2, NF-κB, PGE2, IL-1β, and TNF-α. Furthermore, STVRE also reversed HUA-induced abnormalities in kidneys compared with the MC group. The results of our study suggest that STVRE has potential to attenuate hyperuricemia and renal protective effects, and may be used as a natural supplement for the possible treatment of UA-related disorders.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives School of Food and Chemical Technology, Beijing Technology and Business University, Beijing 100048, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Marahatha R, Basnet S, Bhattarai BR, Budhathoki P, Aryal B, Adhikari B, Lamichhane G, Poudel DK, Parajuli N. Potential natural inhibitors of xanthine oxidase and HMG-CoA reductase in cholesterol regulation: in silico analysis. BMC Complement Med Ther 2021; 21:1. [PMID: 33386071 PMCID: PMC7775628 DOI: 10.1186/s12906-020-03162-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/23/2020] [Indexed: 12/30/2022] Open
Abstract
Background Hypercholesterolemia has posed a serious threat of heart diseases and stroke worldwide. Xanthine oxidase (XO), the rate-limiting enzyme in uric acid biosynthesis, is regarded as the root of reactive oxygen species (ROS) that generate atherosclerosis and cholesterol crystals. β-Hydroxy β-methylglutaryl-coenzyme A reductase (HMGR) is a rate-limiting enzyme in cholesterol biosynthesis. Although some commercially available enzyme inhibiting drugs have effectively reduced cholesterol levels, most of them have failed to meet potential drug candidates’ requirements. Here, we have carried out an in-silico analysis of secondary metabolites that have already shown good inhibitory activity against XO and HMGR in a wet lab setup. Methods Out of 118 secondary metabolites reviewed, sixteen molecules inhibiting XO and HMGR were selected based on the IC50 values reported in in vitro assays. Further, receptor-based virtual screening was carried out against secondary metabolites using GOLD Protein-Ligand Docking Software, combined with subsequent post-docking, to study the binding affinities of ligands to the enzymes. In-silico ADMET analysis was carried out to explore their pharmacokinetic properties, followed by toxicity prediction through ProTox-II. Results The molecular docking of amentoflavone (GOLD score 70.54, ∆G calc. = − 10.4 Kcal/mol) and ganomycin I (GOLD score 59.61, ∆G calc. = − 6.8 Kcal/mol) displayed that the drug has effectively bound at the competitive site of XO and HMGR, respectively. Besides, 6-paradol and selgin could be potential drug candidates inhibiting XO. Likewise, n-octadecanyl-O-α-D-glucopyranosyl (6′ → 1″)-O-α-D-glucopyranoside could be potential drug candidates to maintain serum cholesterol. In-silico ADMET analysis has shown that these sixteen metabolites were optimal within the categorical range compared to commercially available XO and HMGR inhibitors, respectively. Toxicity analysis through ProTox-II revealed that 6-gingerol, ganoleucoin K, and ganoleucoin Z are toxic for human use. Conclusion This computational analysis supports earlier experimental evidence towards the inhibition of XO and HMGR by natural products. Further study is necessary to explore the clinical efficacy of these secondary molecules, which might be alternatives for the treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Rishab Marahatha
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Saroj Basnet
- Center for Drug Design and Molecular Simulation Division, Cancer Care Nepal and Research Center, Jorpati, Kathmandu, Nepal
| | - Bibek Raj Bhattarai
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Prakriti Budhathoki
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Babita Aryal
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Bikash Adhikari
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Ganesh Lamichhane
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Darbin Kumar Poudel
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Niranjan Parajuli
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal.
| |
Collapse
|
16
|
Singh JV, Bedi PMS, Singh H, Sharma S. Xanthine oxidase inhibitors: patent landscape and clinical development (2015–2020). Expert Opin Ther Pat 2020; 30:769-780. [DOI: 10.1080/13543776.2020.1811233] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jatinder Vir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | | | - Harbinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Yang Q, Wang Q, Feng Y, Wei Q, Sun C, Firempong CK, Adu-Frimpong M, Li R, Bao R, Toreniyazov E, Ji H, Yu J, Xu X. Anti-hyperuricemic property of 6-shogaol via self-micro emulsifying drug delivery system in model rats: formulation design, in vitro and in vivo evaluation. Drug Dev Ind Pharm 2019; 45:1265-1276. [DOI: 10.1080/03639045.2019.1594885] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Qiuxuan Yang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Yingshu Feng
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Qiuyu Wei
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Congyong Sun
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Caleb Kesse Firempong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
- Department of Biochemistry and Biotechnology, College of Science, KwameNkrumah University of Science and Technology, Kumasi, Ghana
| | - Michael Adu-Frimpong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Ran Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Rui Bao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Elmurat Toreniyazov
- Ashkent State Agricultural University (Nukus branch), Avdanberdi str., Nukus, Karakalpakstan
| | - Hao Ji
- Jiangsu Tian Sheng Pharmaceutical Co., Ltd, Zhenjiang, China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| |
Collapse
|
20
|
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
|
21
|
Deciphering the inhibition effect of thymoquinone on xanthine oxidase activity using differential pulse voltammetry in combination with theoretical studies. Enzyme Microb Technol 2019; 121:29-36. [DOI: 10.1016/j.enzmictec.2018.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 02/07/2023]
|
22
|
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
|
23
|
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]
|
24
|
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
|
25
|
Mehmood A, Zhao L, Wang C, Nadeem M, Raza A, Ali N, Shah AA. Management of hyperuricemia through dietary polyphenols as a natural medicament: A comprehensive review. Crit Rev Food Sci Nutr 2017; 59:1433-1455. [PMID: 29278921 DOI: 10.1080/10408398.2017.1412939] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hyperuricemia, a condition due to high serum uric acid level and is notorious to health. It is considered to be a potent risk factor for gout and dramatically associated in the development of many chronic diseases such as malignant tumor, cardiovascular disorders and renal failure. Modern innovative medicinal and therapeutic interventions are underlying these days to combat hyperuricemia. Previously reported studies revealed the significant impact of dietary polyphenols (e.g. anthocyanins, phenolic acids, flavonoids etc.) against hyperurecemia disorder. Dietary plant polyphenols, unlike anti- hyperuricemic agents, are not reported to have any side effects in curing hyperuricemia. The current comprehensive review figure outs the use of dietary polyphenols as a natural remedy for the management of hyperuricemia. The sources, affiliated pathways, mode of actions and factors affecting their efficiency to prevent hyperuricemia are deeply discussed in this article. Additionally, limitations and suggestions regarding previously reported studies are also highlighted.
Collapse
Affiliation(s)
- Arshad Mehmood
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China.,c Institute of Food Science and Nutrition, University of Sargodha , Sargodha , Pakistan
| | - Lei Zhao
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Chengtao Wang
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Muhammad Nadeem
- c Institute of Food Science and Nutrition, University of Sargodha , Sargodha , Pakistan
| | - Ali Raza
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Nawazish Ali
- a Beijing Advance Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University , Beijing , China.,b Beijing Engineering and Technology Research Center of Food Additives , School of Food and Chemical Engineering, Beijing Technology and Business University , Beijing , China
| | - Amjad Abbas Shah
- c Institute of Food Science and Nutrition, University of Sargodha , Sargodha , Pakistan
| |
Collapse
|
26
|
Zhang ZC, Wang HB, Zhou Q, Hu B, Wen JH, Zhang JL. Screening of effective xanthine oxidase inhibitors in dietary anthocyanins from purple sweet potato (Ipomoea batatas L. Cultivar Eshu No.8) and deciphering of the underlying mechanisms in vitro. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
27
|
Xanthine oxidase inhibitors beyond allopurinol and febuxostat; an overview and selection of potential leads based on in silico calculated physico-chemical properties, predicted pharmacokinetics and toxicity. Eur J Med Chem 2017; 135:491-516. [PMID: 28478180 DOI: 10.1016/j.ejmech.2017.04.031] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/28/2017] [Accepted: 04/12/2017] [Indexed: 02/06/2023]
Abstract
Xanthine oxidase (XO), a versatile metalloflavoprotein enzyme, catalyzes the oxidative hydroxylation of hypoxanthine and xanthine to uric acid in purine catabolism while simultaneously producing reactive oxygen species. Both lead to the gout-causing hyperuricemia and oxidative damage of the tissues where overactivity of XO is present. Over the past years, significant progress and efforts towards the discovery and development of new XO inhibitors have been made and we believe that not only experts in the field, but also general readership would benefit from a review that addresses this topic. Accordingly, the aim of this article was to overview and select the most potent recently reported XO inhibitors and to compare their structures, mechanisms of action, potency and effectiveness of their inhibitory activity, in silico calculated physico-chemical properties as well as predicted pharmacokinetics and toxicity. Derivatives of imidazole, 1,3-thiazole and pyrimidine proved to be more potent than febuxostat while also displaying/possessing favorable predicted physico-chemical, pharmacokinetic and toxicological properties. Although being structurally similar to febuxostat, these optimized inhibitors bear some structural freshness and could be adopted as hits for hit-to-lead development and further evaluation by in vivo studies towards novel drug candidates, and represent valuable model structures for design of novel XO inhibitors.
Collapse
|
28
|
Wang LH, Wang MS, Zeng XA, Gong DM, Huang YB. An in vitro investigation of the inhibitory mechanism of β-galactosidase by cinnamaldehyde alone and in combination with carvacrol and thymol. Biochim Biophys Acta Gen Subj 2017; 1861:3189-3198. [DOI: 10.1016/j.bbagen.2016.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/11/2016] [Accepted: 08/10/2016] [Indexed: 10/21/2022]
|
29
|
Durak A, Gawlik-Dziki U, Kowalska I. Evaluation of interactions between coffee and cardamom, their type, and strength in relation to interactions in a model system. CYTA - JOURNAL OF FOOD 2016. [DOI: 10.1080/19476337.2016.1247298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Agata Durak
- Department of Biochemistry and Food Chemistry, University of Life Sciences, Lublin, Poland
| | - Urszula Gawlik-Dziki
- Department of Biochemistry and Food Chemistry, University of Life Sciences, Lublin, Poland
| | - Iwona Kowalska
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation- State Research Institute, Puławy, Poland
| |
Collapse
|
30
|
Ojha R, Singh J, Ojha A, Singh H, Sharma S, Nepali K. An updated patent review: xanthine oxidase inhibitors for the treatment of hyperuricemia and gout (2011-2015). Expert Opin Ther Pat 2016; 27:311-345. [DOI: 10.1080/13543776.2017.1261111] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Ritu Ojha
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | - Jagjeet Singh
- Department of Pharmacy, Rayat-Bahara group of Institutes, Hoshiarpur, India
| | - Anu Ojha
- Department of Pharmacy, DIT University, Dehradun, India
| | - Harbinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Kunal Nepali
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| |
Collapse
|
31
|
Chen C, Lü JM, Yao Q. Hyperuricemia-Related Diseases and Xanthine Oxidoreductase (XOR) Inhibitors: An Overview. Med Sci Monit 2016; 22:2501-12. [PMID: 27423335 PMCID: PMC4961276 DOI: 10.12659/msm.899852] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Uric acid is the final oxidation product of purine metabolism in humans. Xanthine oxidoreductase (XOR) catalyzes oxidative hydroxylation of hypoxanthine to xanthine to uric acid, accompanying the production of reactive oxygen species (ROS). Uric acid usually forms ions and salts known as urates and acid urates in serum. Clinically, overproduction or under-excretion of uric acid results in the elevated level of serum uric acid (SUA), termed hyperuricemia, which has long been established as the major etiologic factor in gout. Accordingly, urate-lowering drugs such as allopurinol, an XOR-inhibitor, are extensively used for the treatment of gout. In recent years, the prevalence of hyperuricemia has significantly increased and more clinical investigations have confirmed that hyperuricemia is an independent risk factor for cardiovascular disease, hypertension, diabetes, and many other diseases. Urate-lowering therapy may also play a critical role in the management of these diseases. However, current XOR-inhibitor drugs such as allopurinol and febuxostat may have significant adverse effects. Therefore, there has been great effort to develop new XOR-inhibitor drugs with less or no toxicity for the long-term treatment or prevention of these hyperuricemia-related diseases. In this review, we discuss the mechanism of uric acid homeostasis and alterations, updated prevalence, therapeutic outcomes, and molecular pathophysiology of hyperuricemia-related diseases. We also summarize current discoveries in the development of new XOR inhibitors.
Collapse
Affiliation(s)
- Changyi Chen
- Division of Surgical Research, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Jian-Ming Lü
- Division of Surgical Research, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Qizhi Yao
- Division of Surgical Research, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|