1
|
Xu H, Yang C, Li L, Du J, Yin Q, Zhao P, Wang N, Huang W, Li Y. Design, synthesis, and evaluation of chalcone derivatives as xanthine oxidase inhibitors. Eur J Med Chem 2024; 279:116893. [PMID: 39348762 DOI: 10.1016/j.ejmech.2024.116893] [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: 07/29/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024]
Abstract
Xanthine oxidase (XO) is an important enzyme that catalyzes the oxidation of hypoxanthine to xanthine and xanthine to uric acid in the catabolism of purines in humans. This makes XO a well-recognized target in alleviating hyperuricemia. The present study adapted a structure-based drug discovery approach to develop potent and low-toxicity XO inhibitors with the chalcone skeleton. We introduced a carboxyl group and a hydroxyl group to the B ring and modified the A ring. 35 chalcone derivatives were designed and synthesized. All the 35 derivatives exhibited higher XO inhibition activities (IC50 = 0.064-0.559 μM) compared with allopurinol (IC50 = 2.588 μM). Their high affinity was attributed to strong hydrogen bond interactions formed between the introduced carboxyl and hydroxyl groups with key amino acid residues in XO. SAR analysis disclosed that carboxyl, hydroxyl, ethyl (12c), methylamino (12h), dimethylamino (12i), indolin (13k), and indol (13l) groups played important roles in improving the whole molecules' inhibition potency against XO. ADME predictions and cytotoxicity assays suggested their pharmacokinetic characteristics and biocompatibility were desirable. Additionally, 12c exhibited a significant hypouricemic effect on potassium oxonate-induced hyperuricemia rats after orally administrated at a dose range of 10-40 mg/kg, representing a promising anti-hyperuricemia potential for further optimization and development.
Collapse
Affiliation(s)
- Haiqi Xu
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Can Yang
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Lizi Li
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Jiana Du
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Qin Yin
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Puchen Zhao
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Na Wang
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Wencai Huang
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China.
| | - Yanfang Li
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
2
|
Yao Y, Wu T, Zhang M, Fu D, Yang H, Chen S. An Improved Test Method for Assaying the Inhibition of Bioflavonoids on Xanthine Oxidase Activity in vitro. ChemistryOpen 2024; 13:e202400127. [PMID: 39246250 PMCID: PMC11625940 DOI: 10.1002/open.202400127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/27/2024] [Indexed: 09/10/2024] Open
Abstract
The difference on inhibitory effects of bioflavonoids inhibiting XOD activity assayed by varying test methods cause of us to be further in consideration. The reported test method creating a micro-environment surrounding XOD in the absence of ⋅O2 -, which is seemly different from the assay in vivo. So, the vitro test method for assaying XOD activity is necessary to be improved for selection of potential inhibitors in the presence of ⋅O2 -. The inhibitory results demonstrated that bioflavonoids of MY, DMY, QUE and LUT are capable to be on effective IC50 values, but others are not. As well, their resulting inhibitions determined by the improved test method are much less than that reported in the literature, indicating that their chemical affinities with XOD become weaker. Moreover, DMY assayed on the inhibitions of XOD in the improved test method performs to be a better inhibitor, as compared to the assay of the reported test methods. Abasing on the transformation of DMY into MY in the presence of ⋅O2 -, the good inhibition of DMY on XOD activity can be explained by the synergistic effect of MY.
Collapse
Affiliation(s)
- Yuanyong Yao
- State Ethnic Affairs Commission Key Development Laboratory of Chinese Veterinary Medicine & National and Local Joint Engineering Center of Chinese Veterinary Medicine Separation and Purification TechnologyTongren Vocational and Technical UniversityTongren554300China
- Institute of Material and Chemical EngineeringTongren UniversityTongren554300China
| | - Tao Wu
- Institute of Material and Chemical EngineeringTongren UniversityTongren554300China
| | - Meng Zhang
- Institute of Material and Chemical EngineeringTongren UniversityTongren554300China
| | - Daihua Fu
- Institute of Material and Chemical EngineeringTongren UniversityTongren554300China
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of EducationYunnan UniversityKunming650091China
| | - Hai Yang
- Institute of Material and Chemical EngineeringTongren UniversityTongren554300China
| | - Shixue Chen
- Institute of Material and Chemical EngineeringTongren UniversityTongren554300China
| |
Collapse
|
3
|
Zhang Y, Ban C, Su D, Liu Y, Zhou S, Fan J. Xanthine oxidase inhibitors: Virtual screening and mechanism of inhibition studies. Int J Biol Macromol 2024; 281:136281. [PMID: 39368582 DOI: 10.1016/j.ijbiomac.2024.136281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Xanthine oxidase (XO), which plays a key role in purine metabolism, is an important target enzyme for the prevention and treatment of hyperuricemia. Inhibitory activity against XO is a common criterion for the screening of compounds with potential anti-hyperuricemic activity. In this study, 22 XO inhibitors were used to construct a 3D-QSAR pharmacophore model. Subsequently, molecular docking and in vitro activity evaluations were used to identify strong XO inhibitors from a list of 2000 natural compounds. The interaction mechanisms of these compounds with XO were analyzed based on inhibition kinetics and multi-spectral analyses. The pharmacophore model was composed of three hydrogen bond receptors and a hydrophobic center. The screened compounds - Diosmetin, Fisetin, and Genistein - all showed good XO inhibitory activity, with IC50 values of 1.86 ± 0.11 μM, 5.83 ± 0.08 μM, and 7.56 ± 0.10 μM, respectively. Kinetic analysis, fluorescence quenching assays, and molecular docking experiments showed that Diosmetin, Fisetin, and Genistein docked near the same active site of XO, mainly affecting the microenvironment of tryptophan residues. These molecules showed static binding to XO via hydrogen bonds, hydrophobic interactions, and van der Waals forces. Diosmetin and Genistein were competitive inhibitors, whereas Fisetin was a mixed inhibitor. Infrared spectroscopy showed that Diosmetin, Fisetin, and Genistein increased the α-helix content of XO from 7.4 % to 16.6 %, 21.4 %, and 11.2 %, respectively, thereby enhancing its stability. In summary, the pharmacophore model constructed in this study was accurate. The flavonoids Diosmetin, Fisetin, and Genistein effectively inhibited the activity of XO, and the amino acid residues LEU257, ILE353, and VAL259 played a key role in the interaction between the flavonoids and XO. These findings are of great significance for the screening and development of new XO inhibitors.
Collapse
Affiliation(s)
- Yingxin Zhang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China
| | - Chenyu Ban
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China
| | - Dongyu Su
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China
| | - Yi Liu
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China
| | - Suzhen Zhou
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR China
| | - Jinbo Fan
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, PR 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
|
Vyas K, Prabaker S, Prabhu D, Sakthivelu M, Rajamanikandan S, Velusamy P, Su CH, Gopinath SCB, Pachaiappan R. Study of an inhibitory effect of plant polyphenolic compounds against digestive enzymes using bench-working experimental evidence predicted by molecular docking and dynamics. Int J Biol Macromol 2024; 259:129222. [PMID: 38185307 DOI: 10.1016/j.ijbiomac.2024.129222] [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: 10/21/2023] [Revised: 12/16/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
The substantial nutritional content and diversified biological activity of plant-based nutraceuticals are due to polyphenolic chemicals. These chemicals are important and well-studied plant secondary metabolites. Their protein interactions are extensively studied. This relationship is crucial for the logical development of functional food and for enhancing the availability and usefulness of polyphenols. This study highlights the influence of protein types and polyphenols on the interaction, where the chemical bindings predominantly consist of hydrophobic interactions and hydrogen bonds. The interaction between polyphenolic compounds (PCs) and digestive enzymes concerning their inhibitory activity has not been fully studied. Therefore, we have examined the interaction of four digestive enzymes (α-amylase, pepsin, trypsin, and α-chymotrypsin) with four PCs (curcumin, diosmin, morin, and 2',3',4'-trihydroxychalcone) through in silico and in vitro approaches. In vitro plate assays, enzyme kinetics, spectroscopic assays, molecular docking, and simulations were performed. We observed all these PCs have significant docking scores and preferable interaction with the active site of the digestive enzymes, resulting in the reduction of enzyme activity. The enzyme-substrate binding mechanism was determined using the Lineweaver Burk plot, indicating that the inhibition occurred competitively. Among four PCs diosmin and morin has the highest interaction energy over digestive enzymes with IC50 value of 1.13 ± 0.0047 and 1.086 ± 0.0131 μM. Kinetic studies show that selected PCs inhibited pepsin, trypsin, and chymotrypsin competitively and inhibited amylase in a non-competitive manner, especially by 2',3',4'-trihydroxychalcone. This study offers insights into the mechanisms by which the selected PCs inhibit the enzymes and has the potential to enhance the application of curcumin, diosmin, morin, and 2',3',4'-trihydroxychalcone as natural inhibitors of digestive enzymes.
Collapse
Affiliation(s)
- Kaushal Vyas
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Supraja Prabaker
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Dhamodharan Prabhu
- Centre for Drug Discovery, Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Meenakumari Sakthivelu
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Sundararaj Rajamanikandan
- Centre for Drug Discovery, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Palaniyandi Velusamy
- Research & Development, Sree Balaji Medical College and Hospital (SBMCH), Bharath Institute of Higher Education and research (BIHER), Chromepet 600 044, Tamil Nadu, India.
| | - Chia-Hung Su
- Department of Chemical Engineering, Ming Chi University of Technology, Taishan, Taipei 24301, Taiwan
| | - Subash C B Gopinath
- Faculty of Chemical Engineering & Technology, Universiti Malaysia Perlis (UniMAP), 02600 Arau, Perlis, Malaysia; Institute of Nano Electronic Engineering, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia; Micro System Technology, Centre of Excellence (CoE), Universiti Malaysia Perlis (UniMAP), Pauh Campus, 02600 Arau, Perlis, Malaysia; Department of Computer Science and Engineering, Faculty of Science and Information Technology, Daffodil International University, Daffodil Smart City, Birulia, Savar, Dhaka 1216, Bangladesh
| | - Raman Pachaiappan
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
6
|
Guo J, Gan C, Cheng B, Cui B, Yi F. Exploration of binding mechanism of apigenin to pepsin: Spectroscopic analysis, molecular docking, enzyme activity and antioxidant assays. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 290:122281. [PMID: 36584639 DOI: 10.1016/j.saa.2022.122281] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/07/2022] [Accepted: 12/25/2022] [Indexed: 06/17/2023]
Abstract
Pepsin plays an important role in nutrient metabolism. Apigenin (AP) is a beneficial polyphenol to human health. To enhance the bioavailability of AP and elucidate the inhibitory effect of AP on pepsin, the interaction mechanism of AP with pepsin was investigated using spectroscopic analysis and molecular docking, and the activity of pepsin and antioxidant activity of AP was also evaluated. Specifically, AP performed static quenching of pepsin and had only one binding site on pepsin. More interestingly, the interaction between AP and pepsin was spontaneous, while hydrogen bonds and van der Waals forces were the main binding forces. Generally, synchronous and three-dimensional fluorescence confirmed that AP induced the conformational changes of pepsin, and molecular docking proved the above results and illustrated the specific binding patterns. Specifically, AP inhibited the activity of pepsin, while pepsin decreased the antioxidant activity of AP. These results provided useful information for elucidating the interactions between AP and pepsin.
Collapse
Affiliation(s)
- Jinying Guo
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, PR China.
| | - Chuanfa Gan
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Bo Cheng
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Bo Cui
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, PR China
| | - Fankai Yi
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471023, PR China
| |
Collapse
|
7
|
Dong Y, Sun N, Ge Q, Lv R, Lin S. Antioxidant soy peptide can inhibit xanthine oxidase activity and improve LO2 cell damage. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
8
|
Zhang G, Zhu M, Liao Y, Gong D, Hu X. Action mechanisms of two key xanthine oxidase inhibitors in tea polyphenols and their combined effect with allopurinol. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:7195-7208. [PMID: 35727856 DOI: 10.1002/jsfa.12085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Tea polyphenols have been reported to have the effect of lowering uric acid. However, there are few studies on the inhibitory effects and molecular mechanisms of specific catechins on the urate-metabolizing enzyme xanthine oxidase (XO). In this research, multiple spectroscopic methods and computer simulations were used to determine the inhibitory ability and mechanisms of epigallocatechin gallate (EGCG) and gallocatechin gallate (GCG) on XO. RESULTS Herein, EGCG and GCG reversibly inhibited XO activity in a mixed manner, with IC50 values of 40.50 ± 0.32 and 33.60 ± 0.53 μmol L-1 , and also decreased the superoxide anion radical (O2 - ) of the catalytic system by reducing the XO molecule and inhibiting the formation of uric acid. The combination of EGCG or GCG with allopurinol showed synergistic inhibition on XO. The binding of EGCG or GCG to XO with moderate affinity formed a stable complex by hydrogen bonds and van der Waals forces. The presence of EGCG and GCG made the structure of XO more stable and compact. The two inhibitors bound to the vicinity of flavin adenine dinucleotide (FAD) in XO, hindering the entry of substrate; thus the activity of XO was suppressed. CONCLUSION Both EGCG and GCG are excellent natural XO inhibitors, and inhibited the activity of XO by occupying the channel of the substrate to enter the active center and interfering with the dual substrate reaction catalyzed by XO. These findings provide a scientific basis for the application of catechins in dietary supplements and medicines with lowering uric acid effects. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Guowen Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Miao Zhu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yijing Liao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Deming Gong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xing Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Network pharmacology and molecular docking analysis on molecular targets: Mechanisms of baicalin and baicalein against hyperuricemic nephropathy. Toxicol Appl Pharmacol 2021; 424:115594. [PMID: 34044073 DOI: 10.1016/j.taap.2021.115594] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022]
Abstract
Oxidative stress and inflammation in kidney are the main causes for hyperuricemic nephropathy (HN). Baicalin and baicalein, two flavonoids, have anti-inflammatory and anti-oxidative effects and they are interconvertible in the body. In this study, both baicalin and baicalein were administered by intragastric administration (i.g.) or intraperitoneal injection (i.p.) at the dose of 50 mg kg-1, once a day for 15 consecutive days to HN mice, a model established by i.g. of yeast extract combined with i.p. of potassium oxonate. In HN mice, baicalin and baicalein reduced serum uric acid (SUA) levels and protected kidneys by anti-inflammatory and anti-oxidative effects. Mechanistically, the effect of baicalin and baicalein on reducing SUA levels might due to their inhibitory effect on xanthine oxidase (XO) activity in vivo and in vitro. Furthermore, the mechanisms of baicalin and baicalein against HN were analyzed with network pharmacology and molecular docking technology. The network pharmacology indicated that the protective effects of baicalin and baicalein against HN were mainly related to their down-regulating effects on TLRs, NF-κB, MAPK, PI3K/AKT and NOD-like receptor signaling pathways. Molecular docking indicated high binding affinity of baicalin/baicalein to targets such as AKT1 and MAPK1. In summary, baicalin and baicalein are promising drug candidates for the treatment of HN by inhibiting XO activity, reducing inflammation and cell apoptosis through down-regulating TLRs/NLRP3/NF-κB, MAPK, PI3K/AKT/NF-κB pathways.
Collapse
|
10
|
Wang J, Sun S, Zhao K, Shi H, Fan J, Wang H, Liu Y, Liu X, Wang W. Insights into the inhibitory mechanism of purpurogallin on xanthine oxidase by multiple spectroscopic techniques and molecular docking. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
11
|
Chazelas P, Steichen C, Favreau F, Trouillas P, Hannaert P, Thuillier R, Giraud S, Hauet T, Guillard J. Oxidative Stress Evaluation in Ischemia Reperfusion Models: Characteristics, Limits and Perspectives. Int J Mol Sci 2021; 22:ijms22052366. [PMID: 33673423 PMCID: PMC7956779 DOI: 10.3390/ijms22052366] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemia reperfusion injury is a complex process consisting of a seemingly chaotic but actually organized and compartmentalized shutdown of cell function, of which oxidative stress is a key component. Studying oxidative stress, which results in an imbalance between reactive oxygen species (ROS) production and antioxidant defense activity, is a multi-faceted issue, particularly considering the double function of ROS, assuming roles as physiological intracellular signals and as mediators of cellular component damage. Herein, we propose a comprehensive overview of the tools available to explore oxidative stress, particularly in the study of ischemia reperfusion. Applying chemistry as well as biology, we present the different models currently developed to study oxidative stress, spanning the vitro and the silico, discussing the advantages and the drawbacks of each set-up, including the issues relating to the use of in vitro hypoxia as a surrogate for ischemia. Having identified the limitations of historical models, we shall study new paradigms, including the use of stem cell-derived organoids, as a bridge between the in vitro and the in vivo comprising 3D intercellular interactions in vivo and versatile pathway investigations in vitro. We shall conclude this review by distancing ourselves from "wet" biology and reviewing the in silico, computer-based, mathematical modeling, and numerical simulation options: (a) molecular modeling with quantum chemistry and molecular dynamic algorithms, which facilitates the study of molecule-to-molecule interactions, and the integration of a compound in a dynamic environment (the plasma membrane...); (b) integrative systemic models, which can include many facets of complex mechanisms such as oxidative stress or ischemia reperfusion and help to formulate integrated predictions and to enhance understanding of dynamic interaction between pathways.
Collapse
Affiliation(s)
- Pauline Chazelas
- Maintenance Myélinique et Neuropathies Périphériques, Université de Limoges, EA 6309, 87032 Limoges, France; (P.C.); (F.F.)
- Laboratoire de Biochimie et Génétique Moléculaire, CHU de Limoges, 87042 Limoges, France
| | - Clara Steichen
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86074 Poitiers, France
| | - Frédéric Favreau
- Maintenance Myélinique et Neuropathies Périphériques, Université de Limoges, EA 6309, 87032 Limoges, France; (P.C.); (F.F.)
- Laboratoire de Biochimie et Génétique Moléculaire, CHU de Limoges, 87042 Limoges, France
| | - Patrick Trouillas
- INSERM U1248, IPPRITT, Université de Limoges, 87032 Limoges, France;
- RCPTM, University Palacký of Olomouc, 771 47 Olomouc, Czech Republic
| | - Patrick Hannaert
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
| | - Raphaël Thuillier
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86074 Poitiers, France
- Service de Biochimie, CHU de Poitiers, 86021 Poitiers, France
| | - Sébastien Giraud
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Service de Biochimie, CHU de Poitiers, 86021 Poitiers, France
| | - Thierry Hauet
- INSERM U1082, IRTOMIT, 86021 Poitiers, France; (C.S.); (P.H.); (R.T.); (S.G.); (T.H.)
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86074 Poitiers, France
- Service de Biochimie, CHU de Poitiers, 86021 Poitiers, France
- FHU SUPORT Survival Optimization in Organ Transplantation, 86021 Poitiers, France
- IBiSA Plateforme Modélisation Préclinique-Innovations Chirurgicale et Technologique (MOPICT), Do-maine Expérimental du Magneraud, 17700 Surgères, France
| | - Jérôme Guillard
- UMR CNRS 7285 IC2MP, Team 5 Chemistry, Université de Poitiers, 86073 Poitiers, France
- Correspondence: ; Tel.: +33-5-49-44-38-59
| |
Collapse
|
12
|
Chen Y, Gao Y, Wu F, Luo X, Ju X, Liu G. Computationally exploring novel xanthine oxidase inhibitors using docking-based 3D-QSAR, molecular dynamics, and virtual screening. NEW J CHEM 2020. [DOI: 10.1039/d0nj03221b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Computationally exploring novel potential xanthine oxidase inhibitors using a systematic modeling study.
Collapse
Affiliation(s)
- Yanming Chen
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Ya Gao
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Fengshou Wu
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Xiaogang Luo
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Xiulian Ju
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| | - Genyan Liu
- Key Laboratory for Green Chemical Process of Ministry of Education
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology
- School of Chemical Engineering and Pharmacy
- Wuhan Institute of Technology
- Wuhan 430205
| |
Collapse
|