1
|
Su W, Chen H, Li Y, Wang Y, Chen T, Shi H, Yang J, Zhang C, Wang T, Xiong L. Design, synthesis and biological evaluation of 2-arylbenzo[b]furan-4-vinylcarbonyl derivatives based on Salvianolic acid C as antioxidant neuroprotective agents for the treatment of ischemic stroke. Eur J Med Chem 2025; 290:117506. [PMID: 40132493 DOI: 10.1016/j.ejmech.2025.117506] [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: 01/09/2025] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Ischemic stroke is a globally recognized disease characterized by high mortality and disability rates, with limited clinical treatment options available. The development of neuroprotective agents with antioxidant properties continues to be a focal point of current research. In this study, we designed and synthesized 42 derivatives using α, β-unsaturated carbonyl and 2-arylbenzo[b]furan in Salvianolic acid C as the core skeleton, and evaluated their biological activities. Among these, compound 6p demonstrated notable antioxidant neuroprotective activity and low cytotoxicity. Furthermore, it exhibited the most potent cell protective activity and ROS scavenging capacity in t-BHP-induced PC12 cells. In a rat model of ischemia-reperfusion injury, 6p was found to reduce ROS levels and neuronal apoptosis in brain tissue, enhance neurological function, and decrease the size of cerebral infarction in rats. Additionally, 6p promotes the nuclear translocation of NRF2 and elevates the expression of the antioxidant protein HO-1. Molecular docking results indicated that 6p can bind to key sites within KEAP1 complex. In conclusion, these findings suggest that compound 6p serves as a potential neuroprotective agent for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Wei Su
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Heming Chen
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yi Li
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yun Wang
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Ting Chen
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Hao Shi
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jian Yang
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Chuan Zhang
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Tingfang Wang
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Liyan Xiong
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
2
|
Er-Rajy M, El Fadili M, Zarougui S, Mujwar S, Aloui M, Zerrouk M, Hammouti B, Rhazi L, Sabbahi R, Alanazi MM, Azzaoui K, Salghi R, Elhallaoui M. Design and evaluation of novel triazole derivatives as potential anti-gout inhibitors: a comprehensive molecular modeling study. Front Chem 2025; 13:1518777. [PMID: 40115054 PMCID: PMC11922854 DOI: 10.3389/fchem.2025.1518777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/30/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Gout is the most common inflammatory arthritis, characterized by hyperuricemia, tophus formation, joint disease, and kidney stones. Uric acid, the final byproduct of purine catabolism, is eliminated via the kidneys and digestive system. Xanthine oxidase (XO) catalyzes the conversion of hypoxanthine and xanthine into uric acid, making XO inhibitors crucial for treating hyperuricemia and gout. Currently, three XO inhibitors are clinically used, showing significant efficacy. A molecular modeling study on triazole derivatives aims to identify novel XO inhibitors using 3D-QSAR, molecular docking, MD simulations, ADMET analysis, and DFT calculations. These computational approaches facilitate drug discovery while reducing research costs. Methods Our work focuses on a series of synthesized anti-xanthine oxidase inhibitors, aiming to develop new inhibitors. A computational study was carried out to identify the xanthine oxidase inhibitory structural features of a series of triazole inhibitors using computational method. Results A model based on CoMFA and CoMSIA/SEA has been built to predict new triazole derivatives. Discussion The optimal model established from CoMFA and CoMSIA/SEA was successfully evaluated for its predictive capability. Visualization of the contour maps of both models showed that modifying the substituents plays a key role in enhancing the biological activity of anti-gout inhibitors. Molecular docking results for complexes N°8-3NVY and N°22-3NVY showed scores of -7.22 kcal/mol and -8.36 kcal/mol, respectively, indicating substantial affinity for the enzyme. Complex N°8-3NVY forms two hydrogen bonds with SER 69 and ASN 71, three alkyl bonds with ALA 70, LEU 74, and ALA 75, and one Pi-Pi T-shaped bond with PHE 68. Complex N°22-3NVY forms three hydrogen bonds with HIS 99, ARG 29, and ILE 91, and one halogen bond with LEU 128 at 3.60 Å. A MD study revealed that the N°22-3NVY complex remained highly stable throughout the simulation. Therefore, we proposed six new molecules, their anti-gout inhibitory activities were predicted using two models, and they were evaluated for Lipinski's rule, and ADMET properties. The results show that both Pred 4 and Pred 5 have better pharmacokinetic properties than the height potent molecule in the studied series, making these two compounds valuable candidates for new anti-gout drugs. Subsequently, using DFT study to evaluate the chemical reactivity properties of these two proposed compounds, the energy gap results revealed that both molecules exhibit moderate chemical stability and reactivity.
Collapse
Affiliation(s)
- Mohammed Er-Rajy
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Euromed University of Fes, UMF, Fez, Morocco
| | - Mohamed El Fadili
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Sara Zarougui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Mourad Aloui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mohammed Zerrouk
- Engineering Laboratory of Organometallic, Molecular Materials and Environment, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | | | - Larbi Rhazi
- Institut Polytechnique UniLaSalle, Université d'Artois, Beauvais, France
| | - Rachid Sabbahi
- Research Team in Science and Technology, Higher School of Technology, Ibn Zohr University, Laayoune, Morocco
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalil Azzaoui
- Engineering Laboratory of Organometallic, Molecular Materials and Environment, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez, Morocco
- Laboratory of Industrial Engineering, Energy and the Environment (LI3E) SUPMTI, Rabat, Morocco
| | - Rachid Salghi
- Laboratory of Applied Chemistry and Environment, National School of Applied Sciences, University Ibn Zohr, Agadir, Morocco
| | - Menana Elhallaoui
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
3
|
Han C, Wu Y, Rong J, Xia Q, Du D. Unveiling the Emerging Role of Xanthine Oxidase in Acute Pancreatitis: Beyond Reactive Oxygen Species. Antioxidants (Basel) 2025; 14:95. [PMID: 39857429 PMCID: PMC11759826 DOI: 10.3390/antiox14010095] [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: 10/22/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Acute pancreatitis (AP) is a potentially fatal acute digestive disease that is widespread globally. Although significant progress has been made in the previous decade, the study of mechanisms and therapeutic strategies is still far from being completed. Xanthine oxidase (XO) is an enzyme that catalyzes hypoxanthine and xanthine to produce urate and is accompanied by the generation of reactive oxygen species (ROS) in purine catabolism. Considerable preclinical and clinical studies have been conducted over many decades to investigate the role of XO in the pathogenesis of AP and its potential targeting therapeutic value. There is no doubt that the ROS generated by irreversibly activated XO participates in the local pancreas and multiple organ failure during AP. However, the optimal timing and doses for therapeutic interventions targeting XO in animal studies and the clinic, as well as the additional molecular mechanisms through which XO contributes to disease onset and progression, including metabolic regulation, remain to be elucidated. This review summarized the benefits and contradictions of using XO inhibitors in animal models, offered mechanisms other than ROS, and discussed the difficulties faced in clinical trials. We hope to provide a perspective on the future worthwhile basic and clinical research on XO by analyzing its chemical and biological characteristics, as well as the progress of its regulatory mechanisms in AP.
Collapse
Affiliation(s)
- Chenxia Han
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaling Wu
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Juan Rong
- Department of Gastroenterology, The Third People’s Hospital of Chengdu, Chengdu 610031, China
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Du
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Rashad AY, Daabees HG, Elagawany M, Shahin M, Abdel Moneim AE, Marei MY, Rostom SAF. Dual inhibition strategy addressing hyperuricemia and oxidative stress: design, biological evaluation and stability studies of febuxostat-probenecid mutual prodrug. Med Chem Res 2024; 33:2476-2490. [DOI: 10.1007/s00044-024-03317-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/17/2024] [Indexed: 05/14/2025]
|
5
|
Sun Z, Zhang X, Zhao Z, Li X, Pang J, Chen J. Recent Progress and Future Perspectives on Anti-Hyperuricemic Agents. J Med Chem 2024; 67:19966-19987. [PMID: 39513478 DOI: 10.1021/acs.jmedchem.4c01260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Increased biosynthesis or underexcretion of uric acid (UA or urate) in the body ultimately leads to the development of hyperuricemia. Epidemiological studies indicate that hyperuricemia is closely associated with the occurrence of various diseases such as gout and cardiovascular diseases. Currently, the first-line therapeutic medications used to reduce UA levels primarily include xanthine oxidase (XO) inhibitors, which limit UA production, and urate transporter 1 (URAT1) inhibitors, which decrease urate reabsorption and enhance urate excretion. Despite significant progress in urate-lowering therapies, long-term use of these drugs can cause hepatorenal toxicity as well as cardiovascular complications. Therefore, there is an urgent need for novel anti-hyperuricemic agents with better efficacy and lower toxicity. This perspective mainly focuses on the current research progress and design strategy of anti-hyperuricemic agents, particularly those targeting XO and URAT1. It is our hope that this perspective will provide insights into the challenges and opportunities for anti-hyperuricemic drug discovery.
Collapse
Affiliation(s)
- Zhiqiang Sun
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuewen Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zean Zhao
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoxun Li
- Chengdu Easton Biopharmaceuticals Co., Ltd., Chengdu 611731, China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
6
|
Guo S, Sun Q, Zhang X, Li SY, Liu HY, Ge GH, Wang J, Liu XY, Xu B, Li TT, Zhou XF, Wang YP, Meng FH, Zhang TJ. Discovery of 4-(isopentyloxy)-3-nitrobenzamide derivatives as xanthine oxidase inhibitors through a non-anthraquinone exploration. Arch Pharm (Weinheim) 2024; 357:e2400137. [PMID: 38963324 DOI: 10.1002/ardp.202400137] [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: 02/21/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
In our previous study, we reported a series of N-(9,10-anthraquinone-2-carbonyl) amino acid derivatives as novel inhibitors of xanthine oxidase (XO). Recognizing the suboptimal drug-like properties associated with the anthraquinone moiety, we embarked on a nonanthraquinone medicinal chemistry exploration in the current investigation. Through systematic structure-activity relationship (SAR) studies, we identified a series of 4-(isopentyloxy)-3-nitrobenzamide derivatives exhibiting excellent in vitro potency against XO. The optimized compound, 4-isopentyloxy-N-(1H-pyrazol-3-yl)-3-nitrobenzamide (6k), demonstrated exceptional in vitro potency with an IC50 value of 0.13 μM. Compound 6k showed favorable drug-like characteristics with ligand efficiency (LE) and lipophilic ligand efficiency (LLE) values of 0.41 and 3.73, respectively. In comparison to the initial compound 1d, 6k exhibited a substantial 24-fold improvement in IC50, along with a 1.6-fold enhancement in LE and a 3.7-fold increase in LLE. Molecular modeling studies provided insights into the strong interactions of 6k with critical amino acid residues within the active site. Furthermore, in vivo hypouricemic investigations convincingly demonstrated that 6k significantly reduced serum uric acid levels in rats. The MTT results revealed that compound 6k is nontoxic to healthy cells. The gastric and intestinal stability assay demonstrated that compound 6k exhibits good stability in the gastric and intestinal environments. In conclusion, compound 6k emerges as a promising lead compound, showcasing both exceptional in vitro potency and favorable drug-like characteristics, thereby warranting further exploration.
Collapse
Affiliation(s)
- Shuai Guo
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Qi Sun
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Xu Zhang
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Song-Ye Li
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Hong-Ye Liu
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Gong-Hui Ge
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Jing Wang
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Xing-Yang Liu
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Ben Xu
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Ting-Ting Li
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Xian-Feng Zhou
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Yan-Ping Wang
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Fan-Hao Meng
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| | - Ting-Jian Zhang
- School of Pharmacy/Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang, China
| |
Collapse
|
7
|
Sun Q, Yu W, Gong M, Ma J, Liu G, Mei T, Luo X. Xanthine oxidase immobilized cellulose membrane-based colorimetric biosensor for screening and detecting the bioactivity of xanthine oxidase inhibitors. Int J Biol Macromol 2024; 275:133450. [PMID: 38944077 DOI: 10.1016/j.ijbiomac.2024.133450] [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: 12/21/2023] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
Xanthine oxidase (XO) is a typical target for hyperuricemia and gout, for which there are only three commercial xanthine oxidase inhibitors (XOIs): febuxostat, topiroxostat and allopurinol. However, these inhibitors have problems such as low bioactivity and several side effects. Therefore, the development of novel XOIs with high bioactivity for the treatment of hyperuricemia and gout is urgently needed. In this work we constructed a XO immobilized cellulose membrane colorimetric biosensor (XNCM) by the TEMPO oxidation, amide bond coupling and nitro blue tetrazolium chloride (NBT) loading method. As expected, the XNCM was able to detect xanthine, with high selectivity and sensitivity by colorimetric method with a distinctive color change from yellow to purple, which can be easily observed by the naked-eye in just 8 min without any complex instrumentation. In addition, the XNCM sensor performed screening of 21 different compounds and have been successfully pre-screened out XOIs with biological activity. Most importantly, the XNCM was able to quantitatively detect the IC50 values of two commercial inhibitors (febuxostat and allopurinol). All the results confirmed that the XNCM is a simple and effective tool which can be used for the accelerated screening of XOIs and has the potential to uncover additional XOIs.
Collapse
Affiliation(s)
- Qi Sun
- School of Chemistry and Environmental Engineering, Hubei key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum, Wuhan Institute of Technology, Wuhan 430205, PR China
| | - Wenlong Yu
- School of Chemistry and Environmental Engineering, Hubei key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum, Wuhan Institute of Technology, Wuhan 430205, PR China
| | - Mixue Gong
- School of Chemistry and Environmental Engineering, Hubei key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum, Wuhan Institute of Technology, Wuhan 430205, PR China
| | - Jingfang Ma
- School of Chemistry and Environmental Engineering, Hubei key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum, Wuhan Institute of Technology, Wuhan 430205, PR China
| | - Genyan Liu
- School of Chemistry and Environmental Engineering, Hubei key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum, Wuhan Institute of Technology, Wuhan 430205, PR China.
| | - Tao Mei
- Key Laboratory of Polymer Materials, Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science and Engineering, Hubei University, Wuhan 430062, PR China.
| | - Xiaogang Luo
- School of Chemistry and Environmental Engineering, Hubei key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum, Wuhan Institute of Technology, Wuhan 430205, PR China.
| |
Collapse
|
8
|
Huang D, Li W, Zhao Y, Xie C, Luo X, Wu F, Xu Z, Sun Q, Liu G. Design, synthesis, and biological evaluation of 5-(1H-indol-5-yl)isoxazole-3-carboxylic acids as novel xanthine oxidase inhibitors. Eur J Med Chem 2024; 271:116443. [PMID: 38691887 DOI: 10.1016/j.ejmech.2024.116443] [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: 11/03/2023] [Revised: 04/07/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
Xanthine oxidase (XO) is a key enzyme for the production of uric acid in the human body. XO inhibitors (XOIs) are clinically used for the treatment of hyperuricemia and gout, as they can effectively inhibit the production of uric acid. Previous studies indicated that both indole and isoxazole derivatives have good inhibitory effects against XO. Here, we designed and synthesized a novel series of N-5-(1H-indol-5-yl)isoxazole-3-carboxylic acids according to bioisosteric replacement and hybridization strategies. Among the obtained target compounds, compound 6c showed the best inhibitory activity against XO with an IC50 value of 0.13 μM, which was 22-fold higher than that of the classical antigout drug allopurinol (IC50 = 2.93 μM). Structure-activity relationship analysis indicated that the hydrophobic group on the nitrogen atom of the indole ring is essential for the inhibitory potencies of target compounds against XO. Enzyme kinetic studies proved that compound 6c acted as a mixed-type XOI. Molecular docking studies showed that the target compound 6c could not only retain the key interactions similar to febuxostat at the XO binding site but also generate some new interactions, such as two hydrogen bonds between the oxygen atom of the isoxazole ring and the amino acid residues Ser876 and Thr1010. These results indicated that 5-(1H-indol-5-yl)isoxazole-3-carboxylic acid might be an efficacious scaffold for designing novel XOIs and compound 6c has the potential to be used as a lead for further the development of novel anti-gout candidates.
Collapse
Affiliation(s)
- Dongqian Huang
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Wenye Li
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Yilan Zhao
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Cheng Xie
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Xiaogang Luo
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China; School of Materials Science and Engineering, Zhengzhou University, No.100 Science Avenue, Zhengzhou City, 450001, Henan Province, PR China
| | - Fengshou Wu
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Zhiqiang Xu
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China
| | - Qi Sun
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China.
| | - Genyan Liu
- Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China.
| |
Collapse
|
9
|
Sun ZG, Wu KX, Ullah I, Zhu HL. Recent Advances in Xanthine Oxidase Inhibitors. Mini Rev Med Chem 2024; 24:1177-1186. [PMID: 37711003 DOI: 10.2174/1389557523666230913091558] [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: 04/25/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 09/16/2023]
Abstract
Uric acid is a product of purine nucleotide metabolism, and high concentrations of uric acid can lead to hyperuricemia, gout and other related diseases. Xanthine oxidase, the only enzyme that catalyzes xanthine and hypoxanthine into uric acid, has become a target for drug development against hyperuricemia and gout. Inhibition of xanthine oxidase can reduce the production of uric acid, so xanthine oxidase inhibitors are used to treat hyperuricemia and related diseases, including gout. In recent years, researchers have obtained new xanthine oxidase inhibitors through drug design, synthesis, or separation of natural products. This paper summarizes the research on xanthine oxidase inhibitors since 2015, mainly including natural products, pyrimidine derivatives, triazole derivatives, isonicotinamide derivatives, chalcone derivatives, furan derivatives, coumarin derivatives, pyrazole derivatives, and imidazole derivatives, hoping to provide valuable information for the research and development of novel xanthine oxidase inhibitors.
Collapse
Affiliation(s)
- Zhi-Gang Sun
- Central Laboratory, Linyi Central Hospital, No. 17 Jiankang Road, Linyi, 276400, China
| | - Kai-Xiang Wu
- School of Clinical Medicine, Jining Medical University, No. 133 Hehua Road, Jining, 272067, China
| | - Inam Ullah
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No. 163 Xianlin Road, Nanjing, 210023, China
| | - Hai-Liang Zhu
- Central Laboratory, Linyi Central Hospital, No. 17 Jiankang Road, Linyi, 276400, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No. 163 Xianlin Road, Nanjing, 210023, China
| |
Collapse
|
10
|
Singh A, Singh K, Sharma A, Kaur K, Chadha R, Singh Bedi PM. Past, present and future of xanthine oxidase inhibitors: design strategies, structural and pharmacological insights, patents and clinical trials. RSC Med Chem 2023; 14:2155-2191. [PMID: 37974965 PMCID: PMC10650961 DOI: 10.1039/d3md00316g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/06/2023] [Indexed: 11/19/2023] Open
Abstract
Xanthine oxidase, a molybdo-flavoenzyme, and an isoform of xanthine dehydrogenase both exist as xanthine oxidoreductase and are responsible for purine catabolism. Xanthine oxidase is more involved in pathological conditions when extensively modulated. Elevation of xanthine oxidase is not only the prime cause of gout but is also responsible for various hyperuricemia associated pathological conditions like diabetes, chronic wounds, cardiovascular disorders, Alzheimer's disease, etc. Currently available xanthine oxidase inhibitors in clinical practice (allopurinol, febuxostat and topiroxostat) suffer from fatal side effects that pose a serious problem to the healthcare system, raising global emergency to develop novel, potent and safer xanthine oxidase inhibitors. This review will provide key and systematic information about: a. design strategies (inspired from both marketed drugs in clinical practice and natural products), structural insights and pharmacological output (xanthine oxidase inhibition and associated activities) of various pre-clinical candidates reported by various research groups across the globe in the past two decades; b. patented xanthine oxidase inhibitors published in the last three decades and c. clinical trials and their outcomes on approved drug candidates. Information generated in this review has suggested fragment-based drug design (FBDD) and molecular hybridization techniques to be most suitable for development of desired xanthine oxidase inhibitors as one provides high selectivity toward the enzyme and the other imparts multifunctional properties to the structure and both may possess capabilities to surpass the limitations of currently available clinical drugs. All in combination will exclusively update researchers working on xanthine oxidase inhibitors and allied areas and potentially help in designing rational, novel, potent and safer xanthine oxidase inhibitors that can effectively tackle xanthine oxidase related disease conditions and disorders.
Collapse
Affiliation(s)
- Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Karanvir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Aman Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Kirandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Renu Chadha
- University Institute of Pharmaceutical Sciences, Panjab University Chandigarh 160014 India
| | - Preet Mohinder Singh Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
- Drug and Pollution Testing Laboratory, Guru Nanak Dev University Amritsar Punjab 143005 India
| |
Collapse
|
11
|
Rashad AY, Daabees HG, Elagawany M, Shahin M, Abdel Moneim AE, Rostom SAF. A New Avenue for Enhanced Treatment of Hyperuricemia and Oxidative Stress: Design, Synthesis and Biological Evaluation of Some Novel Mutual Prodrugs Involving Febuxostat Conjugated with Different Antioxidants. Bioorg Chem 2023; 140:106818. [PMID: 37688830 DOI: 10.1016/j.bioorg.2023.106818] [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: 06/02/2023] [Revised: 08/13/2023] [Accepted: 08/27/2023] [Indexed: 09/11/2023]
Abstract
Febuxostat (FEB) is the first non-purine xanthine oxidase inhibitor (XOI) used for the treatment of hyperuricemia and gout. The oxidative stress induced by reactive oxygen species (ROS) which accompany purine metabolism by XO, could contribute to cellular damage and several pathological conditions. In this view, the present work addresses the evaluation of combining the hypouricemic effect of FEB and the free radical scavenging potential of various natural antioxidants in a single chemical entity by implementing the "mutual prodrug" strategy. Accordingly, a series of five ester prodrugs containing FEB together with different naturally occurring antioxidants namely, thioctic acid (4), thymol (5), menthol (6), vanillin (7), and guaiacol (8) was synthesized. Prominently, all the chemically conjugated prodrugs (4 - 8) revealed an obvious increase in the hypouricemic and antioxidant potentials when compared with their corresponding promoieties and physical mixtures. Moreover, they showed a potential protective effect against CCl4-induced hepatotoxicity and oxidative stress, together with no cytotoxicity on normal breast cells (MCF10A). Furthermore, the in vitro chemical and enzymatic stability studies of the prodrugs (4 - 8) using a developed HPLC method, verified their stability in different pHs, and rapid hydrolysis in rabbit plasma and liver homogenate to their parent metabolites. Moreover, the prodrugs (4 - 8) showed higher lipophilicity and lower aqueous solubility when compared to the parent drugs. Finally, the obtained merits from the implementation of the mutual prodrug strategy would encourage further application in the development of promising candidates with high therapeutic efficacy and improved safety profiles.
Collapse
Affiliation(s)
- Aya Y Rashad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira 22516, Egypt
| | - Hoda G Daabees
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira 22516, Egypt
| | - Mohamed Elagawany
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira 22516, Egypt
| | - Mohamed Shahin
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, El-Buhaira 22516, Egypt
| | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Sherif A F Rostom
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
12
|
Li R, Zhang Z, Chen J, Li H, Tang H. Investigating of zein-gum arabic-tea polyphenols ternary complex nanoparticles for luteolin encapsulation: Fabrication, characterization, and functional performance. Int J Biol Macromol 2023:125059. [PMID: 37244348 DOI: 10.1016/j.ijbiomac.2023.125059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Luteolin has extensive biological effects, but its low water-solubility and oral bioavailability have restricted its application. In this study, we successfully prepared new zein-gum arabic (GA)-tea polyphenols (TP) ternary complex nanoparticles (ZGTL) as a delivery system to encapsulate luteolin using an anti-solvent precipitation method. Consequently, ZGTL nanoparticles showed negatively charged smooth spherical structures with smaller particle size and higher encapsulation ability. X-ray diffraction revealed the amorphous state of luteolin in the nanoparticles. Hydrophobic, electrostatic, and hydrogen bonding interactions contributed to the formation and stability of ZGTL nanoparticles, as indicated by fluorescence and Fourier transform infrared spectra analyses. The inclusion of TP improved the physicochemical stability and luteolin retention rate of ZGTL nanoparticles by forming more compact nanostructures under different environmental conditions, including pH, salt ion concentration, temperature, and storage. Additionally, ZGTL nanoparticles exhibited stronger antioxidant activity and better sustainable release capacity under simulated gastrointestinal conditions due to TP incorporation. These findings demonstrate that ZGT complex nanoparticles have potential applications as an effective delivery system for encapsulating bioactive substances in food and medicine fields.
Collapse
Affiliation(s)
- Renjie Li
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Zhuangwei Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Jin Chen
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Huihui Li
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Hongjin Tang
- College of Biological and Food Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| |
Collapse
|
13
|
Hu SS, Zhang TJ, Wang ZR, Xu EY, Wang QY, Zhang X, Guo S, Ge GH, Wang J, Meng FH. Design, synthesis and structure-activity relationship of N-phenyl aromatic amide derivatives as novel xanthine oxidase inhibitors. Bioorg Chem 2023; 133:106403. [PMID: 36801790 DOI: 10.1016/j.bioorg.2023.106403] [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: 10/15/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Our previous studies suggested that N-phenyl aromatic amides are a class of promising xanthine oxidase (XO) inhibitor chemotypes. In this effort, several series of N-phenyl aromatic amide derivatives (4a-h, 5-9, 12i-w, 13n, 13o, 13r, 13s, 13t and 13u) were designed and synthesized to carry out an extensive structure-activity relationship (SAR). The investigation provided some valuable SAR information and identified N-(3-(1H-imidazol-1-yl)-4-((2-methylbenzyl)oxy)phenyl)-1H-imidazole-4-carboxamide (12r, IC50 = 0.028 µM) as the most potent XO inhibitor with close in vitro potency to that of topiroxostat (IC50 = 0.017 µM). Molecular docking and molecular dynamics simulation rationalized the binding affinity through a series of strong interactions with the residues Glu1261, Asn768, Thr1010, Arg880, Glu802, etc. In vivo hypouricemic studies also suggested that the uric acid lowering effect of compound 12r was improved compared with the lead g25 (30.61 % vs 22.4 % reduction in uric acid levels at 1 h; 25.91 % vs 21.7 % reduction in AUC of uric acid) . Pharmacokinetic studies revealed that compound 12r presented a short t1/2 of 0.25 h after oral administration. In addition, 12r has non-cytotoxicity against normal cell HK-2. This work may provide some insights for further development of novel amide-based XO inhibitors.
Collapse
Affiliation(s)
- Sen-Sen Hu
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Ting-Jian Zhang
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Zhao-Ran Wang
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - En-Yu Xu
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Qiu-Yin Wang
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Xu Zhang
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Shuai Guo
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Gong-Hui Ge
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Jing Wang
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, 77, Puhe Road, North New Area, Shenyang 110122, China.
| |
Collapse
|
14
|
Rashad AY, Daabees HG, Elagawany M, Shahin M, Abdel Moneim AE, Rostom SA. Towards the Development of Dual Hypouricemic and Anti-inflammatory Candidates: Design, Synthesis, Stability Studies and Biological Evaluation of Some Mutual Ester Prodrugs of Febuxostat-NSAIDs. Bioorg Chem 2023; 135:106502. [PMID: 37030108 DOI: 10.1016/j.bioorg.2023.106502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/03/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Treatment of gout involves two basic approaches: reducing the serum uric acid mainly by xanthine oxidase inhibitors (XOIs) and alleviating the intensity of the accompanying acute arthritic inflammation using non-steroidal anti-inflammatory drugs (NSAIDs). Febuxostat (FEB) is the first non-purine XOI approved for the treatment of hyperuricemia and gout. The present study aims at combining the hypouricemic effect of FEB and the anti-inflammatory (AI) properties of NSAIDs in a single entity by adopting the "mutual prodrug" approach. Accordingly, a series of seven ester prodrugs comprising basically FEB together with different NSAIDs namely, diclofenac (4), ibuprofen (5), ketoprofen (6), indomethacin (7), naproxen (8), ketorolac (9) and etodolac (10) was synthesized. All the investigated seven prodrugs (4-10) were equipotent or even superior to their corresponding parent drugs in the hypouricemic and AI activities, together with a gastrointestinal (GI) safety profile. Among this series, the prodrug FEB-DIC (4) showed excellent dual in vivo hypouricemic and anti-inflammatory activity (43.60 % and 15.96 %, respectively) when compared to the parent drugs FEB and diclofenac (36.82 % and 12.10 %, respectively) and its physical mixture (37.28 % and 12.41 %, respectively). Investigation of the in vitro chemical stability and hydrolysis of the prodrug (4) in aqueous and biological samples using a developed HPLC method confirmed its stability in various pHs, whereas rapid hydrolysis to the parent drugs in liver homogenate and human plasma was proven. Finally, it is concluded that the mutual prodrug approach could be successfully used in drug design and development for overcoming undesirable difficulties without losing the desired activities of the parent drugs.
Collapse
|
15
|
Ten Years Milestones in Xanthine Oxidase Inhibitors Discovery: Febuxostat-Based Inhibitors Trends, Bifunctional Derivatives, and Automatized Screening Assays. ORGANICS 2022. [DOI: 10.3390/org3040026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Xanthine oxidase (XO) is an enzyme involved in the oxidative process of hypoxanthine and xanthine to uric acid (UA). This process also produces reactive oxygen species (ROS) as byproducts. Both UA and ROS are dangerous for human health, and some health conditions trigger upregulation of XO activity, which results in many diseases (cancer, atherosclerosis, hepatitis, gout, and others) given the worsened scenario of ROS and UA overproduction. So, XO became an attractive target to produce and discover novel selective drugs based on febuxostat, the most recent XO inhibitor out of only two approved by FDA. Under this context, high-performance liquid chromatography (HPLC) and capillary electrophoresis (CE) have been successfully applied to rapidly and easily screen for bioactive compounds, isolated or in complex natural matrixes, that act as enzyme inhibitors through the use of an immobilized enzyme reactor (IMER). This article’s goal is to present advances comprising febuxostat-based XO inhibitors as a new trend, bifunctional moieties capable of inhibiting XO and modulating ROS activity, and in-flow techniques employing an IMER in HPLC and CE to screen for synthetic and natural compounds that act as XO inhibitors.
Collapse
|
16
|
Chen JS, Wang MX, Wang MM, Zhang YK, Guo X, Chen YY, Zhang MQ, Sun JY, Liu YF, Liu C. Synthesis and biological evaluation of geniposide derivatives as inhibitors of hyperuricemia, inflammatory and fibrosis. Eur J Med Chem 2022; 237:114379. [DOI: 10.1016/j.ejmech.2022.114379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 01/16/2023]
|
17
|
Kaur G, Singh A, Arora G, Monga A, Jassal AK, Uppal J, Bedi PMS, Bora KS. Synthetic heterocyclic derivatives as promising xanthine oxidase inhibitors: An overview. Chem Biol Drug Des 2022; 100:443-468. [PMID: 35763448 DOI: 10.1111/cbdd.14109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/19/2022] [Accepted: 06/26/2022] [Indexed: 11/28/2022]
Abstract
Inhibition of xanthine oxidase is an effective and most prominent therapeutic approach for the management of gout. Discovery of its association in the pathophysiology of diabetes, cardiovascular disorders, etc., widened its therapeutic horizons. Limited drug candidates in clinical practice along with side effects forced researchers to develop more efficacious and safer xanthine oxidase inhibitors for the management of gout and other disorders associated with xanthine oxidase hyperactivity. In this regard, this review focus on: (a) Various drug candidates in clinical practice and under clinical trials, (b) Development of various heterocyclic motifs as xanthine oxidase inhibitors in last two decades and (c) Various patented synthetic xanthine oxidase inhibitors.
Collapse
Affiliation(s)
- Gurinder Kaur
- University Institute of Pharma. Sciences, Chandigarh University, Mohali, Punjab, India
| | - Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Geetakshi Arora
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Aditi Monga
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Anupmjot Kaur Jassal
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Jasreen Uppal
- University Institute of Pharma. Sciences, Chandigarh University, Mohali, Punjab, India
| | - Preet Mohinder Singh Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India.,Drug and Pollution testing Laboratory, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Kundan Singh Bora
- University Institute of Pharma. Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
18
|
Zhang TJ, Zhang Y, Zhang ZH, Wang ZR, Zhang X, Hu SS, Lu PF, Guo S, Meng FH. Discovery of 4-(phenoxymethyl)-1H-1,2,3-triazole derivatives as novel xanthine oxidase inhibitors. Bioorg Med Chem Lett 2022; 60:128582. [DOI: 10.1016/j.bmcl.2022.128582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/23/2021] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
|
19
|
Tu S, Zhang TJ, Zhang Y, Zhang X, Zhang ZH, Meng FH. N-(3-cyano-1H-indol-5-yl)isonicotinamide and N-(3-cyano-1H-indol-5-yl)-1H-benzo[d]imidazole-5-carboxamide derivatives: Novel amide-based xanthine oxidase inhibitors. Bioorg Chem 2021; 115:105181. [PMID: 34329991 DOI: 10.1016/j.bioorg.2021.105181] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022]
Abstract
Our previous work demonstrated that amide is an efficient linker to explore chemical space of xanthine oxidase (XO) inhibitors that are entirely different from febuxostat and topiroxostat. In this effort, with 3-cyano-1H-indol-5-yl as a key moiety, two series of amide-based XO inhibitors, N-(3-cyano-1H-indol-5-yl)isonicotinamides (2a-w) and N-(3-cyano-1H-indol-5-yl)-1H-benzo[d]imidazole-5-carboxamides (3a-i), were designed and synthesized. The structure-activity relationship investigation identified N-(3-cyano-1-cyclopentyl-1H-indol-5-yl)-1H-benzo[d]imidazole-5-carboxamide (3i, IC50 = 0.62 μM) as the most promising compound, with 14.4-fold higher in vitro inhibitory potency than allopurinol (IC50 = 8.91 μM). Molecular simulations provided reasonable interaction modes for the representative compounds. Furthermore, in vivo activity evaluation demonstrated that compound 3i (oral dose of 12.8 mg/kg) has obviously hypouricemic effect on a potassium oxonate induced hyperuricemic rat model. Cytotoxicity assay and ADME prediction also supported that 3i is an excellent lead for further exploration of amide-based XO inhibitors.
Collapse
Affiliation(s)
- Shun Tu
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Ting-Jian Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Yi Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Xu Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Zhen-Hao Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China.
| |
Collapse
|
20
|
Febuxostat-based amides and some derived heterocycles targeting xanthine oxidase and COX inhibition. Synthesis, in vitro and in vivo biological evaluation, molecular modeling and in silico ADMET studies. Bioorg Chem 2021; 113:104948. [PMID: 34052736 DOI: 10.1016/j.bioorg.2021.104948] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022]
Abstract
Various febuxostat derivatives comprising carboxamide functionalities and different substituted heterocycles were synthesized and evaluated for their biological activities as xanthine oxidase (XO) and cyclooxygenase (COX) inhibitors. All the tested compounds exhibited variable in vitro XO inhibitory activities (IC50 values 0.009-0.077 µM), among which the analog 17 has emerged as the most potent derivative (IC50 0.009 µM), representing nearly 3-times the potency of febuxostat (IC50 0.026 µM). The same analogs were further investigated for their in vitro COX-1 and COX-2 inhibitory activity, where fifteen analogs demonstrated recognizable COX-2 inhibitory potential (IC50 values range 0.04 - 0.1 µM), when correlated with celecoxib (IC50 0.05 µM), together with appreciable selectivity indices. Compounds 5a, 14b, 17, 19c, 19e and 21b that showed significant in vitro XO and/ or COX inhibitory potentials were further investigated for their in vivo hypouricemic as well as anti-inflammatory activities. Interestingly, the in vivo results were concordant with the collected in vitro data. Docking of compounds 5a, 14b, 17, 19c, 19e and 21b with the active sites of XO and COX-2 isozymes demonstrated superior binding profile compared with the reported ligands (febuxostat and celecoxib, respectively). Their docking scores were reasonable and cohering to a great extent with their corresponding in vitro IC50 values. Moreover, in silico computation of the predicted pharmacokinetic and toxicity properties (ADMET), together with the ligand efficiency (LE) of the same six compounds suggesting their liability to act as new orally active drug candidates with a predicted high safety profile.
Collapse
|
21
|
Orhan IE, Deniz FSS. Natural Products and Extracts as Xantine Oxidase Inhibitors - A Hope for Gout Disease? Curr Pharm Des 2021; 27:143-158. [PMID: 32723252 DOI: 10.2174/1381612826666200728144605] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 06/25/2020] [Indexed: 11/22/2022]
Abstract
Xanthine oxidase (EC 1.17.3.2) (XO) is one of the main enzymatic sources that create reactive oxygen species (ROS) in the living system. It is a dehydrogenase enzyme that performs electron transfer to nicotinamide adenine dinucleotide (NAD+), while oxidizing hypoxanthin, which is an intermediate compound in purine catabolism, first to xanthine and then to uric acid. XO turns into an oxidant enzyme that oxidizes thiol groups under certain stress conditions in the tissue. The last metabolic step, in which hypoxanthin turns into uric acid, is catalyzed by XO. Uric acid, considered a waste product, can cause kidney stones and gouty-type arthritis as it is crystallized, when present in high concentrations. Thus, XO inhibitors are one of the drug classes used against gout, a purine metabolism disease that causes urate crystal storage in the joint and its surroundings caused by hyperuricemia. Urate-lowering therapy includes XO inhibitors that reduce uric acid production as well as uricosuric drugs that increase urea excretion. Current drugs that obstruct uric acid synthesis through XO inhibition are allopurinol, febuxostat, and uricase. However, since the side effects, safety and tolerability problems of some current gout medications still exist, intensive research is ongoing to look for new, effective, and safer XO inhibitors of natural or synthetic origins for the treatment of the disease. In the present review, we aimed to assess in detail XO inhibitory capacities of pure natural compounds along with the extracts from plants and other natural sources via screening Pubmed, Web of Science (WoS), Scopus, and Google Academic. The data pointed out to the fact that natural products, particularly phenolics such as flavonoids (quercetin, apigenin, and scutellarein), tannins (agrimoniin and ellagitannin), chalcones (melanoxethin), triterpenes (ginsenoside Rd and ursolic acid), stilbenes (resveratrol and piceatannol), alkaloids (berberin and palmatin) have a great potential for new XO inhibitors capable of use against gout disease. In addition, not only plants but other biological sources such as microfungi, macrofungi, lichens, insects (silk worms, ants, etc) seem to be the promising sources of novel XO inhibitors.
Collapse
Affiliation(s)
- Ilkay E Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Turkey
| | - Fatma S S Deniz
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330 Ankara, Turkey
| |
Collapse
|
22
|
Synthesis and biological evaluation of 2-(4-alkoxy-3-cyano)phenylpyrimidine derivatives with 4-amino or 4-hydroxy as a pharmacophore element binding with xanthine oxidase active site. Bioorg Med Chem 2021; 38:116117. [PMID: 33838610 DOI: 10.1016/j.bmc.2021.116117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/26/2021] [Accepted: 03/17/2021] [Indexed: 12/20/2022]
Abstract
Xanthine oxidase is the rate-limiting enzyme critical for the synthesis of uric acid, and therefore xanthine oxidase inhibitors are considered as one of the promising therapies for hyperuricemia and gout. In our previous study, series of 2-(4-alkoxy-3-cyano)phenyl-6-oxo-1,6-dihydropyrimidine-5-carboxylic acids and 2-(4-alkoxy-3-cyano)phenyl-6-imino-1,6-dihydropyrimidine-5-carboxylic acids were synthesized that presented excellent in vitro xanthine oxidase inhibitory potency. Interestingly, molecular docking studies revealed that the interaction behavior of these compounds with xanthine oxidase was changed after the conversion from a hydroxy group to amine group. To further investigate the structure-activity relationships of these pyrimidine-containing xanthine oxidase inhibitors and explore the contribution of amino or hydroxy group on xanthine oxidase inhibitory potency, several 2-phenylpyrimidine derivatives with amino or hydroxy functional group were designed and synthesized. Thereafter, the structure-activity research and molecular modeling study proved that hydroxy and amino groups could be used as pharmacophore elements for the design of 2-phenylpyrimidines xanthine oxidase inhibitors. Particularly, the optimized compound, 2-(3-cyano-4-isopentoxy)phenylpyrimidine-4-ol, emerged the strongest xanthine oxidase inhibitor potency, with an IC50 value of 0.046 µM, which was approximately 120-fold more potent than that of allopurinol (IC50 = 5.462 µM). Additionally, Lineweaver-Burk plot analysis revealed that the optimized compound acted as a mixed-type inhibitor. Furthermore, the in vivo hypouricemic effect of the optimized compound was investigated in a hyperuricemia rat model induced by potassium oxonate, and the results showed that the optimized compound could effectively reduce serum uric acid levels at an oral dose of 30 mg/kg.
Collapse
|
23
|
Wu Y, He H, Hou T. Purification, identification, and computational analysis of xanthine oxidase inhibitory peptides from kidney bean. J Food Sci 2021; 86:1081-1088. [PMID: 33565626 DOI: 10.1111/1750-3841.15603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/11/2020] [Accepted: 12/23/2020] [Indexed: 01/11/2023]
Abstract
Hyperuricemia is related to plenty of diseases, seriously damaging human health. Current clinical drugs used to treat hyperuricemia have many adverse effects. In this study, kidney bean hydrolysate (KBH) was found to exert high xanthine oxidase inhibitory (XOI) activity. Compared to KBH (50.31 ± 2.73%), XOI activities of three fractions (Mw <5 kDa, Mw <3 kDa, Mw < 1 kDa) by ultrafiltration were higher and increased to 58.58 ± 3.57%, 59.34 ± 1.78%, and 55.05 ± 5.00%, respectively (P < 0.05). A total of 69 peptides were identified by HPLC-ESI-MS/MS and analyzed binding affinities with XO with the help of molecular docking. AVDSLVPIGR, DWYDIK, LDNLLR, ISPIPVLK, ISSLEMTR showed well binding affinities with XO and DWYDIK presented the highest XOI activity (68.63 ± 5.07%) among five synthetic peptides (P < 0.05). Additionally, visual analysis results indicated that DWYDIK was pushed into the hydrophobic channel and formed hydrogen bonds with pivotal amino acids of xanthine oxidase. Overall, KBH could be a promising candidate as anti- hyperuricemia functional food. PRACTICAL APPLICATION: This research initially revealed that kidney bean peptides could significantly inhibit the activity of xanthine oxidase, indicating kidney bean peptides could be a treatment for hyperuricemia. Kidney bean peptides may have commercial potentials as a safer alternative with few side effects to drugs.
Collapse
Affiliation(s)
- Yuqun Wu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, 43000, China
| | - Hui He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, 43000, China
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.,Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan, 43000, China
| |
Collapse
|
24
|
Zhao J, Huang L, Sun C, Zhao D, Tang H. Studies on the structure-activity relationship and interaction mechanism of flavonoids and xanthine oxidase through enzyme kinetics, spectroscopy methods and molecular simulations. Food Chem 2020; 323:126807. [PMID: 32330646 DOI: 10.1016/j.foodchem.2020.126807] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/14/2022]
Abstract
In this study, some flavonoids were screened as potent xanthine oxidase (XO) inhibitors in vitro. Flavonoid 9 was demonstrated to exhibit the inhibitory activity through a ping-pong mechanism. Further structure-activity relationship revealed that different structural elements had greatly influenced the inhibition effect on XO and underlined the requirement of hydroxyl groups at C5 and C4' of flavonoid type I. Moreover, some bioactive flavonoids could efficiently quench the intrinsic fluorescence of XO by either static or static-dynamic mixed mechanism. The synchronous fluorescence, ANS-binding fluorescence, Fourier transform infrared spectra and circular dichroism suggested that active flavonoids could bind to the active center of XO, prevent the entrance of substrate, and induce the rearrangement and conformation change of its secondary structures, ultimately resulting in the significant inhibition effect. Additionally, molecular docking further confirmed these conclusions and highlighted the great importance of hydrophobic interactions and hydrogen bonds for the formation of stable complex conformation.
Collapse
Affiliation(s)
- Jie Zhao
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China
| | - Lin Huang
- Blood Purification Center, Affiliated Yijishan Hospital of Wannan Medical College, Wuhu 241001, PR China
| | - Chunyong Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Dongsheng Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Hongjin Tang
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu 241000, PR China.
| |
Collapse
|
25
|
Gao J, Liu X, Zhang B, Mao Q, Zhang Z, Zou Q, Dai X, Wang S. Design, synthesis and biological evaluation of 1-alkyl-5/6-(5-oxo-4,5-dihydro-1,2,4-oxadiazol-3-yl)-1H-indole-3-carbonitriles as novel xanthine oxidase inhibitors. Eur J Med Chem 2020; 190:112077. [DOI: 10.1016/j.ejmech.2020.112077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/11/2020] [Accepted: 01/16/2020] [Indexed: 12/17/2022]
|
26
|
From Xanthine Oxidase Inhibition to In Vivo Hypouricemic Effect: An Integrated Overview of In Vitro and In Vivo Studies with Focus on Natural Molecules and Analogues. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9531725. [PMID: 32184901 PMCID: PMC7060854 DOI: 10.1155/2020/9531725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/10/2019] [Accepted: 12/24/2019] [Indexed: 01/05/2023]
Abstract
Hyperuricemia is characterized by elevated uric acid (UA) levels on blood, which can lead to gout, a common pathology. These high UA levels are associated with increased purine ingestion and metabolization and/or its decreased excretion. In this field, xanthine oxidase (XO), by converting hypoxanthine and xanthine to UA, plays an important role in hyperuricemia control. Based on limitations and adverse effects associated with the use of allopurinol and febuxostat, the most known approved drugs with XO inhibitory effect, the search for new molecules with XO activity is growing. However, despite the high number of studies, it was found that the majority of tested products with relevant XO inhibition were left out, and no further pharmacological evaluation was performed. Thus, in the present review, available information published in the past six years concerning isolated molecules with in vitro XO inhibition complemented with cytotoxicity evaluation as well as other relevant studies, including in vivo hypouricemic effect, and pharmacokinetic/pharmacodynamic profile was compiled. Interestingly, the analysis of data collected demonstrated that molecules from natural sources or their mimetics and semisynthetic derivatives constitute the majority of compounds being explored at the moment by means of in vitro and in vivo animal studies. Therefore, several of these molecules can be useful as lead compounds and some of them can even have the potential to be considered in the future clinical candidates for the treatment of hyperuricemia.
Collapse
|
27
|
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
|
28
|
Novel 3-[4-alkoxy-3-(1H-tetrazol-1-yl) phenyl]-1,2,4-oxadiazol-5(4H)-ones as promising xanthine oxidase inhibitors: Design, synthesis and biological evaluation. Bioorg Chem 2020; 95:103564. [DOI: 10.1016/j.bioorg.2019.103564] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/27/2019] [Accepted: 12/30/2019] [Indexed: 12/23/2022]
|
29
|
Zhang TJ, Zhang Y, Tu S, Wu YH, Zhang ZH, Meng FH. Design, synthesis and biological evaluation of N-(3-(1H-tetrazol-1-yl)phenyl)isonicotinamide derivatives as novel xanthine oxidase inhibitors. Eur J Med Chem 2019; 183:111717. [PMID: 31557611 DOI: 10.1016/j.ejmech.2019.111717] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/06/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022]
Abstract
In our previous study, we reported a series of N-phenylisonicotinamide derivatives as novel xanthine oxidase (XO) inhibitors and identified N-(3-cyano-4-((2-cyanobenzyl)oxy)phenyl)isonicotinamide (compound 1) as the most potent one with an IC50 value of 0.312 μM. To further optimize the structure and improve the potency, a structure-based drug design (SBDD) strategy was performed to construct the missing H-bond between the small molecule and the Asn768 residue of XO. We introduced a tetrazole moiety at the 3'-position of the phenyl to serve as an H-bond acceptor and obtained a series of N-(3-(1H-tetrazol-1-yl)phenyl)isonicotinamide derivatives (2a-t and 6-8). Besides, to investigate the influence of the amide-reversal, some N-(pyridin-4-yl)-3-(1H-tetrazol-1-yl)benzamide derivatives (3c, 3e, 3i, 3k and 3u) were also synthesized and evaluated. Biological evaluation and structure-activity relationship analysis demonstrated that the 3'-(1H-tetrazol-1-yl) moiety was an excellent fragment for the N-phenylisonicotinamide scaffold; a substituted benzyloxy, especially, an m-cyanobenzyloxy (e.g., 2s), linking at the 4'-position was welcome for the potency; and the amide-reversal could damage the potency, so maintenance of the N-phenylisonicotinamide scaffold was essential. In summary, starting from compound 1, the SBDD effort successfully identified a promising XO inhibitor 2s (IC50 = 0.031 μM), with a 10-fold gain in potency. Its potency was very close to the positive control topiroxostat (IC50 = 0.021 μM). A Lineweaver-Burk plot indicated that compound 2s acted as a mixed-type XO inhibitor. Molecular docking and molecular dynamics simulations revealed that the tetrazole moiety could occupy the Asn768-sub-pocket with N-4 atom accepting an H-bond from the Asn768 residue, as expected.
Collapse
Affiliation(s)
- Ting-Jian Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang, 110122, China
| | - Yi Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang, 110122, China
| | - Shun Tu
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang, 110122, China
| | - Yu-Hang Wu
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang, 110122, China
| | - Zhen-Hao Zhang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang, 110122, China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang, 110122, China.
| |
Collapse
|
30
|
Hendrickson C, Linden K, Kreyer S, Beilman G, Scaravilli V, Wendorff D, Necsoiu C, Batchinsky AI, Cancio LC, Chung KK, Lusczek ER. 1H-NMR Metabolomics Identifies Significant Changes in Metabolism over Time in a Porcine Model of Severe Burn and Smoke Inhalation. Metabolites 2019; 9:E142. [PMID: 31336875 PMCID: PMC6680385 DOI: 10.3390/metabo9070142] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
Burn injury initiates a hypermetabolic response leading to muscle catabolism and organ dysfunction but has not been well-characterized by high-throughput metabolomics. We examined changes in metabolism over the first 72 h post-burn using proton nuclear magnetic resonance (1H-NMR) spectroscopy and serum from a porcine model of severe burn injury. We sought to quantify the changes in metabolism that occur over time in response to severe burn and smoke inhalation in this preliminary study. Fifteen pigs received 40% total body surface area (TBSA) burns with additional pine bark smoke inhalation. Arterial blood was drawn at baseline (pre-burn) and every 24 h until 72 h post-injury or death. The aqueous portion of each serum sample was analyzed using 1H-NMR spectroscopy and metabolite concentrations were used for principal component analysis (PCA). Thirty-eight metabolites were quantified in 39 samples. Of these, 31 showed significant concentration changes over time (p < 0.05). PCA revealed clustering of samples by time point on a 2D scores plot. The first 48 h post-burn were characterized by high concentrations of histamine, alanine, phenylalanine, and tyrosine. Later timepoints were characterized by rising concentrations of 2-hydroxybutyrate, 3-hydroxybutyrate, acetoacetate, and isovalerate. No significant differences in metabolism related to mortality were observed. Our work highlights the accumulation of organic acids resulting from fatty acid catabolism and oxidative stress. Further studies will be required to relate accumulation of the four organic carboxylates identified in this analysis to outcomes from burn injury.
Collapse
Affiliation(s)
- Cole Hendrickson
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katharina Linden
- US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, USA
- Department for Pediatric Cardiology, University Hospital Bonn, 53113 Bonn, Germany
| | - Stefan Kreyer
- US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, USA
- Clinic for Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127 Bonn, Germany
| | - Gregory Beilman
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vittorio Scaravilli
- US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, USA
- Department of Anesthesia Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Daniel Wendorff
- US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, USA
| | - Corina Necsoiu
- US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, USA
| | | | - Leopoldo C Cancio
- US Army Institute of Surgical Research, Fort Sam Houston, TX 78234, USA
| | - Kevin K Chung
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | |
Collapse
|
31
|
Tang H, Zhao D. Studies of febuxostat analogues as xanthine oxidase inhibitors through 3D-QSAR, Topomer CoMFA and molecular modeling. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2019. [DOI: 10.1007/s13738-019-01726-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
Luna G, Dolzhenko AV, Mancera RL. Inhibitors of Xanthine Oxidase: Scaffold Diversity and Structure-Based Drug Design. ChemMedChem 2019; 14:714-743. [PMID: 30740924 DOI: 10.1002/cmdc.201900034] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Indexed: 12/19/2022]
Abstract
Xanthine oxidase (XO) is the enzyme responsible for the catabolism of purines and their conversion into uric acid. XO is thus the target for the treatment of hyperuricemia and gout. For more than 50 years the only XO inhibitor drug available on the market was the purine analogue allopurinol. In the last decade there has been a resurgence in the search for new inhibitors of XO, as the activity of XO and hyperuricemia have also been associated with a variety of conditions such as diabetes, hypertension, and other cardiovascular diseases. In recent years the non-purine inhibitor febuxostat was approved in Europe and the USA for the treatment of hyperuricemia. This drug was followed by another XO inhibitor called topiroxostat. This review discusses the molecular structures and activities of the multiple classes of inhibitors that have been developed since the discovery of allopurinol, with a brief review of the molecular interactions between inhibitors and XO active site residues for the most important molecules. The challenges ahead for the discovery of new inhibitors of XO with novel chemical structures are discussed.
Collapse
Affiliation(s)
- Giuseppe Luna
- School of Pharmacy and Biomedical Sciences and Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA, 6845, Australia
| | - Anton V Dolzhenko
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia
| | - Ricardo L Mancera
- School of Pharmacy and Biomedical Sciences and Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA, 6845, Australia
| |
Collapse
|