1
|
Morais Costa NE, Dos Santos PHC, Silva Medeiros VG, Guimarães AS, Caldas Santos JC, Lins Freire NM, da Silva JCS, de Aquino TM, Modolo LV, Alberto EE, de Fátima Â. Synthesis and anti-ureolitic activity of Biginelli adducts derived from formylphenyl boronic acids. Bioorg Chem 2024; 152:107735. [PMID: 39213798 DOI: 10.1016/j.bioorg.2024.107735] [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: 07/02/2024] [Revised: 08/10/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Urease is a metalloenzyme that contains two Ni(II) ions in its active site and catalyzes the hydrolysis of urea into ammonia and carbon dioxide. The development of effective urease inhibitors is crucial not only for mitigating nitrogen losses in agriculture but also for offering an alternative treatment against infections caused by resistant pathogens that utilize urease as a virulence factor. This study focuses on synthesizing and investigating the urease inhibition potential of Biginelli Adducts bearing a boric acid group. An unsubstituted or hydroxy-substituted boronic group in the Biginelli adducts structure enhances the urease inhibitory activity. Biophysical and kinetics studies revealed that the best Biginelli adduct (4e; IC50 = 132 ± 12 µmol/L) is a mixed inhibitor with higher affinity to the urease active site over an allosteric one. Docking studies confirm the interactions of 4e with residues essential for urease activity and demonstrate its potential to coordinate with the nickel atoms through the oxygen atoms of carbonyl or boronic acid groups. Overall, the Biginelli adduct 4e shows great potential as an additive for developing enhanced efficiency fertilizers and/or for medical applications.
Collapse
Affiliation(s)
- Nathália Evelyn Morais Costa
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departamento de Química, ICEx, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Pedro Henrique Costa Dos Santos
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departamento de Química, ICEx, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Victorya Gabryelle Silva Medeiros
- Laboratório de Instrumentação e Desenvolvimento em Química Analítica, LINQA, Instituto de Químico e Biotecnologia, 57072-900 Maceió, AL, Brazil
| | - Ari Souza Guimarães
- Laboratório de Instrumentação e Desenvolvimento em Química Analítica, LINQA, Instituto de Químico e Biotecnologia, 57072-900 Maceió, AL, Brazil
| | - Josué Carinhanha Caldas Santos
- Laboratório de Instrumentação e Desenvolvimento em Química Analítica, LINQA, Instituto de Químico e Biotecnologia, 57072-900 Maceió, AL, Brazil
| | - Nathalia Monteiro Lins Freire
- Research Group on Therapeutic Strategies - GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, Brazil
| | | | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies - GPET, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, AL, Brazil
| | - Luzia V Modolo
- Departamento de Botânica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Eduardo E Alberto
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departamento de Química, ICEx, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Ângelo de Fátima
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departamento de Química, ICEx, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil.
| |
Collapse
|
2
|
Li Y, Zou H, Sun-Waterhouse D, Chen Y. Chlorogenic acid, caffeic acid and luteolin from dandelion as urease inhibitors: insights into the molecular interactions and inhibition mechanism. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:8079-8088. [PMID: 38877786 DOI: 10.1002/jsfa.13637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/02/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Dandelion contains hundreds of active compounds capable of inhibiting urease activity, but the individual compounds have not yet been fully identified, and their effects and underlying mechanisms are not clear. The present study aimed to screen the urease inhibition active compounds of dandelion by urease inhibitory activity evaluation HPLC-tandem mass spectrometry analysis, their mechanism of urease inhibition by polyphenols was explored using enzyme kinetic studies via Lineweaver-Burk plots. Other investigations included isothermal titration calorimetry and surface plasmon resonance sensing, fluorescence quenching experiments, and single ligand molecular docking and two-ligand simultaneous docking techniques. RESULTS The results indicated that the ethyl acetate fraction of dandelion flower exhibited the greatest inhibition (lowest IC50 0.184 ± 0.007 mg mL-1). Chlorogenic acid, caffeic acid and luteolin could be effective urease inhibitors that acted in a non-competitive inhibition manner. Individually, chlorogenic acid could not only fast bind to urease, but also dissociate rapidly, whereas luteolin might interact with urease with the weakest affinity. The chlorogenic acid-caffeic acid combination exhibited an additive effect in urease inhibition. However, the chlorogenic acid-luteolin and caffeic acid-luteolin combinations exhibited antagonistic effects, with the caffeic acid-luteolin combination showing greater antagonism. CONCLUSION The present study reveals that chlorogenic acid, caffeic acid and luteolin are major bioactive compounds for urease inhibition, indicating the molecular mechanisms. The antagonistic effects were observed between luteolin and chlorogenic acid/caffeic acid, and the interactions of the catalytic site and flap may account for the antagonistic effects. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yanni Li
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Healthy in Universities of Shandong, Shandong Agricultural University, Taian, China
| | - Hui Zou
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Healthy in Universities of Shandong, Shandong Agricultural University, Taian, China
| | - Dongxiao Sun-Waterhouse
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Healthy in Universities of Shandong, Shandong Agricultural University, Taian, China
| | - Yilun Chen
- College of Food Science and Engineering, Key Laboratory of Food Nutrition and Healthy in Universities of Shandong, Shandong Agricultural University, Taian, China
| |
Collapse
|
3
|
Khair-ul-Bariyah S, Sarfraz M, Arshad M, Waseem A, Khan HU, Khan S, Sharif A, Farooqi ZH, Ahmed E. Synthesis of 2-aminothiazole sulfonamides as potent biological agents: Synthesis, structural investigations and docking studies. Heliyon 2024; 10:e34980. [PMID: 39157352 PMCID: PMC11327602 DOI: 10.1016/j.heliyon.2024.e34980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/20/2024] Open
Abstract
A simplified synthetic approach involving sulfonylation followed by amino group alkylation produced new 2-aminothiazole derivatives. UV/Vis, infrared, and NMR spectroscopies confirmed their structures. Compounds 36, 22, 34, and 35 showed strong inhibition against Jack bean and Bacillus Pasteurii urease, with IC50 values from 14.06 to 20.21 μM/mL. Compounds 20, 26, 21, 29, 30, 31, and 32 exhibited potent inhibitory effects against α-glucosidase and α-amylase, with IC50 values between 20.34 and 37.20 μM/mL. Compounds 33, 26, and 27 demonstrated potent DPPH scavenging, with IC50 values around 34.4-39.2 μM/mL. FMO analysis showed compounds 21, 22, 24, and 25 having parallel aromatic ring systems due to π cloud interactions, while compounds 32 and 38 had distinct electronic density distributions. Compound 22 had HOMO and LUMO energy gaps of 5.805 eV, with bromo and fluoro substitutions in compounds 21 and 24 slightly increasing the gaps to 6.089 eV and 6.078 eV, respectively. Nitro groups in compounds 25 and 32 reduced the gaps to 0.384 eV and 1.187 eV. All compounds demonstrated high gastrointestinal absorption, non-permeability to the blood-brain barrier, and optimal skin permeation (Log Kp between -5.83 and -6.54 cm/s). Compounds 22, 24, and 38 had promising QED scores of 0.719, 0.707, and 0.860, respectively, with synthetic accessibility scores from 2.057 to 2.517. ADMET predictions indicated minimal toxicity, cardiovascular safety, and significant inhibitory potential for CYP enzymes. Strong in silico binding affinities (binding energies -5.75 to -7.63 kcal/mol) and metabolic stability suggest these derivatives are promising candidates for further drug development.
Collapse
Affiliation(s)
| | - Muhammad Sarfraz
- Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Muhammad Arshad
- Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Amir Waseem
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hidayat Ullah Khan
- Department of Chemistry, University of Science and Technology, Bannu, 28100, Pakistan
| | - Shahnaz Khan
- Department of Chemistry, University of Science and Technology, Bannu, 28100, Pakistan
| | - Ahsan Sharif
- School of Chemistry, University of the Punjab, 54590, Pakistan
| | | | - Ejaz Ahmed
- School of Chemistry, University of the Punjab, 54590, Pakistan
| |
Collapse
|
4
|
Viana LPS, Naves GM, Medeiros IG, Guimarães AS, Sousa ES, Santos JCC, Freire NML, de Aquino TM, Modolo LV, de Fátima Â, da Silva CM. Synergizing structure and function: Cinnamoyl hydroxamic acids as potent urease inhibitors. Bioorg Chem 2024; 146:107247. [PMID: 38493635 DOI: 10.1016/j.bioorg.2024.107247] [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: 12/27/2023] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/19/2024]
Abstract
The current investigation encompasses the structural planning, synthesis, and evaluation of the urease inhibitory activity of a series of molecular hybrids of hydroxamic acids and Michael acceptors, delineated from the structure of cinnamic acids. The synthesized compounds exhibited potent urease inhibitory effects, with IC50 values ranging from 3.8 to 12.8 µM. Kinetic experiments unveiled that the majority of the synthesized hybrids display characteristics of mixed inhibitors. Generally, derivatives containing electron-withdrawing groups on the aromatic ring demonstrate heightened activity, indicating that the increased electrophilicity of the beta carbon in the Michael Acceptor moiety positively influences the antiureolytic properties of this compounds class. Biophysical and theoretical investigations further corroborated the findings obtained from kinetic assays. These studies suggest that the hydroxamic acid core interacts with the urease active site, while the Michael acceptor moiety binds to one or more allosteric sites adjacent to the active site.
Collapse
Affiliation(s)
- Luciana P S Viana
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Giovanna M Naves
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isabela G Medeiros
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ari S Guimarães
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Emilly S Sousa
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Josué C C Santos
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Nathália M L Freire
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Thiago M de Aquino
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Luzia V Modolo
- Departamento de Botânica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ângelo de Fátima
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cleiton M da Silva
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
5
|
Lansdorp BM, Lamberg P, Hamid R. Screen-Printed Silver/Silver Chloride Electrodes Inhibit Alcohol Oxidase Activity. ECS SENSORS PLUS 2023; 2:030602. [PMID: 37469623 PMCID: PMC10353475 DOI: 10.1149/2754-2726/ace5a9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Indexed: 07/21/2023]
Abstract
Silver/silver chloride (Ag/AgCl) is ubiquitous in the field of electrochemical biosensing due to its suitability as a reference electrode material. However, we recently discovered that screen-printed Ag/AgCl ink has a detrimental effect on Alcohol Oxidase enzyme stability. We performed an optical absorbance assay to isolate the interaction of enzyme and electrode to discover a surprisingly strong inhibition effect. The halftime of enzymatic activity was reduced from nearly 1 week in buffer to 10 h in the presence of the Ag/AgCl electrode. We expect this discovery to have broad implications on enzymatic biosensors that use Ag/AgCl as reference electrode material.
Collapse
Affiliation(s)
- Bob M. Lansdorp
- Milo Sensors, Inc. Santa Barbara, CA, 93101, United States of America
| | - Peter Lamberg
- Milo Sensors, Inc. Santa Barbara, CA, 93101, United States of America
| | - Rashad Hamid
- Milo Sensors, Inc. Santa Barbara, CA, 93101, United States of America
| |
Collapse
|
6
|
Jiang J, Liang P, Li A, Xue Q, Yu H, You Z. Synthesis, Crystal Structures and Urease Inhibition of Zinc(II) and Copper(II) Complexes Derived from 2-Amino-N′-(1-(Pyridin-2-yl) Ethylidene)Benzohydrazide. J STRUCT CHEM+ 2023. [DOI: 10.1134/s0022476623030034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
7
|
Cephalosporin as Potent Urease and Tyrosinase Inhibitor: Exploration through Enzyme Inhibition, Kinetic Mechanism, and Molecular Docking Studies. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1092761. [PMID: 35937399 PMCID: PMC9352478 DOI: 10.1155/2022/1092761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022]
Abstract
In present study, eleven cephalosporin drugs were selected to explore their new medically important enzyme targets with inherited safety advantage. To this end, selected drugs with active ingredient, cefpodoxime proxetil, ceftazidime, cefepime, ceftriaxone sodium, cefaclor, cefotaxime sodium, cefixime trihydrate, cephalexin, cefadroxil, cephradine, and cefuroxime, were evaluated and found to have significant activity against urease (IC50 = 0.06 ± 0.004 to 0.37 ± 0.046 mM) and tyrosinase (IC50 = 0.01 ± 0.0005 to 0.12 ± 0.017 mM) enzymes. Urease activity was lower than standard thiourea; however, tyrosinase activity of all drugs outperforms (ranging 6 to 18 times) the positive control: hydroquinone (IC50 = 0.18 ± 0.02 mM). Moreover, the kinetic analysis of the most active drugs, ceftriaxone sodium and cefotaxime sodium, revealed that they bind irreversibly with both the enzymes; however, their mode of action was competitive for urease and mixed-type, preferentially competitive for tyrosinase enzyme. Like in vitro activity, ceftriaxone sodium and cefotaxime sodium docking analysis showed their considerable binding affinity and significant interactions with both urease and tyrosinase enzymes sufficient for downstream signaling responsible for observed enzyme inhibition in vitro, purposing them as potent candidates to control enzyme-rooted obstructions in future.
Collapse
|
8
|
He Y, Zhang X, Li M, Zheng N, Zhao S, Wang J. Coptisine: A natural plant inhibitor of ruminal bacterial urease screened by molecular docking. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 808:151946. [PMID: 34843773 DOI: 10.1016/j.scitotenv.2021.151946] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/20/2021] [Accepted: 11/20/2021] [Indexed: 06/13/2023]
Abstract
Inhibition of ruminal bacterial urease activity could slow down the decomposition of urea to ammonia, which would lead to a decrease in urea synthesis in the liver and urea-N emission in the urine. In order to find a rumen bacterial urease specific inhibitor that is environmentally friendly, we used the homology model of rumen bacterial urease as the target to screen natural compounds from plants by molecular docking. The screening results showed that coptisine had the most potential to inhibit the activity of rumen bacterial urease with an IC50 of 2.45 μM, which was superior to the traditional inhibitor acetohydroxamic acid. The enzyme kinetics results indicated coptisine was mixed type inhibitor of rumen bacterial urease with a Ki value of 0.68 μM. Coptisine significantly decreased the release of NH3 and decomposition of urea and improved microbial fermentation in a rumen fermentation system in vitro. Thiol-containing compounds or boric acid significantly decreased the inhibitory capacity of coptisine toward rumen bacterial urease, which indicated that coptisine could interact with both the urease active center Ni and amino acid residues possessing sulfhydryl groups in the flap area. The molecular docking results showed that coptisine acted as the metal acceptor for one nickel ion in the active site, and formed hydrogen bonds with the amino acid residues His320 and His362, which were located in the active site and flap region, respectively. These findings emphasized the potential role of coptisine in reducing nitrogen emissions that originate from ruminants by regulating rumen bacterial urease activity.
Collapse
Affiliation(s)
- Yue He
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaoyin Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ming Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
9
|
Ji J, Wang S, Zhao J, Yang T, Wang J, You Z. Synthesis, crystal structures and urease inhibition of copper, nickel and zinc complexes derived from 4-chloro-2-((pyridin-2-ylmethylene)amino)phenol. J COORD CHEM 2022. [DOI: 10.1080/00958972.2022.2032005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jing Ji
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| | - Shiyi Wang
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| | - Jie Zhao
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| | - Ting Yang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, P.R. China
| | - Jiaqi Wang
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| | - Zhonglu You
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| |
Collapse
|
10
|
Lu Q, Tan D, Xu Y, Liu M, He Y, Li C. Inactivation of Jack Bean Urease by Nitidine Chloride from Zanthoxylum nitidum: Elucidation of Inhibitory Efficacy, Kinetics and Mechanism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:13772-13779. [PMID: 34767340 DOI: 10.1021/acs.jafc.1c04801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Urease is a metalloenzyme that catalyzes the hydrolysis of urea into ammonia and carbon dioxide, which has a negative impact on human health and agriculture. In this study, the inactivation of jack bean urease by nitidine chloride (NC) was investigated to elucidate the inhibitory effect, kinetics, and underlying mechanism of action. The results showed that NC acted as a concentration- and time-dependent inhibitor with an IC50 value of 33.2 ± 4.8 μM and exhibited a similar inhibitory effect to acetohydroxamic acid (IC50 = 31.7 ± 5.8 μM). Further kinetic analysis demonstrated that NC was a slow-binding and non-competitive inhibitor for urease. Thiol-blocking reagents (dithiothreitol, glutathione, and l-cysteine) significantly retarded urease inactivation, while Ni2+ competitive inhibitors (boric acid and sodium fluoride) synergetically suppressed urease with NC, suggesting that the active site sulfhydryl groups were possibly obligatory for NC blocking urease. Molecular docking simulation further argued its inhibition mechanism. Additionally, NC-induced deactivation of urease was verified to be reversible since the inactivated enzyme could be reactivated by glutathione. Taking together, NC was a non-competitive inhibitor targeting the thiol group at the active site of urease with characteristics of concentration dependence, reversibility, and slow binding, serving as a promising novel urease suppressant.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, PR China
| | - Daopeng Tan
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, PR China
| | - Yifei Xu
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518005, PR China
| | - Meigui Liu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, PR China
| | - Yuqi He
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, PR China
| | - Cailan Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai 519041, PR China
| |
Collapse
|
11
|
Chen K, Wu W, Hou X, Yang Q, Li Z. A review: antimicrobial properties of several medicinal plants widely used in Traditional Chinese Medicine. FOOD QUALITY AND SAFETY 2021. [DOI: 10.1093/fqsafe/fyab020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Abstract
Due to the dramatic increase in the use of antibiotics and growing health threat of bacterial resistance to many commonly used antibiotics, many studies have been directed at developing new and effective antibacterial compounds, among which many new, natural, and effective antibacterial compounds discovered from medicinal plants have drawn great interest and raised new hope for treating the challenges of antibiotic resistance. This review aimed to summarize the most important and widely used medicinal plants that were reported to have antibacterial activities. A general literature search from 2010 to 2020 was conducted using different databases, including Science Direct, Web of Science, and PubMed. According to the literature, three medicinal plants with outstanding antibacterial activities, Taraxacum officinale, Coptis Rhizome, and Scutellaria baicalensis, were screened and reviewed by prioritization. The extraction methods, antibacterial activities of different parts of plants or the plant-derived compounds, spectra of antibacterial activities, and toxicity were described, respectively. However, the antibacterial activities of the extracts or pure compounds as reported in the reviewed literature were mostly based on in vitro assays, and moreover, the deeper antibacterial mechanisms have not been elucidated clearly. Therefore, further studies are required in the fields of purification and identification of the antibacterial compounds, its mechanisms of action, and synergistic effects in combination with other antibacterial drugs, which may be helpful in the development of new antibacterial drugs.
Collapse
|
12
|
Tavares MC, Oliveira KA, de Fátima Â, Coltro WKT, Santos JCC. Paper-based analytical device with colorimetric detection for urease activity determination in soils and evaluation of potential inhibitors. Talanta 2021; 230:122301. [PMID: 33934769 DOI: 10.1016/j.talanta.2021.122301] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/24/2021] [Accepted: 03/06/2021] [Indexed: 10/21/2022]
Abstract
Urease is an enzyme associated with the degradation of urea, an important nitrogen fertilizer in agriculture. Thus, this current report describes the use of a paper-based analytical device (UrePAD) designed to contain a microzone array for colorimetric determination of urease activity in soils in the absence/presence of potential enzyme inhibitors. The UrePAD can be used at the point-of-need (point-of-care), and it offers advantages such as low cost, simplicity in handling, low sample/reagent volumes, and no use of toxic reagents. The acid-base indicator phenol red was used to monitor the urea hydrolysis reaction catalyzed by urease in the evaluated systems. The images were digitalized in a bench scanner, and the analysis was performed using Corel Draw X8 software. The device offered a LOD of 0.10 U mL-1 with linearity between 0.25 and 4.0 U mL-1 and a relative standard deviation ≤ 1.38%. UrePAD was tested in four soil samples of different characteristics and with eight urease inhibitors of varied classes. The results obtained through the proposed device did not differ statistically (95% confidence interval) from those employing the classic method based on the Berthelot reaction, thus indicating that UrePAD was effective for determining urease activity and screening inhibitors, besides showing the capacity to simplify fieldwork involving the application of urea in the soil.
Collapse
Affiliation(s)
- Maria Célia Tavares
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Campus A.C. Simões, 57072-900, Maceió, Alagoas, Brazil
| | | | - Ângelo de Fátima
- Departmento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG 31270-901, Brazil
| | - Wendell K T Coltro
- Instituto de Química, Universidade Federal de Goiás, Goiânia, GO 74690-900, Brazil.
| | - Josué Carinhanha Caldas Santos
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Campus A.C. Simões, 57072-900, Maceió, Alagoas, Brazil.
| |
Collapse
|
13
|
Mamidala R, Bhimathati SRS, Vema A. Discovery of Novel Dihydropyrimidine and hydroxamic acid hybrids as potent Helicobacter pylori Urease inhibitors. Bioorg Chem 2021; 114:105010. [PMID: 34102519 DOI: 10.1016/j.bioorg.2021.105010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/25/2022]
Abstract
Two novel series of Dihydropyrimidine-hydroxamic acid hybrids (4a-4l and 5a-5l) were designed, synthesized and evaluated for in vitro Helicobacter pylori urease inhibition. In vitro enzyme inhibition screening led to the discovery of three potent urease inhibitors 2-[[4-(4-hydroxy phenyl)-6-oxo-1,6-dihydropyrimidine-2-yl]-amino]-N-hydroxy acetamide (4g), 2-[[4-(4-chloro phenyl)-6-oxo-1,6-dihydropyrimidine-2-yl]-amino]-N-hydroxy acetamide (4b) and 3-[[4-(3-methoxy phenyl)-6-oxo-1,6-dihydropyrimidine-2-yl]-amino]-N-hydroxy propanamide (5l). Compound 4g showed excellent urease inhibition with IC50 value of 14 ± 1 nM, indicated by its strong interactions with both metallic Ni++ ions, Gly279, His221, Ala365, Asp362, Asn168, Arg338 and His322 residues of the active site of urease. Further, compounds 4b and 5l displayed very good activity with IC50 value of 0.082 ± 0.004 µM and 0.14 ± 0.013 µM respectively compared to standard Acetohydroxamic acid (IC50 - 27.4 ± 1.2 µM). Kinetic studies revealed that a mixed inhibition with both competitive and non-competitive aspects is involved in the urease inhibition mechanism. The in vitro urease inhibition results were supported by molecular docking studies. Collectively, this study indicates that 4g could be considered as promising lead molecule that can be further developed as a potent drug molecule for the treatment of Helicobacter pylori caused gastritis for further studies.
Collapse
Affiliation(s)
- Ravinder Mamidala
- Department of Medicinal Chemistry, St. Peter's Institute of Pharmaceutical Sciences, Hanamkonda, Warangal, Telangana 506001, India; Department of Pharmaceutical Chemistry, Institute of Science and Technology, Jawaharlal Nehru Technological University, Hyderabad, Telangana 500085, India
| | - Solomon Raj S Bhimathati
- Department of Pharmacology, Gland Institute of Pharmaceutical Sciences, Medak, Telangana 502220, India
| | - Aparna Vema
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India.
| |
Collapse
|
14
|
Khan MU, Aslam M, Shahzad SA, Khan ZA, Khan NA, Ali M, Naz S, Rahman J, Farooq U. Design and synthesis of thiobarbituric acid analogues as potent urease inhibitors. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.129959] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
A Novel Urease Inhibitor of Ruminal Microbiota Screened through Molecular Docking. Int J Mol Sci 2020; 21:ijms21176006. [PMID: 32825454 PMCID: PMC7503308 DOI: 10.3390/ijms21176006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/28/2020] [Accepted: 08/17/2020] [Indexed: 11/20/2022] Open
Abstract
Inhibition of the urease activity of ruminal microbiota is not only beneficial for increasing dietary and endogenic urea-N utilization efficiency in ruminants but also might be applicable for the preservation of nitrogen fertilizer in soil and treatment of gastrointestinal and urinary tract infections caused by ureolytic bacteria. To discover urease inhibitors to efficiently target ruminal microbiota, the identified ruminal microbial metagenomic urease gene was used to construct a homology model to virtually screen urease inhibitors from the ChemDiv database by molecular docking. The GMQE and QMEAN values of the homology model were 0.85 and −0.37, respectively, indicating a good model quality. The inhibition effect of the screened urease inhibitor for ruminal urea degradation was assessed by ruminal microbial fermentation in vitro. The toxic effect of the candidate inhibitor was performed using gut Caco-2 cells in vitro. The results showed that compound 3-[1-[(aminocarbonyl)amino]-5-(4-methoxyphenyl)-1H-pyrrol-2-yl] propanoic acid (ChemDiv_ID: 6238-0047, IC50 = 65.86 μM) was found to be the most effective urease inhibitor among the candidate compounds. Compound 6238-0047 significantly lowered the amount of urea degradation and ammonia production in ruminal microbial fermentation. The 24 h degradation rate of compound 6238-0047 in ruminal microbial fermentation was 3.32%–16.00%. In addition, compound 6238-0047 (10–100 μM) had no significant adverse effect on the cell viability of Caco-2 cells. Molecular docking showed that compound 6238-0047 could interact with Asp359 in the active site and Cys318 in the flap region by the hydrogen bond and Pi-Alkyl interaction, respectively. Compound 6238-0047 could be used as a novel inhibitor for decreasing the urease activity of ruminal microbiota.
Collapse
|
16
|
Jagannathan V, Venkatesan A, Viswanathan P. Kinetics and Computational Evaluation of Eugenol and Vanillic Acid on Inhibition of a Potential Enzyme of a Nosocomial Pathogen that Promotes Struvite Formation. CURRENT ENZYME INHIBITION 2020; 16:162-171. [DOI: 10.2174/1573408016999200415115754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/12/2020] [Accepted: 04/12/2020] [Indexed: 02/08/2023]
Abstract
Background:
Struvite/infection stone is one of the major clinical burdens in urinary tract
infections that is caused by the ureolytic behavior of pathogenic bacteria.
Objective:
The current strategy for treating infective stones is mostly antibiotic therapy, which ends
in promoting resistance to the organisms. Hence in the present study, we investigated two phytocompounds,
eugenol (an allyl-substituted guaiacol) and vanillic acid (a phenolic acid) that are found to be
effective in inhibiting the urease enzyme of a nosocomial pathogen Proteus mirabilis.
Methods:
The enzyme was purified to apparent homogeneity and the kinetic parameters were studied
in the presence and in the absence of eugenol and vanillic acid. Molecular docking and simulation
were done to understand the level of protein-ligand interactions and the interacting residues.
Results:
Kinetic parameters obtained from the Michaelis-Menten plot show that both eugenol and
vanillic acid exhibit non-competitive inhibition of urease enzyme in a dose-dependent manner. In silico
studies showed that eugenol and vanillic acid have almost similar binding affinities to the regulatory
pocket of the modeled protein. Dynamics and simulation results indicate that the interaction of
ligands with the ARG373 residue of the protein provides a stable bound conformation.
Conclusion:
Overall, our results suggest that both the phytocompounds eugenol and vanillic acid
have a potential application as a new therapy for the inhibition of urease enzyme that could possibly
replace the complexions related to struvite stone formation.
Collapse
Affiliation(s)
- Venkataseshan Jagannathan
- Renal Research Laboratory, Department of Biosciences, Centre for Biomedical Research, Vellore - 632 014, Tamil Nadu, India
| | - Arthi Venkatesan
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore - 632 014, Tamil Nadu, India
| | - Pragasam Viswanathan
- Renal Research Laboratory, Department of Biosciences, Centre for Biomedical Research, Vellore - 632 014, Tamil Nadu, India
| |
Collapse
|
17
|
Barakat A, Soliman SM, Ali M, Elmarghany A, Al-Majid AM, Yousuf S, Ul-Haq Z, Choudhary MI, El-Faham A. Synthesis, crystal structure, evaluation of urease inhibition potential and the docking studies of cobalt(III) complex based on barbituric acid Schiff base ligand. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2019.119405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
18
|
Salehi Ashani R, Azizian H, Sadeghi Alavijeh N, Fathi Vavsari V, Mahernia S, Sheysi N, Biglar M, Amanlou M, Balalaie S. Synthesis, Biological Evaluation and Molecular Docking of Deferasirox and Substituted 1,2,4-Triazole Derivatives as Novel Potent Urease Inhibitors: Proposing Repositioning Candidate. Chem Biodivers 2020; 17:e1900710. [PMID: 32187446 DOI: 10.1002/cbdv.201900710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/16/2020] [Indexed: 01/19/2023]
Abstract
A series of new deferasirox derivatives were synthesized through the reaction of monosubstituted hydrazides with 2-(2-hydroxyphenyl)-4H-benzo[e][1,3]oxazin-4-one. For the first time, deferasirox and some of its derivatives were evaluated for their in vitro inhibitory activity against Jack bean urease. The potencies of the members of this class of compounds are higher than that of acetohydroxamic acid. Two compounds, bearing tetrazole and hydrazine derivatives (bioisoester of carboxylate group), represented the most potent urease inhibitory activity with IC50 values of 1.268 and 3.254 μm, respectively. In silico docking studies were performed to delineate possible binding modes of the compounds with the enzyme, urease. Docking analysis suggests that the synthesized compounds were anchored well in the catalytic site and extending to the entrance of binding pocket and thus restrict the mobility of the flap by interacting with its crucial amino acid residues, CME592 and His593. The overall results of urease inhibition have shown that these target compounds can be further optimized and developed as a lead skeleton for the discovery of novel urease inhibitors.
Collapse
Affiliation(s)
- Razieh Salehi Ashani
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, P.O. Box, 15875-4416, Tehran, Iran
| | - Homa Azizian
- Department of Medicinal Chemistry, School of Pharmacy, International Campus, Iran University of Medical Sciences, P.O. Box, 14665-354, Tehran, Iran
| | - Nahid Sadeghi Alavijeh
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, P.O. Box, 15875-4416, Tehran, Iran
| | - Vaezeh Fathi Vavsari
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, P.O. Box, 15875-4416, Tehran, Iran
| | - Shabnam Mahernia
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, P.O. Box, 14155-6451, Tehran, Iran
| | - Niloofar Sheysi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box, 14155-6451, Tehran, Iran
| | - Mahmood Biglar
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, P.O. Box, 14155-6451, Tehran, Iran
| | - Massoud Amanlou
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, P.O. Box, 14155-6451, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. Box, 14155-6451, Tehran, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, P.O. Box, 15875-4416, Tehran, Iran.,Medical Biology Research Center, Kermanshah University of Medical Sciences, P.O. Box, 67155-1616, Kermanshah, Iran
| |
Collapse
|
19
|
Lu Q, Li C, Wu G. Insight into the inhibitory effects of Zanthoxylum nitidum against Helicobacter pylori urease and jack bean urease: Kinetics and mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2020; 249:112419. [PMID: 31759110 DOI: 10.1016/j.jep.2019.112419] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zanthoxylum nitidum (Roxb.) DC. is a traditional Chinese medicine characterised by anti-inflammatory and anti-Helicobacter pylori, which is widely used to treat H. pylori-induced gastric disease in China. However, the underlying mechanism related to its anti-H. pylori activity remains unclear. Urease plays a crucial role in the colonisation and survival of H. pylori. AIM OF THE STUDY The root aqueous extract of Z. nitidum against H. pylori urease (HPU) and jack bean urease (JBU) was investigated to illuminate the inhibitory potency, kinetics and potential mechanism. MATERIALS AND METHODS Z. nitidum components were determined by UPLC. The enzyme inhibitory effects of Z. nitidum were examined using modified spectrophotometric Berthelot (phenol-hypochlorite) method. Urease inhibition kinetics were determined by Lineweaver-Burk plots. Sulfhydryl group reagents and Ni2+-binding inhibitors were used in the mechanism study. Moreover, the molecular docking technique was used to investigate the binding conformations of the main compounds of Z. nitidum on Urease. RESULTS According to UPLC results, the major components of Z. nitidum were magnoflorine, sanguinarine, nitidine chloride, chelerythrine, skimmianine and L-Sesamin. Z. nitidum has higher enzyme inhibitory activity on HPU (IC50 = 1.29 ± 0.10 mg/mL) than on JBU (IC50 = 2.04 ± 0.27 mg/mL). Enzyme inhibitory kinetic analysis revealed that the type of Z. nitidum inhibition against HPU was a slow-binding and mixed-type, whereas a slow-binding and non-competitive type inhibited JBU. Further mechanism study indicated that the active site of sulfhydryl group might be the target of inhibition by Z. nitidum. The molecular docking study indicated that the above six main components of Z. nitidum exhibited stronger affinity to HPU than to JBU through interacting with the key amino acid residues located on the mobile flap or interacting with the active site Ni2+. Results indicated that these components are potential active ingredients directed against urease. CONCLUSIONS Z. nitidum inactivated urease in a concentration-dependent manner through slow-binding inhibition and binding to the urease active site sulfhydryl group. Our investigation might provide experimental evidence for the traditional application of Z. nitidum in the treatment of H. pylori-associated gastric disorders.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, PR China.
| | - Cailan Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, PR China.
| | - Guosong Wu
- Pharmacy Department, Guangzhou the People's Hospital of Baiyun District, Guangzhou, 510500, PR China.
| |
Collapse
|
20
|
Akkas T, Zakharyuta A, Taralp A, Ow-Yang CW. Cross-linked enzyme lyophilisates (CLELs) of urease: A new method to immobilize ureases. Enzyme Microb Technol 2020; 132:109390. [DOI: 10.1016/j.enzmictec.2019.109390] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 08/01/2019] [Accepted: 08/04/2019] [Indexed: 12/31/2022]
|
21
|
Li Y, Xu L, Duan M, Zhang B, Wang Y, Guan Y, Wu J, Jing C, You Z. Syntheses, characterization, crystal structures and Jack bean urease inhibitory activities of ZnII, CoII/III and NiII complexes derived from reduced Schiff base ligand. Polyhedron 2019. [DOI: 10.1016/j.poly.2019.03.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Li Y, Xu L, Duan M, Wu J, Wang Y, Dong K, Han M, You Z. An acetohydroxamate-coordinated oxidovanadium(V) complex derived from pyridinohydrazone ligand with urease inhibitory activity. INORG CHEM COMMUN 2019. [DOI: 10.1016/j.inoche.2019.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Arqué X, Romero-Rivera A, Feixas F, Patiño T, Osuna S, Sánchez S. Intrinsic enzymatic properties modulate the self-propulsion of micromotors. Nat Commun 2019; 10:2826. [PMID: 31249381 PMCID: PMC6597730 DOI: 10.1038/s41467-019-10726-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/24/2019] [Indexed: 11/17/2022] Open
Abstract
Bio-catalytic micro- and nanomotors self-propel by the enzymatic conversion of substrates into products. Despite the advances in the field, the fundamental aspects underlying enzyme-powered self-propulsion have rarely been studied. In this work, we select four enzymes (urease, acetylcholinesterase, glucose oxidase, and aldolase) to be attached on silica microcapsules and study how their turnover number and conformational dynamics affect the self-propulsion, combining both an experimental and molecular dynamics simulations approach. Urease and acetylcholinesterase, the enzymes with higher catalytic rates, are the only enzymes capable of producing active motion. Molecular dynamics simulations reveal that urease and acetylcholinesterase display the highest degree of flexibility near the active site, which could play a role on the catalytic process. We experimentally assess this hypothesis for urease micromotors through competitive inhibition (acetohydroxamic acid) and increasing enzyme rigidity (β-mercaptoethanol). We conclude that the conformational changes are a precondition of urease catalysis, which is essential to generate self-propulsion. Self-propulsion of biocatalytic micro- and nanomotors is facilitated by enzymes converting substrates into products. Here, the authors show that intrinsic enzymatic properties such as conformational changes are crucial for the self-propulsion of silica microcapsules modified with urease.
Collapse
Affiliation(s)
- Xavier Arqué
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Spain
| | - Adrian Romero-Rivera
- CompBioLab Group, Institut de Química Computacional i Catàlisi (IQCC) and Departament de Química, Universitat de Girona, Carrer Maria Aurèlia Capmany 69, 17003, Girona, Spain
| | - Ferran Feixas
- CompBioLab Group, Institut de Química Computacional i Catàlisi (IQCC) and Departament de Química, Universitat de Girona, Carrer Maria Aurèlia Capmany 69, 17003, Girona, Spain
| | - Tania Patiño
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Spain.
| | - Sílvia Osuna
- CompBioLab Group, Institut de Química Computacional i Catàlisi (IQCC) and Departament de Química, Universitat de Girona, Carrer Maria Aurèlia Capmany 69, 17003, Girona, Spain. .,Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, 08010, Barcelona, Spain.
| | - Samuel Sánchez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, 08028, Barcelona, Spain. .,Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
24
|
Gholivand K, Pooyan M, Mohammadpanah F, Pirastefar F, Junk PC, Wang J, Ebrahimi Valmoozi AA, Mani-Varnosfaderani A. Synthesis, crystal structure and biological evaluation of new phosphoramide derivatives as urease inhibitors using docking, QSAR and kinetic studies. Bioorg Chem 2019; 86:482-493. [DOI: 10.1016/j.bioorg.2019.01.064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/20/2019] [Accepted: 01/27/2019] [Indexed: 12/24/2022]
|
25
|
Duan M, Li Y, Xu L, Yang H, Luo F, Guan Y, Zhang B, Jing C, You Z. Synthesis, crystal structure and urease inhibition of a trinuclear copper(II) complex with reduced Schiff base ligand. INORG CHEM COMMUN 2019. [DOI: 10.1016/j.inoche.2018.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
26
|
Robidillo CJT, Aghajamali M, Faramus A, Sinelnikov R, Veinot JGC. Interfacing enzymes with silicon nanocrystals through the thiol-ene reaction. NANOSCALE 2018; 10:18706-18719. [PMID: 30270384 DOI: 10.1039/c8nr05368e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This study reports the preparation of functional bioinorganic hybrids, through application of the thiol-ene reaction, that exhibit catalytic activity and photoluminescent properties from enzymes and freestanding silicon nanocrystals. Thermal hydrosilylation of 1,7-octadiene and alkene-terminated poly(ethylene oxide)methyl ether with hydride-terminated silicon nanocrystals afforded nanocrystals functionalized with alkene residues and poly(ethylene oxide) moieties. These silicon nanocrystals were conjugated with representative enzymes through the photochemical thiol-ene reaction to afford bioinorganic hybrids that are dispersible and photostable in buffer, and that exhibit photoluminescence (λmax = 630 nm) and catalytic activity. They were characterized using Fourier transform infrared spectroscopy (FTIR), X-ray photoelectron spectroscopy (XPS), transmission electron microscopy (TEM), dynamic light scattering analysis (DLS), absorption spectroscopy, steady-state and time-resolved photoluminescence spectroscopy, and pertinent enzyme activity assays. The general derivatization approach presented for interfacing enzymes with biocompatible silicon nanocrystals has far reaching implications for many applications ranging from sensors to therapeutic agents. The bioinorganic hybrids presented herein have potential applications in the chemical detection of nitrophenyl esters and urea. They can also be employed in enzyme-based theranostics as they combine long-lived silicon nanocrystal photoluminescence with substrate-specific enzymatic activity.
Collapse
|
27
|
Li C, Huang P, Wong K, Xu Y, Tan L, Chen H, Lu Q, Luo C, Tam C, Zhu L, Su Z, Xie J. Coptisine-induced inhibition of Helicobacter pylori: elucidation of specific mechanisms by probing urease active site and its maturation process. J Enzyme Inhib Med Chem 2018; 33:1362-1375. [PMID: 30191728 PMCID: PMC6136390 DOI: 10.1080/14756366.2018.1501044] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In this study, we examined the anti-Helicobactor pylori effects of the main protoberberine-type alkaloids in Rhizoma Coptidis. Coptisine exerted varying antibacterial and bactericidal effects against three standard H. pylori strains and eleven clinical isolates, including four drug-resistant strains, with minimum inhibitory concentrations ranging from 25 to 50 μg/mL and minimal bactericidal concentrations ranging from 37.5 to 125 μg/mL. Coptisine’s anti-H. pylori effects derived from specific inhibition of urease in vivo. In vitro, coptisine inactivated urease in a concentration-dependent manner through slow-binding inhibition and involved binding to the urease active site sulfhydryl group. Coptisine inhibition of H. pylori urease (HPU) was mixed type, while inhibition of jack bean urease was non-competitive. Importantly, coptisine also inhibited HPU by binding to its nickel metallocentre. Besides, coptisine interfered with urease maturation by inhibiting activity of prototypical urease accessory protein UreG and formation of UreG dimers and by promoting dissociation of nickel from UreG dimers. These findings demonstrate that coptisine inhibits urease activity by targeting its active site and inhibiting its maturation, thereby effectively inhibiting H. pylori. Coptisine may thus be an effective anti-H. pylori agent.
Collapse
Affiliation(s)
- Cailan Li
- a Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Ping Huang
- b School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Kambo Wong
- c School of Life Sciences , Center for Protein Science and Crystallography, The Chinese University of Hong Kong , P. R. China
| | - Yifei Xu
- b School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Lihua Tan
- a Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Hanbin Chen
- d The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Qiang Lu
- e Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education and Research Center of Chinese Herbal Resource Science and Engineering , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Chaodan Luo
- a Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Chunlai Tam
- c School of Life Sciences , Center for Protein Science and Crystallography, The Chinese University of Hong Kong , P. R. China
| | - Lixiang Zhu
- b School of Pharmaceutical Sciences , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Ziren Su
- a Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine , Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| | - Jianhui Xie
- f Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome , The Second Affiliated Hospital, Guangzhou University of Chinese Medicine , Guangzhou , P. R. China
| |
Collapse
|
28
|
You Z, Yu H, Li Z, Zhai W, Jiang Y, Li A, Guo S, Li K, Lv C, Zhang C. Inhibition studies of Jack bean urease with hydrazones and their copper(II) complexes. Inorganica Chim Acta 2018. [DOI: 10.1016/j.ica.2018.05.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
29
|
Sangeeta S, Ahmad K, Noorussabah N, Bharti S, Mishra M, Sharma S, Choudhary M. Synthesis, crystal structures, molecular docking and urease inhibition studies of Ni(II) and Cu(II) Schiff base complexes. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2017.11.062] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
30
|
Lage TCA, Maciel TMS, Mota YCC, Sisto F, Sabino JR, Santos JCC, Figueiredo IM, Masia C, de Fátima Â, Fernandes SA, Modolo LV. In vitroinhibition ofHelicobacter pyloriand interaction studies of lichen natural products with jack bean urease. NEW J CHEM 2018. [DOI: 10.1039/c8nj00072g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The interaction between lichen metabolites and jack bean urease was investigated by molecular spectroscopy at pH 7.4 and kinetics assay.
Collapse
Affiliation(s)
- Tiago C. A. Lage
- Grupo de Química Supramolecular e Biomimética (GQSB), Departamento de Química, Universidade Federal de Viçosa
- Viçosa
- Brazil
| | - Thamilla Maria S. Maciel
- Laboratório de Instrumentação e Desenvolvimento em Química Analítica (LINQA), Instituto de Química e Biotecnologia, Universidade Federal de Alagoas
- Maceió
- Brazil
| | - Yane C. C. Mota
- Grupo de Estudos em Bioquímica de Plantas (GEBioPlan), Departamento de Botânica, Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Francesca Sisto
- Dipartimento di Scienze Biomediche
- Chirurgiche ed Odontoiatriche
- Università degli Studi di
- Milano
- Italy
| | - José R. Sabino
- Grupo de Cristalografia, Instituto de Física, Universidade Federal de Goiás
- Goiânia
- Brazil
| | - Josué C. C. Santos
- Laboratório de Instrumentação e Desenvolvimento em Química Analítica (LINQA), Instituto de Química e Biotecnologia, Universidade Federal de Alagoas
- Maceió
- Brazil
| | - Isis M. Figueiredo
- Laboratório de Instrumentação e Desenvolvimento em Química Analítica (LINQA), Instituto de Química e Biotecnologia, Universidade Federal de Alagoas
- Maceió
- Brazil
| | - Carla Masia
- Dipartimento di Scienze Biomediche
- Chirurgiche ed Odontoiatriche
- Università degli Studi di
- Milano
- Italy
| | - Ângelo de Fátima
- Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Departamento de Química, Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| | - Sergio A. Fernandes
- Grupo de Química Supramolecular e Biomimética (GQSB), Departamento de Química, Universidade Federal de Viçosa
- Viçosa
- Brazil
| | - Luzia V. Modolo
- Grupo de Estudos em Bioquímica de Plantas (GEBioPlan), Departamento de Botânica, Universidade Federal de Minas Gerais
- Belo Horizonte
- Brazil
| |
Collapse
|
31
|
Syntheses, structures, and inhibition studies of Jack bean urease by copper(II) complexes derived from a tridentate hydrazone ligand. Inorganica Chim Acta 2018. [DOI: 10.1016/j.ica.2017.09.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
32
|
Wang H, Zhang X, Zhao Y, Zhang D, Jin F, Fan Y. Three Co(II) complexes with a sexidentate N 2 O 4 -donor bis-Schiff base ligand: Synthesis, crystal structures, DFT studies, urease inhibition and molecular docking studies. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.07.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
33
|
Krajewska B. Urease-aided calcium carbonate mineralization for engineering applications: A review. J Adv Res 2017; 13:59-67. [PMID: 30094083 PMCID: PMC6077181 DOI: 10.1016/j.jare.2017.10.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 12/21/2022] Open
Abstract
Inducing calcium carbonate precipitation is another important function of urease in nature. The process takes advantage of the supply of carbonate ions derived from urea hydrolysis and of an increase in pH generated by the reaction, effects that in the presence of Ca2+ ions lead to the precipitation of CaCO3. Further to its importance in nature, if performed in a biomimetic manner, the urease-aided CaCO3 mineralization offers enormous potential in innovative engineering applications as an eco-friendly technique operative under mild conditions, to be used for remediation and cementation/deposition in field applications in situ. These include among others, the strengthening and consolidation of soil/sand, the protection and restoration of stone and concrete structures, conservation of stone cultural heritage materials, cleaning waste- and groundwater of toxic metals and radionuclides, and plugging geological formations for the enhancement of oil recovery and geologic CO2 sequestration. In view of the potential of this newly emerging interdisciplinary branch of engineering, this article presents the principles of urease-aided calcium carbonate mineralization apposed to other biomineralization processes, and reviews the advantages and limitations of the technique compared to the conventional techniques presently in use. Further, it presents areas of its existing and potential applications, notably in geotechnical, construction and environmental engineering, and its future perspectives.
Collapse
|
34
|
Guo S, Wang T, Xin J, Hu Q, Ren S, Sheng G, Pan L, Zhang C, Li K, You Z. Syntheses, crystal structures, and Jack bean urease inhibitory activities of copper(II) complexes derived from 4-tert-butyl-N′-(1-(pyridin-2-yl)ethylidene)benzohydrazide. J COORD CHEM 2017. [DOI: 10.1080/00958972.2017.1390569] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Sihan Guo
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P. R. China
| | - Tianrui Wang
- School of Life Sciences, Shandong University of Technology, Zibo, P. R. China
| | - Jiajin Xin
- School of Life Sciences, Shandong University of Technology, Zibo, P. R. China
| | - Qiqige Hu
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P. R. China
| | - Shanfa Ren
- School of Life Sciences, Shandong University of Technology, Zibo, P. R. China
| | - Guihua Sheng
- School of Life Sciences, Shandong University of Technology, Zibo, P. R. China
| | - Lin Pan
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P. R. China
| | - Chenglu Zhang
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P. R. China
| | - Kun Li
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P. R. China
| | - Zhonglu You
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P. R. China
| |
Collapse
|
35
|
Design, synthesis, in vitro Evaluation and docking studies on dihydropyrimidine-based urease inhibitors. Bioorg Chem 2017; 74:53-65. [DOI: 10.1016/j.bioorg.2017.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 12/28/2022]
|
36
|
Yousaf M. A Comparative Kinetic Study of Free and Immobilized Urease on Commercial and Glutaraldehyde Activated Cotton. ACTA ACUST UNITED AC 2017. [DOI: 10.6000/1927-5129.2017.13.53] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
37
|
Tan L, Li C, Chen H, Mo Z, Zhou J, Liu Y, Ma Z, Xu Y, Yang X, Xie J, Su Z. Epiberberine, a natural protoberberine alkaloid, inhibits urease of Helicobacter pylori and jack bean: Susceptibility and mechanism. Eur J Pharm Sci 2017; 110:77-86. [PMID: 28167234 DOI: 10.1016/j.ejps.2017.02.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 01/28/2017] [Accepted: 02/02/2017] [Indexed: 12/31/2022]
Abstract
In our previous study, Rhizoma Coptidis extract was found to exert more potent inhibitory effect than its major component berberine towards urease from Helicobacter pylori (HPU) and jack bean (JBU). In continuation of our work, the present study was designed to further comparatively investigate the urease inhibitory activities of five major protoberberine alkaloids in Rhizoma Coptidis, namely berberine, palmatine, coptisine, epiberberine, jateorhizine to identify the bioactive constituent, and illuminate the potential mechanism of action. Results indicated that the five protoberberine alkaloids acted as concentration-dependent inactivators of urease with IC50 values ranging between 3.0 and 5087μM for HPU and 2.3->10,000μM for JBU, respectively. Notably, epiberberine (EB) was found to be the most potent inhibitor against both ureases with IC50 values of 3.0±0.01μM for HPU and 2.3±0.01μM for JBU, which was more effective than the standard urease inhibitor, acetohydroxamic acid (83±0.01μM for HPU and 22±0.01μM for JBU, respectively). Further kinetic analysis revealed that the type of EB inhibition against HPU was slow-binding and uncompetitive, with Ki of 10.6±0.01μM, while slow-binding and competitive against JBU with Ki of 4.6±0.01μM. Addition of thiol reagents, such as l-cysteine, glutathione and dithiothreitol, significantly abolished the inhibition, while Ni2+ competitive inhibitors, boric acid and sodium fluoride, synergetically inhibited urease with EB, indicating the obligatory role of the active site sulfhydryl group for the inhibition. In addition, binding of EB with the urease proved to be reversible, as about 65% and 90% enzymatic activity of HPU and JBU, respectively, could be restored by dithiothreitol application. These findings highlighted the potential role of Rhizoma Coptidis protoberberine alkaloids, especially EB, as a lead urease inhibitor in the treatment of diseases associated with ureolytic bacteria. Thus, EB had good potential for further development into a promising therapeutic approach for the treatment of urease-related diseases.
Collapse
Affiliation(s)
- Lihua Tan
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China; Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Guangzhou 510006, People's Republic of China
| | - Cailan Li
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China; Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Guangzhou 510006, People's Republic of China
| | - Hanbin Chen
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, People's Republic of China
| | - Zhizhun Mo
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China; Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Guangzhou 510006, People's Republic of China
| | - Jiangtao Zhou
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China; Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Guangzhou 510006, People's Republic of China
| | - Yuhong Liu
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China; Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Guangzhou 510006, People's Republic of China
| | - Zhilin Ma
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Yuyao Xu
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China
| | - Xiaobo Yang
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, People's Republic of China
| | - Jianhui Xie
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, People's Republic of China.
| | - Ziren Su
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou 510006, People's Republic of China; Guangdong Provincial Key Laboratory of New Chinese Medicinal Development and Research, Guangzhou 510006, People's Republic of China; Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan 523808, People's Republic of China.
| |
Collapse
|
38
|
|
39
|
Wang J, Qu D, Lei JX, You Z. Synthesis, crystal structures and Jack bean urease inhibitory activity of copper(II) complexes with 4-bromo-N′-(2-hydroxy-5-methoxybenzylidene)benzohydrazide. J COORD CHEM 2016. [DOI: 10.1080/00958972.2016.1262538] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Jing Wang
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| | - Dan Qu
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| | - Jin-Xian Lei
- College of Chemistry and Chemical Engineering, Jinzhong University, Jinzhong, P.R. China
| | - Zhonglu You
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, P.R. China
| |
Collapse
|
40
|
Pan L, Wang C, Yan K, Zhao K, Sheng G, Zhu H, Zhao X, Qu D, Niu F, You Z. Synthesis, structures and Helicobacter pylori urease inhibitory activity of copper(II) complexes with tridentate aroylhydrazone ligands. J Inorg Biochem 2016; 159:22-8. [DOI: 10.1016/j.jinorgbio.2016.02.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 01/07/2016] [Accepted: 02/10/2016] [Indexed: 10/22/2022]
|
41
|
You Z, Liu M, Wang C, Sheng G, Zhao X, Qu D, Niu F. Inhibition studies of Helicobacter pylori urease with Schiff base copper(ii) complexes. RSC Adv 2016. [DOI: 10.1039/c6ra00500d] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Nine new copper(ii) complexes derived from various Schiff bases were prepared. Five complexes show effective urease inhibitory activities. Complex5has the most effective activity against urease, with a mixed competitive inhibition mechanism.
Collapse
Affiliation(s)
- Zhonglu You
- Department of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- P. R. China
| | - Mingyang Liu
- Department of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- P. R. China
| | - Cunfang Wang
- School of Life Sciences
- Shandong University of Technology
- Zibo 255049
- P. R. China
| | - Guihua Sheng
- School of Life Sciences
- Shandong University of Technology
- Zibo 255049
- P. R. China
| | - Xinlu Zhao
- Department of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- P. R. China
| | - Dan Qu
- Department of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- P. R. China
| | - Fang Niu
- Department of Chemistry and Chemical Engineering
- Liaoning Normal University
- Dalian 116029
- P. R. China
| |
Collapse
|
42
|
Jiang XY, Sheng LQ, Song CF, Du NN, Xu HJ, Liu ZD, Chen SS. Mechanism, kinetics, and antimicrobial activities of 2-hydroxy-1-naphthaldehyde semicarbazone as a new Jack bean urease inhibitor. NEW J CHEM 2016. [DOI: 10.1039/c5nj01601k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new inhibitor of jack bean urease, 2-hydroxy-1-naphthaldehyde semicarbazone, was synthesized and employed to investigate the inhibitory mechanism of HNDSC on jack bean urease by kinetic and fluorescence titration assay, and its antibacterial activities were also investigated.
Collapse
Affiliation(s)
- Xue-Yue Jiang
- School of Chemistry and Material Engineering
- Fuyang Normal College
- Fuyang
- China
| | - Liang-Quan Sheng
- School of Chemistry and Material Engineering
- Fuyang Normal College
- Fuyang
- China
- College of Chemistry and Chemical Engineering
| | - Chong-Fu Song
- School of Chemistry and Material Engineering
- Fuyang Normal College
- Fuyang
- China
| | - Na-Na Du
- School of Chemistry and Material Engineering
- Fuyang Normal College
- Fuyang
- China
| | - Hua-Jie Xu
- School of Chemistry and Material Engineering
- Fuyang Normal College
- Fuyang
- China
| | - Zhao-Di Liu
- School of Chemistry and Material Engineering
- Fuyang Normal College
- Fuyang
- China
| | - Shui-Sheng Chen
- School of Chemistry and Material Engineering
- Fuyang Normal College
- Fuyang
- China
| |
Collapse
|
43
|
Jing C, Wang C, Yan K, Zhao K, Sheng G, Qu D, Niu F, Zhu H, You Z. Synthesis, structures and urease inhibitory activity of cobalt(III) complexes with Schiff bases. Bioorg Med Chem 2015; 24:270-6. [PMID: 26712097 DOI: 10.1016/j.bmc.2015.12.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/04/2015] [Accepted: 12/07/2015] [Indexed: 11/27/2022]
Abstract
A series of new cobalt(III) complexes were prepared. They are [CoL(1)(py)3]·NO3 (1), [CoL(2)(bipy)(N3)]·CH3OH (2), [CoL(3)(HL(3))(N3)]·NO3 (3), and [CoL(4)(MeOH)(N3)] (4), where L(1), L(2), L(3) and L(4) are the deprotonated form of N'-(2-hydroxy-5-methoxybenzylidene)-3-methylbenzohydrazide, N'-(2-hydroxybenzylidene)-3-hydroxylbenzohydrazide, 2-[(2-dimethylaminoethylimino)methyl]-4-methylphenol, and N,N'-bis(5-methylsalicylidene)-o-phenylenediamine, respectively, py is pyridine, and bipy is 2,2'-bipyridine. The complexes were characterized by infrared and UV-Vis spectra, and single crystal X-ray diffraction. The Co atoms in the complexes are in octahedral coordination. Complexes 1 and 4 show effective urease inhibitory activities, with IC50 values of 4.27 and 0.35 μmol L(-1), respectively. Complex 2 has medium activity against urease, with IC50 value of 68.7 μmol L(-1). While complex 3 has no activity against urease. Molecular docking study of the complexes with Helicobacter pylori urease was performed.
Collapse
Affiliation(s)
- Changling Jing
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, PR China
| | - Cunfang Wang
- School of Life Sciences, Shandong University of Technology, Zibo 255049, PR China
| | - Kai Yan
- School of Life Sciences, Shandong University of Technology, Zibo 255049, PR China
| | - Kedong Zhao
- School of Life Sciences, Shandong University of Technology, Zibo 255049, PR China
| | - Guihua Sheng
- School of Life Sciences, Shandong University of Technology, Zibo 255049, PR China
| | - Dan Qu
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, PR China
| | - Fang Niu
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, PR China
| | - Hailiang Zhu
- School of Life Sciences, Shandong University of Technology, Zibo 255049, PR China
| | - Zhonglu You
- Department of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, PR China.
| |
Collapse
|
44
|
Urease immobilized polymer hydrogel: Long-term stability and enhancement of enzymatic activity. J Colloid Interface Sci 2015; 463:164-72. [PMID: 26520823 DOI: 10.1016/j.jcis.2015.10.051] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/17/2015] [Accepted: 10/20/2015] [Indexed: 11/24/2022]
Abstract
A method has been developed in which an enzyme namely urease was immobilized inside hydrogel matrix to study the stability and enzymatic activity in room temperature (∼27-30°C). This urease coupled hydrogel (UCG) was obtained by amine-acid coupling reaction and this procedure is such that it ensured the wider opening of mobile flap of enzyme active site. A systematic comparison of urea-urease assay and the detailed kinetic data clearly revealed that the urease shows activity for more than a month when stored at ∼27-30°C in case of UCG whereas it becomes inactive in case of free urease (enzyme in buffer solution). The aqueous microenvironment inside the hydrogel, unusual morphological features and thermal behaviour were believed to be the reasons for unexpected behaviour. UCG displayed enzyme activity at basic pH and up to 60°C. UCG showed significant enhancement in activity against thermal degradation compared to free urease. In summary, this method is a suitable process to stabilize the biomacromolecules in standard room temperature for many practical uses.
Collapse
|
45
|
Mo ZZ, Wang XF, Zhang X, Su JY, Chen HM, Liu YH, Zhang ZB, Xie JH, Su ZR. Andrographolide sodium bisulphite-induced inactivation of urease: inhibitory potency, kinetics and mechanism. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:238. [PMID: 26179287 PMCID: PMC4504079 DOI: 10.1186/s12906-015-0775-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/13/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND The inhibitory effect of andrographolide sodium bisulphite (ASB) on jack bean urease (JBU) and Helicobacter pylori urease (HPU) was performed to elucidate the inhibitory potency, kinetics and mechanism of inhibition in 20 mM phosphate buffer, pH 7.0, 2 mM EDTA, 25 °C. METHODS The ammonia formations, indicator of urease activity, were examined using modified spectrophotometric Berthelot (phenol-hypochlorite) method. The inhibitory effect of ASB was characterized with IC50 values. Lineweaver-Burk and Dixon plots for JBU inhibition of ASB was constructed from the kinetic data. SH-blocking reagents and competitive active site Ni2+ binding inhibitors were employed for mechanism study. Molecular docking technique was used to provide some information on binding conformations as well as confirm the inhibition mode. RESULTS The IC50 of ASB against JBU and HPU was 3.28±0.13 mM and 3.17±0.34 mM, respectively. The inhibition proved to be competitive and concentration- dependent in a slow-binding progress. The rapid formation of initial ASB-JBU complex with an inhibition constant of Ki=2.86×10(-3) mM was followed by a slow isomerization into the final complex with an overall inhibition constant of Ki*=1.33×10(-4) mM. The protective experiment proved that the urease active site is involved in the binding of ASB. Thiol reagents (L-cysteine and dithiothreithol) strongly protect the enzyme from the loss of enzymatic activity, while boric acid and fluoride show weaker protection, indicating that the active-site sulfhydryl group of JBU was potentially involved in the blocking process. Moreover, inhibition of ASB proved to be reversible since ASB-inactivated JBU could be reactivated by dithiothreitol application. Molecular docking assay suggested that ASB made contacts with the important sulfhydryl group Cys-592 residue and restricted the mobility of the active-site flap. CONCLUSIONS ASB was a competitive inhibitor targeting thiol groups of urease in a slow-binding manner both reversibly and concentration-dependently, serving as a promising urease inhibitor for the treatment of urease-related diseases.
Collapse
Affiliation(s)
- Zhi-Zhun Mo
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Xiu-Fen Wang
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Xie Zhang
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Ji-Yan Su
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Hai-Ming Chen
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Yu-Hong Liu
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Zhen-Biao Zhang
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
| | - Jian-Hui Xie
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, P. R. China.
| | - Zi-Ren Su
- Guangdong Provincial Key Laboratory of of New Chinese Medicinals Development and Research, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China.
- Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Dongguan, 523000, P. R. China.
| |
Collapse
|
46
|
Khan AL, Ali L, Hussain J, Rizvi TS, Al-Harrasi A, Lee IJ. Enzyme Inhibitory Radicinol Derivative from Endophytic fungus Bipolaris sorokiniana LK12, Associated with Rhazya stricta. Molecules 2015; 20:12198-208. [PMID: 26151116 PMCID: PMC6331987 DOI: 10.3390/molecules200712198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/17/2015] [Accepted: 06/30/2015] [Indexed: 11/16/2022] Open
Abstract
Endophytes, living inside plant tissues, play an essential role in plant growth and development, whilst producing unique bioactive secondary metabolites. In the current study, the endophytic fungus Bipolaris sorokiniana LK12 was isolated from the leaves of ethno-medicinal and alkaloidal rich Rhazya stricta. The bulk amount of ethyl acetate extract of fungus was subjected to advance column chromatographic techniques, which resulted in the isolation of a new radicinol derivative, bipolarisenol (1). It was found to be a derivative of radicinol. The structure elucidation was carried out by the combined use of 1D and 2D nuclear magnetic resonance, infrared spectroscopy, mass, and UV spectrometric analyses. The bipolarisenol was assessed for its potential role in enzyme inhibition of urease and acetyl cholinesterase (AChE). Results showed that bipolarisenol significantly inhibited the AChE activity with low IC50 (67.23 ± 5.12 µg·mL-1). Bipolarisenol inhibited urease in a dose-dependent manner with high IC50 (81.62 ± 4.61 µg·mL-1). The new compound also showed a moderate anti-lipid peroxidation potential (IC50 = 168.91 ± 4.23 µg·mL-1). In conclusion, endophytes isolated from medicinal plants possess a unique potential to be considered for future drug discovery.
Collapse
Affiliation(s)
- Abdul Latif Khan
- UoN Chair of Oman's Medicinal Plants and Marine Natural Products, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman.
| | - Liaqat Ali
- UoN Chair of Oman's Medicinal Plants and Marine Natural Products, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman.
| | - Javid Hussain
- UoN Chair of Oman's Medicinal Plants and Marine Natural Products, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman.
- Department of Biological Sciences and Chemistry, College of Arts and Sciences, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman.
| | - Tania Shamim Rizvi
- UoN Chair of Oman's Medicinal Plants and Marine Natural Products, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman.
| | - Ahmed Al-Harrasi
- UoN Chair of Oman's Medicinal Plants and Marine Natural Products, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman.
- Department of Biological Sciences and Chemistry, College of Arts and Sciences, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman.
| | - In-Jung Lee
- School of Applied Biosciences, Kyungpook National University, Daegu 702-701, Korea.
| |
Collapse
|
47
|
Qu D, Niu F, Zhao X, Yan KX, Ye YT, Wang J, Zhang M, You Z. Synthesis, crystal structures, and urease inhibition of an acetohydroxamate-coordinated oxovanadium(V) complex derived from N′-(3-bromo-2-hydroxybenzylidene)-4-methoxybenzohydrazide. Bioorg Med Chem 2015; 23:1944-9. [DOI: 10.1016/j.bmc.2015.03.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 12/30/2022]
|
48
|
Yu XD, Zheng RB, Xie JH, Su JY, Huang XQ, Wang YH, Zheng YF, Mo ZZ, Wu XL, Wu DW, Liang YE, Zeng HF, Su ZR, Huang P. Biological evaluation and molecular docking of baicalin and scutellarin as Helicobacter pylori urease inhibitors. JOURNAL OF ETHNOPHARMACOLOGY 2015; 162:69-78. [PMID: 25557028 DOI: 10.1016/j.jep.2014.12.041] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 09/07/2014] [Accepted: 12/22/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Baicalin and scutellarin are the principal bioactive components of Scutellaria baicalensis Georgi which has extensively been incorporated into heat-clearing and detoxification formulas for the treatment of Helicobacter pylori-related gastrointestinal disorders in traditional Chinese medicine. However, the mechanism of action remained to be defined. AIM OF THE STUDY To explore the inhibitory effect, kinetics and mechanism of Helicobacter pylori urease (the vital pathogenetic factor for Helicobacter pylori infection) inhibition by baicalin and scutellarin, for their therapeutic potential. MATERIALS AND METHODS The ammonia formations, indicator of urease activity, were examined using modified spectrophotometric Berthelot (phenol-hypochlorite) method. The inhibitory effect of baicalin and scutellarin was characterized with IC50 values, compared to acetohydroxamic acid (AHA), a well known Helicobacter pylori urease inhibitor. Lineweaver-Burk and Dixon plots for the Helicobacter pylori urease inhibition of baicalin and scutellarin was constructed from the kinetic data. SH-blocking reagents and competitive active site Ni(2+) binding inhibitors were employed for mechanism study. Molecular docking technique was used to provide some information on binding conformations as well as confirm the inhibition mode. Moreover, cytotoxicity experiment using Gastric Epithelial Cells (GES-1) was evaluated. RESULTS Baicalin and scutellarin effectively suppressed Helicobacter pylori urease in dose-dependent and time-independent manner with IC50 of 0.82±0.07 mM and 0.47±0.04 mM, respectively, compared to AHA (IC50=0.14±0.05 mM). Structure-activity relationship disclosed 4'-hydroxyl gave flavones an advantage to binding with Helicobacter pylori urease. Kinetic analysis revealed that the types of inhibition were non-competitive and reversible with inhibition constant Ki of 0.14±0.01 mM and 0.18±0.02 mM for baicalin and scutellarin, respectively. The mechanism of urease inhibition was considered to be blockage of the SH groups of Helicobacter pylori urease, since thiol reagents (L,D-dithiothreitol, L-cysteine and glutathione) abolished the inhibitory action and competitive active site Ni(2+) binding inhibitors (boric acid and sodium fluoride) carried invalid effect. Molecular docking study further supported the structure-activity analysis and indicated that baicalin and scutellarin interacted with the key residues Cys321 located on the mobile flap through S-H·π interaction, but did not interact with active site Ni(2+). Moreover, Baicalin (at 0.59-1.05 mM concentrations) and scutellarin (at 0.23-0.71 mM concentrations) did not exhibit significant cytotoxicity to GES-1. CONCLUSIONS Baicalin and scutellarin were non-competitive inhibitors targeting sulfhydryl groups especially Cys321 around the active site of Helicobacter pylori urease, representing potential to be good candidate for future research as urease inhibitor for treatment of Helicobacter pylori infection. Furthermore, our work gave additional scientific support to the use of Scutellaria baicalensis in traditional Chinese medicine (TCM) to treat gastrointestinal disorders.
Collapse
Affiliation(s)
- Xiao-Dan Yu
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Rong-Bo Zheng
- Guangzhou Wanglaoji Pharmaceutical Company Limited, Guangzhou, Guangdong 510450, P.R. China
| | - Jian-Hui Xie
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, P.R. China
| | - Ji-Yan Su
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Xiao-Qi Huang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Yong-Hong Wang
- Guangdong Institute of Microbiology, Guangzhou 510006, P.R. China
| | - Yi-Feng Zheng
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Zhi-Zhun Mo
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Xiao-Li Wu
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Dian-Wei Wu
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Ye-er Liang
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Hui-Fang Zeng
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China.
| | - Zi-Ren Su
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China; Dongguan Mathematical Engineering Academy of Chinese Medicine, Guangzhou, University of Chinese Medicine, Dongguan 523000, P.R. China
| | - Ping Huang
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China.
| |
Collapse
|
49
|
Abstract
Thiosulfinates are a major compound found in most allium plant. Garlic, an allium species shows its potential to inhibit microbial and enzyme activity. The usage of garlic is widely known especially in the agriculture field. Thiosulfinates are formed by reaction of alliinase enzyme from their respective S-alk(en)yl cysteine sulfoxide after the crushing process of the garlic cloves. The stability of the garlic extract has been tested at variations of temperatures and pH values at different incubation time in terms of concentration (mM). After consecutive 4 hour incubation, thiols show higher concentration at temperature 30-35°C. The minimum concentration shows 22% reduction from the original thiols reading. Meanwhile, pH values of 2 (acidic) at 35°C gives higher thiols concentration compared to other conditions.
Collapse
|
50
|
Synthesis, characterization and urease inhibition of a novel acetylhydroxamate-coordinated oxovanadium(V) complex with hydrazone ligand. INORG CHEM COMMUN 2014. [DOI: 10.1016/j.inoche.2014.04.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|