1
|
Güzel-Akdemir Ö, Akdemir A. Urease inhibitors for the treatment of H. pylori. Expert Opin Ther Pat 2025; 35:17-30. [PMID: 39495126 DOI: 10.1080/13543776.2024.2423004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 09/16/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Helicobacter pylori infects almost half of the World population. Although many infected people are symptom free, the microorganism can still cause a variety of gastrointestinal disorders and gastric adenocarcinoma. It is considered a priority pathogen for the development of new antibiotics by the World Health Organisation (WHO). Many virulence factors of H. pylori have been described. This paper will on H. pylori Urease (HPU). AREA COVERED This paper will discuss the (patho)physiology and structure of HPU. In addition, urease inhibitors with known activity against the HPU or inhibitors that show H. pylori growth inhibition will be discussed. EXPERT OPINION Increase in selectivity, affinity and potency of HPU inhibitors can be achieved by the design of compounds that interact with distinct regions within the enzyme active site. Especially, covalent interactions seem promising as they clearly effect the dose requirement of the drug candidate.
Collapse
Affiliation(s)
- Özlen Güzel-Akdemir
- Department of Pharmaceutical Chemistry, Istanbul University, Faculty of Pharmacy, Beyazit/Istanbul, Turkey
| | - Atilla Akdemir
- Department of Pharmacology, Faculty of Pharmacy, Istinye University, Sariyer/Istanbul, Turkey
| |
Collapse
|
2
|
Huang G, Cierpicki T, Grembecka J. Thioamides in medicinal chemistry and as small molecule therapeutic agents. Eur J Med Chem 2024; 277:116732. [PMID: 39106658 DOI: 10.1016/j.ejmech.2024.116732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
Thioamides, which are fascinating isosteres of amides, have garnered significant attention in drug discovery and medicinal chemistry programs, spanning peptides and small molecule compounds. This review provides an overview of the various applications of thioamides in small molecule therapeutic agents targeting a range of human diseases, including cancer, microbial infections (e.g., tuberculosis, bacteria, and fungi), viral infections, neurodegenerative conditions, analgesia, and others. Particular focus is given to design strategies of biologically active thioamide-containing compounds and their biological targets, such as kinases and histone methyltransferase ASH1L. Additionally, the review discusses the impact of the thioamide moiety on key properties, including potency, target interactions, physicochemical characteristics, and pharmacokinetics profiles. We hope that this work will offer valuable insights to inspire the future development of novel bioactive thioamide-containing compounds, facilitating their effective use in combating a wide array of human diseases.
Collapse
Affiliation(s)
- Guang Huang
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
3
|
Ganapathy Vilasam Sreekala A, Gupta KK, Nathan VK. Identification of coastal pesticide pollutants as potent inhibitors of Bacillus pasteurii urease mediated calcium carbonate precipitation: a computational approach. J Biomol Struct Dyn 2024; 42:9628-9638. [PMID: 37691444 DOI: 10.1080/07391102.2023.2252089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023]
Abstract
Microbially induced calcite precipitation (MICP) through urease enzyme has attained a lot of recognition in various fields of civil engineering and geotechnology for stabilizing the strength of soil and various concrete materials. The activity of urease has been found to be affected by various factors like temperature, substrate concentrations, pH of the medium, presence of inhibitors, etc. Through this study, the outcome of the interaction of pesticides (commonly found in Indian coastal regions) on Bacillus pasteurii urease, a major organism reported for MICP studies has been investigated in silico. The results from the study revealed that the enzyme has higher interactions of -4.1, -3.2, and -3.4 kJ/mol with common pesticides like dichloro diphenyl dichloro ethane(DDD), dichloro diphenyl trichloroe thane (DDT), and methyl parathion of organochlorides and organophosphates class. From the molecular dynamics simulation analysis, complex 1 (DDD -receptor) has been found to have the highest and more compact structure followed by methyl parathion -receptor. Prime MM-GBSA analysis also revealed the highest binding energy of -27.8 kcal/mol with the protein and DDD. Thus, it can be inferred from the current study that pesticides, particularly, DDD, DDT, and methyl parathion present in the coastal areas may have an impact on urease. This interaction can result in the inhibition of the urease activity of B. pasteurii, thus preventing the biomineralization process. This study would be the first report on the computational approach to understanding the interaction of prominent pesticides on the coastal region and B. pasteurii urease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Krishna Kant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thirumalasamudram, India
| | - Vinod Kumar Nathan
- School of Chemical and Biotechnology, SASTRA Deemed to be University, Thirumalasamudram, India
| |
Collapse
|
4
|
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
|
5
|
Ivanov NM, Slivkov AI, Huck WTS. A Urease-Based pH Photoswitch: A General Route to Light-to-pH Transduction. Angew Chem Int Ed Engl 2024:e202415614. [PMID: 39263723 DOI: 10.1002/anie.202415614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/13/2024]
Abstract
New approaches for the integration of chemical and physical stimuli to control the dynamics of artificial enzymatic reaction networks (ERNs) are needed. Here, we present a general approach to convert a light stimulus into a time-programmed pH response. We developed and characterized a panel of photoswitchable inhibitors of urease. Urease activity, now regulated by light via the photoinhibitors, leads to an increase in pH upon hydrolysis of urea into ammonia. Careful choice of characteristics of light, and concentrations of enzyme, substrate, and photoinhibitor, allowed us to control the timing of the pH transition. Furthermore, as all enzymes have an activity-pH profile, the urease photoinhibitor system can be used to regulate the activities of other enzymes in small reaction networks.
Collapse
Affiliation(s)
- Nikita M Ivanov
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands
| | - Alexandar I Slivkov
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525AJ, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Heylen RA, Cusick N, White T, Owen EJ, Patenall BL, Alm M, Thomsen P, Laabei M, Jenkins ATA. Rational design and in vitro testing of new urease inhibitors to prevent urinary catheter blockage. RSC Med Chem 2024; 15:d4md00378k. [PMID: 39281800 PMCID: PMC11391341 DOI: 10.1039/d4md00378k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/31/2024] [Indexed: 09/18/2024] Open
Abstract
Catheter associated urinary tract infections (CAUTI) caused by urease-positive organisms can lead to catheter blockage: urease metabolizes urea in urine to ammonia causing an increase in pH and hence precipitation of struvite and apatite salts into the catheter lumen and bladder leading to blockage. Acetohydroxamic acid (AHA) is the only urease inhibitor currently approved for patient use, however, it is rarely used owing to its side effects. Here, we report the identification and development of new urease inhibitors discovered using a rational in silico drug design approach. A series of compounds were designed, the compounds were screened and filtered to identify three compounds which were tested in in vitro urease activity assays. N,N'-Bis(3-pyridinylmethyl)thiourea (Bis-TU) outperformed AHA in activity assays and was tested in an in vitro bladder model, where it significantly extended the lifetime of the catheter compared to AHA. Bis-TU was delivered via a diffusible balloon catheter directly to the site of activity, thus demonstrating localized drug delivery. This cost-effective drug design approach allowed the identification of a potent urease inhibitor, which could be improved through iterative repeats of the method, and the process of design could be utilized to target other diseases.
Collapse
Affiliation(s)
| | - Nicola Cusick
- Department of Chemistry, University of Bath Bath BA2 7AY UK
| | - Tom White
- Department of Chemistry, University of Bath Bath BA2 7AY UK
| | - Emily J Owen
- Department of Chemistry, University of Bath Bath BA2 7AY UK
| | | | - Martin Alm
- Biomodics ApS Fjeldhammervej 15 2610 Rødovre Denmark
| | - Peter Thomsen
- Biomodics ApS Fjeldhammervej 15 2610 Rødovre Denmark
| | - Maisem Laabei
- School of Cellular and Molecular Medicine, University of Bristol BS8 1TD Bristol UK
| | | |
Collapse
|
7
|
Ayaz M, Alam A, Zainab, Elhenawy AA, Ur Rehman N, Ur Rahman S, Ali M, Latif A, Al-Harrasi A, Ahmad M. Designing and Synthesis of Novel Fexofenadine-Derived Hydrazone-Schiff Bases as Potential Urease Inhibitors: In-Vitro, Molecular Docking and DFT Investigations. Chem Biodivers 2024; 21:e202400704. [PMID: 38781003 DOI: 10.1002/cbdv.202400704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Thirteen novel hydrazone-Schiff bases (3-15) of fexofenadine were succesfully synthesized, structurally deduced and finally assessed their capability to inhibit urease enzyme (in vitro). In the series, six compounds 12 (IC50=10.19±0.16 μM), 11 (IC50=15.05±1.11 μM), 10 (IC50=17.01±1.23 μM), 9 (IC50=17.22±0.81 μM), 13 (IC50=19.31±0.18 μM), and 14 (IC50=19.62±0.21 μM) displayed strong inhibitory action better than the standard thiourea (IC50=21.14±0.24 μM), while the remaining compounds displayed significant to less inhibition. LUMO and HOMO showed the transferring of charges from molecules to biological transfer and MEP map showed the chemically reactive zone appropriate for drug action are calculated using DFT. AIM charges, non-bonding orbitals, and ELF are also computed. The urease protein binding analysis benefited from the docking studies.
Collapse
Affiliation(s)
- Muhammad Ayaz
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Aftab Alam
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Zainab
- College of Chemistry and Materials Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Najeeb Ur Rehman
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman
| | - Sajjad Ur Rahman
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Mumtaz Ali
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Abdul Latif
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, 616, Oman
| | - Manzoor Ahmad
- Department of Chemistry, University of Malakand, P.O. Box, 18800, Dir, Lower, Pakistan
| |
Collapse
|
8
|
Wang N, Wu X, Liang J, Liu B, Wang B. Molecular design of hydroxamic acid-based derivatives as urease inhibitors of Helicobacter pylori. Mol Divers 2024; 28:2229-2244. [PMID: 39020133 DOI: 10.1007/s11030-024-10914-9] [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: 04/27/2024] [Accepted: 06/08/2024] [Indexed: 07/19/2024]
Abstract
Helicobacter pylori is the main causative agent of gastric cancer, especially non-cardiac gastric cancers. This bacterium relies on urease producing much ammonia to colonize the host. Herein, the study provides valuable insights into structural patterns driving urease inhibition for high-activity molecules designed via exploring known inhibitors. Firstly, an ensemble model was devised to predict the inhibitory activity of novel compounds in an automated workflow (R2 = 0.761) that combines four machine learning approaches. The dataset was characterized in terms of chemical space, including molecular scaffolds, clustering analysis, distribution for physicochemical properties, and activity cliffs. Through these analyses, the hydroxamic acid group and the benzene ring responsible for distinct activity were highlighted. Activity cliff pairs uncovered substituents of the benzene ring on hydroxamic acid derivatives are key structures for substantial activity enhancement. Moreover, 11 hydroxamic acid derivatives were designed, named mol1-11. Results of molecular dynamic simulations showed that the mol9 exhibited stabilization of the active site flap's closed conformation and are expected to be promising drug candidates for Helicobacter pylori infection and further in vitro, in vivo, and clinical trials to demonstrate in future.
Collapse
Affiliation(s)
- Na Wang
- College of Materials and Energy, South China Agricultural University, Guangzhou, 510630, China
| | - Xiaoyan Wu
- College of Materials and Energy, South China Agricultural University, Guangzhou, 510630, China
| | - Jianhuai Liang
- College of Materials and Energy, South China Agricultural University, Guangzhou, 510630, China
| | - Boping Liu
- College of Materials and Energy, South China Agricultural University, Guangzhou, 510630, China.
| | - Bingfeng Wang
- College of Materials and Energy, South China Agricultural University, Guangzhou, 510630, China.
| |
Collapse
|
9
|
Contini L, Paul A, Mazzei L, Ciurli S, Roncarati D, Braga D, Grepioni F. Is bismuth(III) able to inhibit the activity of urease? Puzzling results in the quest for soluble urease complexes for agrochemical and medicinal applications. Dalton Trans 2024; 53:10553-10562. [PMID: 38847020 DOI: 10.1039/d4dt00778f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Bismuth(III) complexes have been reported to act as inhibitors of the enzyme urease, ubiquitously present in soils and implicated in the pathogenesis of several microorganisms. The general insolubility of Bi(III) complexes in water at neutral pH, however, is an obstacle to their utilization. In our quest to improve the solubility of Bi(III) complexes, we selected a compound reported to inhibit urease, namely [Bi(HEDTA)]·2H2O, and co-crystallized it with (i) racemic DL-histidine to obtain the conglomerate [Bi2(HEDTA)2(μ-D-His)2]·6H2O + [Bi2(HEDTA)2(μ-L-His)2]·6H2O, (ii) enantiopure L-histidine to yield [Bi2(HEDTA)2(μ-L-His)2]·6H2O, and (iii) cytosine to obtain [Bi(HEDTA)]·Cyt·2H2O. All compounds, synthesised by mechanochemical methods and by slurry, were characterized in the solid state by calorimetric (DSC and TGA) and spectroscopic (IR) methods, and their structures were determined using powder X-ray diffraction (PXRD) data. All compounds show an appreciable solubility in water, with values ranging from 6.8 mg mL-1 for the starting compound [Bi(HEDTA)]·2H2O to 36 mg mL-1 for [Bi2(HEDTA)2(μ-L-His)2]·6H2O. The three synthesized compounds as well as [Bi(HEDTA)]·2H2O were then tested for inhibition activity against urease. Surprisingly, no enzymatic inhibition was observed during in vitro assays using Canavalia ensiformis urease and in vivo assays using cultures of Helicobacter pylori, raising questions on the efficacy of Bi(III) compounds to counteract the negative effects of urease activity in the agro-environment and in human health.
Collapse
Affiliation(s)
- Laura Contini
- Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy.
| | - Arundhati Paul
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Viale Giuseppe Fanin 40, Bologna I-40127, Italy.
| | - Luca Mazzei
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Viale Giuseppe Fanin 40, Bologna I-40127, Italy.
| | - Stefano Ciurli
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Viale Giuseppe Fanin 40, Bologna I-40127, Italy.
| | - Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Selmi 3, 40126 Bologna, Italy.
| | - Dario Braga
- Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy.
| | - Fabrizia Grepioni
- Department of Chemistry "G. Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy.
| |
Collapse
|
10
|
Hashem O, Zaib S, Zaraei SO, Javed H, Kedia RA, Anbar HS, Khan I, Ravi A, El-Gamal MI, Khoder G. Design and discovery of urease and Helicobacter pylori inhibitors based on benzofuran/benzothiophene-sulfonate and sulfamate scaffolds for the treatment of ureolytic bacterial infections. Int J Biol Macromol 2024; 271:132502. [PMID: 38768915 DOI: 10.1016/j.ijbiomac.2024.132502] [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: 02/26/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
A series of sulfonate and sulfamate derivatives bearing benzofuran or benzothiophene scaffold exhibited potent inhibitory effect on urease enzyme. Most of the derivatives exhibited significantly higher potency than thiourea, the standard inhibitor. Compound 1s was identified as the most potent urease inhibitor with an IC50 value of 0.42 ± 0.08 μM, which is 53-fold more potent than thiourea, positive control (IC50 = 22.3 ± 0.031 μM). The docking results further revealed the binding interactions towards the urease active site. Phenotypic screening revealed that compounds 1c, 1d, 1e, 1f, 1j, 1n, and 1t exhibit high potency against H. pylori with MIC values ranging from 0.00625 to 0.05 mM and IC50 values ranging from 0.0031 to 0.0095 mM, much more potent than the positive control, acetohydroxamic acid (MIC and IC50 values were 12.5 and 7.38 mM, respectively). Additional studies were performed to investigate the toxicity of these compounds against the gastric epithelial cell line (AGS) and their selectivity profile against E. coli, and five Lactobacillus species representative of the gut microflora. Permeability characteristics of the most promising derivatives were investigated in Caco-2 cell line. The results indicate that the compounds could be targeted in the GIT only without systemic side effects.
Collapse
Affiliation(s)
- Omar Hashem
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan.
| | - Seyed-Omar Zaraei
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hira Javed
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan
| | - Reena A Kedia
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hanan S Anbar
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai 19099, United Arab Emirates.
| | - Imtiaz Khan
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Anil Ravi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammed I El-Gamal
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Ghalia Khoder
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
11
|
Munir R, Zaib S, Zia-ur-Rehman M, Javed H, Roohi A, Zaheer M, Fatima N, Bhat MA, Khan I. Exploration of morpholine-thiophene hybrid thiosemicarbazones for the treatment of ureolytic bacterial infections via targeting urease enzyme: Synthesis, biochemical screening and computational analysis. Front Chem 2024; 12:1403127. [PMID: 38855062 PMCID: PMC11157103 DOI: 10.3389/fchem.2024.1403127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
An important component of the pathogenicity of potentially pathogenic bacteria in humans is the urease enzyme. In order to avoid the detrimental impact of ureolytic bacterial infections, the inhibition of urease enzyme appears to be an appealing approach. Therefore, in the current study, morpholine-thiophene hybrid thiosemicarbazone derivatives (5a-i) were designed, synthesized and characterized through FTIR, 1H NMR, 13C NMR spectroscopy and mass spectrometry. A range of substituents including electron-rich, electron-deficient and inductively electron-withdrawing groups on the thiophene ring was successfully tolerated. The synthesized derivatives were evaluated in vitro for their potential to inhibit urease enzyme using the indophenol method. The majority of compounds were noticeably more potent than the conventional inhibitor, thiourea. The lead inhibitor, 2-(1-(5-chlorothiophen-2-yl)ethylidene)-N-(2-morpholinoethyl)hydrazinecarbothioamide (5g) inhibited the urease in an uncompetitive manner with an IC50 value of 3.80 ± 1.9 µM. The findings of the docking studies demonstrated that compound 5g has a strong affinity for the urease active site. Significant docking scores and efficient binding free energies were displayed by the lead inhibitor. Finally, the ADME properties of lead inhibitor (5g) suggested the druglikeness behavior with zero violation.
Collapse
Affiliation(s)
- Rubina Munir
- Department of Chemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | | | - Hira Javed
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Ayesha Roohi
- Department of Chemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Muhammad Zaheer
- Applied Chemistry Research Centre, PCSIR Laboratories Complex, Lahore, Pakistan
| | - Nabiha Fatima
- Department of Chemistry, Kinnaird College for Women, Lahore, Pakistan
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Imtiaz Khan
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
12
|
Han Y, Wang M, Xie H, Zhou Y, Wang S, Wang G. Fabrication of Au nanoclusters confined on hydroxy double salt-based intelligent biosensor for on-site monitoring of urease and its inhibitors. Talanta 2024; 271:125725. [PMID: 38295444 DOI: 10.1016/j.talanta.2024.125725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/16/2024] [Accepted: 01/26/2024] [Indexed: 02/02/2024]
Abstract
Sensitive and convenient sensing of urease and its inhibitors is exceptionally urgent in clinical diagnosis and new drug development. In this study, the gold nanoclusters (AuNCs) and hydroxyl double salt (HDS) were composited by a simple confinement effect to prepare highly fluorescent AuNCs@HDS composites to monitor urease and its drug inhibitors. HDS was used as a matrix to confine AuNCs (AuNCs@HDS), facilitating the emission intensity of AuNCs. However, acidic conditions (low pH) can disrupt the structure of HDS to break the confinement effect, and quench the fluorescence of AuNCs. Therefore, a sensing platform for pH-related enzyme urease detection was constructed based on the sensitive response of AuNCs@HDS to pH. This sensing platform had a linear response range of 0.5-22.5 U/L and a low limit of detection (LOD) of 0.19 U/L for urease. Moreover, this sensing platform was also applied to monitor urease inhibitors and urease in human saliva samples. Additionally, a portable hydrogel kit combined with a smartphone was developed for urease detection to achieve portable, low-cost, instrument-free, and on-site monitoring of urease.
Collapse
Affiliation(s)
- Yaqing Han
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, PR China; College of Medical Engineering & the Key Laboratory for Medical Functional, Nanomaterials, Jining Medical University, Jining, 272067, PR China
| | - Mengke Wang
- College of Medical Engineering & the Key Laboratory for Medical Functional, Nanomaterials, Jining Medical University, Jining, 272067, PR China
| | - Han Xie
- Shenyang Key Laboratory of Medical Molecular Theranostic Probes, School of Pharmacy, Shenyang Medical University, Shenyang, 110034, PR China
| | - Yitong Zhou
- Shenyang Key Laboratory of Medical Molecular Theranostic Probes, School of Pharmacy, Shenyang Medical University, Shenyang, 110034, PR China
| | - Shun Wang
- College of Medical Engineering & the Key Laboratory for Medical Functional, Nanomaterials, Jining Medical University, Jining, 272067, PR China.
| | - Guannan Wang
- College of Medical Engineering & the Key Laboratory for Medical Functional, Nanomaterials, Jining Medical University, Jining, 272067, PR China; Shenyang Key Laboratory of Medical Molecular Theranostic Probes, School of Pharmacy, Shenyang Medical University, Shenyang, 110034, PR China.
| |
Collapse
|
13
|
Morales N, Valdés-Muñoz E, González J, Valenzuela-Hormazábal P, Palma JM, Galarza C, Catagua-González Á, Yáñez O, Pereira A, Bustos D. Machine Learning-Driven Classification of Urease Inhibitors Leveraging Physicochemical Properties as Effective Filter Criteria. Int J Mol Sci 2024; 25:4303. [PMID: 38673888 PMCID: PMC11049951 DOI: 10.3390/ijms25084303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Urease, a pivotal enzyme in nitrogen metabolism, plays a crucial role in various microorganisms, including the pathogenic Helicobacter pylori. Inhibiting urease activity offers a promising approach to combating infections and associated ailments, such as chronic kidney diseases and gastric cancer. However, identifying potent urease inhibitors remains challenging due to resistance issues that hinder traditional approaches. Recently, machine learning (ML)-based models have demonstrated the ability to predict the bioactivity of molecules rapidly and effectively. In this study, we present ML models designed to predict urease inhibitors by leveraging essential physicochemical properties. The methodological approach involved constructing a dataset of urease inhibitors through an extensive literature search. Subsequently, these inhibitors were characterized based on physicochemical properties calculations. An exploratory data analysis was then conducted to identify and analyze critical features. Ultimately, 252 classification models were trained, utilizing a combination of seven ML algorithms, three attribute selection methods, and six different strategies for categorizing inhibitory activity. The investigation unveiled discernible trends distinguishing urease inhibitors from non-inhibitors. This differentiation enabled the identification of essential features that are crucial for precise classification. Through a comprehensive comparison of ML algorithms, tree-based methods like random forest, decision tree, and XGBoost exhibited superior performance. Additionally, incorporating the "chemical family type" attribute significantly enhanced model accuracy. Strategies involving a gray-zone categorization demonstrated marked improvements in predictive precision. This research underscores the transformative potential of ML in predicting urease inhibitors. The meticulous methodology outlined herein offers actionable insights for developing robust predictive models within biochemical systems.
Collapse
Affiliation(s)
- Natalia Morales
- Magíster en Ciencias de la Computación, Universidad Católica del Maule, Talca 3460000, Chile; (N.M.); (J.G.)
| | - Elizabeth Valdés-Muñoz
- Doctorado en Biotecnología Traslacional, Centro de Biotecnología de los Recursos Naturales, Universidad Católica del Maule, Talca 3480094, Chile;
| | - Jaime González
- Magíster en Ciencias de la Computación, Universidad Católica del Maule, Talca 3460000, Chile; (N.M.); (J.G.)
| | - Paulina Valenzuela-Hormazábal
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile;
| | - Jonathan M. Palma
- Facultad de Ingeniería, Universidad de Talca, Curicó 3344158, Chile;
| | - Christian Galarza
- Departamento de Matemáticas, Facultad de Ciencias Naturales y Matemáticas, Escuela Superior Politécnica del Litoral, Guayaquil EC090903, Ecuador; (C.G.); (Á.C.-G.)
| | - Ángel Catagua-González
- Departamento de Matemáticas, Facultad de Ciencias Naturales y Matemáticas, Escuela Superior Politécnica del Litoral, Guayaquil EC090903, Ecuador; (C.G.); (Á.C.-G.)
| | - Osvaldo Yáñez
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago 7500000, Chile;
| | - Alfredo Pereira
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Bellavista 7, Santiago 8420524, Chile
| | - Daniel Bustos
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile
| |
Collapse
|
14
|
Zhang X, Xiong Z, He Y, Zheng N, Zhao S, Wang J. Epiberberine: a potential rumen microbial urease inhibitor to reduce ammonia release screened by targeting UreG. Appl Microbiol Biotechnol 2024; 108:289. [PMID: 38587649 PMCID: PMC11001712 DOI: 10.1007/s00253-024-13131-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
Rumen microbial urease inhibitors have been proposed for regulating nitrogen emission and improving nitrogen utilization efficiency in ruminant livestock industry. However, studies on plant-derived natural inhibitors of rumen microbial urease are limited. Urease accessory protein UreG, plays a crucial role in facilitating urease maturation, is a new target for design of urease inhibitor. The objective of this study was to select the potential effective inhibitor of rumen microbial urease from major protoberberine alkaloids in Rhizoma Coptidis by targeting UreG. Our results showed that berberine chloride and epiberberine exerted superior inhibition potential than other alkaloids based on GTPase activity study of UreG. Berberine chloride inhibition of UreG was mixed type, while inhibition kinetics type of epiberberine was uncompetitive. Furthermore, epiberberine was found to be more effective than berberine chloride in inhibiting the combination of nickel towards UreG and inducing changes in the second structure of UreG. Molecular modeling provided the rational structural basis for the higher inhibition potential of epiberberine, amino acid residues in G1 motif and G3 motif of UreG formed interactions with D ring of berberine chloride, while interacted with A ring and D ring of epiberberine. We further demonstrated the efficacy of epiberberine in the ruminal microbial fermentation with low ammonia release and urea degradation. In conclusion, our study clearly indicates that epiberberine is a promising candidate as a safe and effective inhibitor of rumen microbial urease and provides an optimal strategy and suitable feed additive for regulating nitrogen excretion in ruminants in the future. KEY POINTS: • Epiberberine is the most effective inhibitor of rumen urease from Rhizoma Coptidis. • Urease accessory protein UreG is an effective target for design of urease inhibitor. • Epiberberine may be used as natural feed additive to reducing NH3 release in ruminants.
Collapse
Affiliation(s)
- Xiaoyin Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zhanbo Xiong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yue He
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
15
|
Acar M, Tatini D, Budroni MA, Ninham BW, Rustici M, Rossi F, Lo Nostro P. Specific anion effects on urease activity: A Hofmeister study. Colloids Surf B Biointerfaces 2024; 236:113789. [PMID: 38367291 DOI: 10.1016/j.colsurfb.2024.113789] [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: 12/19/2023] [Revised: 01/24/2024] [Accepted: 02/04/2024] [Indexed: 02/19/2024]
Abstract
The effects of a range of electrolytes on the hydrolysis of urea by the enzyme urease is explored. The autocatalytic behavior of urease in unbuffered solutions and its pH clock reactions are studied. The concentration dependence of the experimental variables is analyzed in terms of specific ion-enzyme interactions and hydration. The results offer insights into the molecular mechanisms of the enzyme, and on the nature of its interactions with the electrolytes. We found that urease can tolerate mild electrolytes in its environment, while it is strongly inhibited by both strong kosmotropic and strong chaotropic anions. This study may cast light on an alternative therapy for Helicobacter pylori infections and contribute to the design of innovative materials and provide new approaches for the modulation of the enzymatic activity.
Collapse
Affiliation(s)
- Mert Acar
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino, Firenze 50019, Italy
| | - Duccio Tatini
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino, Firenze 50019, Italy
| | - Marcello A Budroni
- Department of Chemistry and Pharmacy, University of Sassari, Sassari 07100, Italy
| | - Barry W Ninham
- Department of Applied Mathematics, Research School of Physical Sciences and Engineering, Australian National University, Canberra, ACT 0200, Australia
| | - Mauro Rustici
- Department of Chemistry and Pharmacy, University of Sassari, Sassari 07100, Italy
| | - Federico Rossi
- Department of Earth, Environmental and Physical Sciences-DEEP Sciences, University of Siena, Italy
| | - Pierandrea Lo Nostro
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Sesto Fiorentino, Firenze 50019, Italy.
| |
Collapse
|
16
|
Valenzuela-Hormazabal P, Sepúlveda RV, Alegría-Arcos M, Valdés-Muñoz E, Rojas-Pérez V, González-Bonet I, Suardíaz R, Galarza C, Morales N, Leddermann V, Castro RI, Benso B, Urra G, Hernández-Rodríguez EW, Bustos D. Unveiling Novel Urease Inhibitors for Helicobacter pylori: A Multi-Methodological Approach from Virtual Screening and ADME to Molecular Dynamics Simulations. Int J Mol Sci 2024; 25:1968. [PMID: 38396647 PMCID: PMC10888695 DOI: 10.3390/ijms25041968] [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: 12/16/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Helicobacter pylori (Hp) infections pose a global health challenge demanding innovative therapeutic strategies by which to eradicate them. Urease, a key Hp virulence factor hydrolyzes urea, facilitating bacterial survival in the acidic gastric environment. In this study, a multi-methodological approach combining pharmacophore- and structure-based virtual screening, molecular dynamics simulations, and MM-GBSA calculations was employed to identify novel inhibitors for Hp urease (HpU). A refined dataset of 8,271,505 small molecules from the ZINC15 database underwent pharmacokinetic and physicochemical filtering, resulting in 16% of compounds for pharmacophore-based virtual screening. Molecular docking simulations were performed in successive stages, utilizing HTVS, SP, and XP algorithms. Subsequent energetic re-scoring with MM-GBSA identified promising candidates interacting with distinct urease variants. Lys219, a residue critical for urea catalysis at the urease binding site, can manifest in two forms, neutral (LYN) or carbamylated (KCX). Notably, the evaluated molecules demonstrated different interaction and energetic patterns in both protein variants. Further evaluation through ADMET predictions highlighted compounds with favorable pharmacological profiles, leading to the identification of 15 candidates. Molecular dynamics simulations revealed comparable structural stability to the control DJM, with candidates 5, 8 and 12 (CA5, CA8, and CA12, respectively) exhibiting the lowest binding free energies. These inhibitors suggest a chelating capacity that is crucial for urease inhibition. The analysis underscores the potential of CA5, CA8, and CA12 as novel HpU inhibitors. Finally, we compare our candidates with the chemical space of urease inhibitors finding physicochemical similarities with potent agents such as thiourea.
Collapse
Affiliation(s)
- Paulina Valenzuela-Hormazabal
- Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile;
| | - Romina V. Sepúlveda
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Av. República 330, Santiago 8370146, Chile;
| | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago 7500000, Chile;
| | - Elizabeth Valdés-Muñoz
- Doctorado en Biotecnología Traslacional, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca 3480094, Chile; (E.V.-M.); (V.R.-P.)
| | - Víctor Rojas-Pérez
- Doctorado en Biotecnología Traslacional, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Talca 3480094, Chile; (E.V.-M.); (V.R.-P.)
| | - Ileana González-Bonet
- Biomedical Research Labs, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile;
| | - Reynier Suardíaz
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Christian Galarza
- Departamento de Matemáticas, Facultad de Ciencias Naturales y Matemáticas, Escuela Superior Politécnica del Litoral, Guayaquil 090112, Ecuador;
| | - Natalia Morales
- Magíster en Ciencias de la Computación, Universidad Católica del Maule, Talca 3460000, Chile; (N.M.); (V.L.)
| | - Verónica Leddermann
- Magíster en Ciencias de la Computación, Universidad Católica del Maule, Talca 3460000, Chile; (N.M.); (V.L.)
| | - Ricardo I. Castro
- Multidisciplinary Agroindustry Research Laboratory, Instituto de Ciencias Aplicadas, Facultad de Arquitectura, Construcción y Medio Ambiente, Universidad Autónoma de Chile, Cinco Pte. N°1670, Talca 3467987, Chile;
| | - Bruna Benso
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile;
| | - Gabriela Urra
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile;
| | - Erix W. Hernández-Rodríguez
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile;
- Unidad de Bioinformática Clínica, Centro Oncológico, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile
| | - Daniel Bustos
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca 3480094, Chile;
| |
Collapse
|
17
|
Gao X, Zhang S, Wang P, Jaroniec M, Zheng Y, Qiao SZ. Urea catalytic oxidation for energy and environmental applications. Chem Soc Rev 2024; 53:1552-1591. [PMID: 38168798 DOI: 10.1039/d3cs00963g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Urea is one of the most essential reactive nitrogen species in the nitrogen cycle and plays an indispensable role in the water-energy-food nexus. However, untreated urea or urine wastewater causes severe environmental pollution and threatens human health. Electrocatalytic and photo(electro)catalytic urea oxidation technologies under mild conditions have become promising methods for energy recovery and environmental remediation. An in-depth understanding of the reaction mechanisms of the urea oxidation reaction (UOR) is important to design efficient electrocatalysts/photo(electro)catalysts for these technologies. This review provides a critical appraisal of the recent advances in the UOR by means of both electrocatalysis and photo(electro)catalysis, aiming to comprehensively assess this emerging field from fundamentals and materials, to practical applications. The emphasis of this review is on the design and development strategies for electrocatalysts/photo(electro)catalysts based on reaction pathways. Meanwhile, the UOR in natural urine is discussed, focusing on the influence of impurity ions. A particular emphasis is placed on the application of the UOR in energy and environmental fields, such as hydrogen production by urea electrolysis, urea fuel cells, and urea/urine wastewater remediation. Finally, future directions, prospects, and remaining challenges are discussed for this emerging research field. This critical review significantly increases the understanding of current progress in urea conversion and the development of a sustainable nitrogen economy.
Collapse
Affiliation(s)
- Xintong Gao
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Shuai Zhang
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Pengtang Wang
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Mietek Jaroniec
- Department of Chemistry and Biochemistry & Advanced Materials and Liquid Crystal Institute, Kent State University, Kent, OH 44242, USA
| | - Yao Zheng
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Shi-Zhang Qiao
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
18
|
Khan M, Nizamani A, Shah L, Ullah I, Waqas M, Halim SA, Ataya FS, Elgazzar AM, Batiha GES, Khan A, Al-Harrasi A. Utilizing the drug repurposing strategy on current drugs: new leads for peptic ulcers via biochemical and biomolecular dynamics studies. J Biomol Struct Dyn 2024:1-14. [PMID: 38225797 DOI: 10.1080/07391102.2024.2302926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/02/2024] [Indexed: 01/17/2024]
Abstract
The hyperactivity of urease enzymes plays a crucial role in the development of hepatic coma, hepatic encephalopathy, urolithiasis, gastric and peptic ulcers. Additionally, these enzymes adversely impact the soil's nitrogen efficiency for crop production. In the current study 100 known drugs were tested against Jack Bean urease and Proteus mirabilis urease and identified three inhibitors i.e. terbutaline (compound 1), Ketoprofen (compound 2) and norepinephrine bitartrate (compound 3). As a result, these compounds showed excellent inhibition against Jack Bean urease i.e. (IC50 = 2.1-11.3 µM), and Proteus mirabilis urease (4.8-11.9 µM). Moreover, in silico studies demonstrate maximum interactions of compounds in the enzyme's active site. Furthermore, intermolecular interactions between compounds and enzyme atoms were examined using STD-NMR spectrophotometry. In parallel, molecular dynamics simulation was carried out to study compounds dynamic behavior within the urease binding region. Urease remained stable during most of the simulation time and ligands were bound in the protein active pocket as observed from the Root mean square deviation (RMSD) and ligand RMSD analyses. Furthermore, these compounds display interactions with the crucial residues, including His492 and Asp633, in 100 ns simulations. In the binding energy analysis, norepinephrine bitartrate exhibited the highest binding energy (-76.32 kcal/mol) followed by Ketoprofen (-65.56 kcal/mol) and terbutaline (-62.15 kcal/mol), as compared to acetohydroxamic acid (-52.86 kcal/mol). The current findings highlight the potential of drug repurposing as an effective approach for identifying novel anti-urease compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Majid Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
- Department of Biochemistry, University of Malakand, Totakan, Pakistan
| | - Arsalan Nizamani
- Muhammad Medical College, Ibn-e-Sina University, Mirpurkhas, Sindh, Pakistan
| | - Luqman Shah
- Department of Biochemistry, Hazara University Mansehra, Mansehra,Pakistan
| | - Imran Ullah
- Department of Biochemistry, Hazara University Mansehra, Mansehra,Pakistan
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Farid Shokry Ataya
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed M Elgazzar
- Department of Veterinary Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Sultanate of Oman
| |
Collapse
|
19
|
Szczerbiec D, Bednarska-Szczepaniak K, Torzewska A. Antibacterial properties and urease suppression ability of Lactobacillus inhibit the development of infectious urinary stones caused by Proteus mirabilis. Sci Rep 2024; 14:943. [PMID: 38200115 PMCID: PMC10781950 DOI: 10.1038/s41598-024-51323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Infectious urolithiasis is a type of urolithiasis, that is caused by infections of the urinary tract by bacteria producing urease such as Proteus mirabilis. Lactobacillus spp. have an antagonistic effect against many pathogens by secreting molecules, including organic acids. The aim of the study was to determine the impact of Lactobacillus strains isolated from human urine on crystallization of urine components caused by P. mirabilis by measuring bacterial viability (CFU/mL), pH, ammonia release, concentration of crystallized salts and by observing crystals by phase contrast microscopy. Moreover, the effect of lactic acid on the activity of urease was examined by the kinetic method and in silico study. In the presence of selected Lactobacillus strains, the crystallization process was inhibited. The results indicate that one of the mechanisms of this action was the antibacterial effect of Lactobacillus, especially in the presence of L. gasseri, where ten times less P. mirabilis bacteria was observed, compared to the control. It was also demonstrated that lactic acid inhibited urease activity by a competitive mechanism and had a higher binding affinity to the enzyme than urea. These results demonstrate that Lactobacillus and lactic acid have a great impact on the urinary stones development, which in the future may help to support the treatment of this health problem.
Collapse
Affiliation(s)
- Dominika Szczerbiec
- Department of Biology of Bacteria, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland
| | | | - Agnieszka Torzewska
- Department of Biology of Bacteria, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland.
| |
Collapse
|
20
|
Sultana A, Wahab A, Fareed G, Rafiq H, Khan KM, Lateef M, Fareed N, Hussain S, Sherwani SK. Identification of potential drug candidates to treat gastritis and associated oxidative stress based on some novel 2-aryl-1 H-naphtho[2,3- d]imidazole: synthesis, in vitro and in silico analysis. RSC Adv 2024; 14:529-537. [PMID: 38173575 PMCID: PMC10759213 DOI: 10.1039/d3ra07412a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
To identify potential scaffolds to treat gastritis and oxidative stress, 2-aryl-1H-naphtho[2,3-d]imidazole derivatives (1-15) were synthesized. The synthesis was conveniently carried out by condensing 2,3-diaminonaphthalene with variously substituted aldehydes to yield 15 new 2-aryl-1H-naphtho[2,3-d]imidazole derivatives. Structures of all synthesized compounds were elucidated using MS and NMR spectroscopic techniques. Compounds containing an imidazole moiety have continued to spark interest in the field of medicinal chemistry due to their unique properties. In continuation of this statement, to further explore the biological potential of these types of compounds, newly synthesized imidazole derivatives were evaluated for their inhibitory potential against urease and antioxidant activities. Compounds 4 and 11 were identified as the most potent urease inhibitors in the series, with IC50 values of 34.2 ± 0.72 and 42.43 ± 0.65 μM, respectively. Compounds 1, 3, 6, 11, and 15, with EC50 values in the range of 37-75 μg ml-1, showed significant antioxidant activity. Molecular docking studies of the selected synthesized compounds 3, 4, 9, and 11 were also performed to determine their binding interaction with the jack bean urease. Through docking studies, it was revealed that all the compounds that showed good inhibitory potential against urease fit well within the protein's binding pocket. Furthermore, ADME analysis was carried out to explore the drug-likeness properties of the compounds. The findings of the present work revealed that compounds 4 and 11 could be better options to treat gastritis and associated oxidative stress.
Collapse
Affiliation(s)
- Amina Sultana
- Department of Chemistry, Federal Urdu University of Arts, Science and Technology Gulshan-e-Iqbal Karachi Pakistan
| | - Aneela Wahab
- Department of Chemistry, Federal Urdu University of Arts, Science and Technology Gulshan-e-Iqbal Karachi Pakistan
| | - Ghulam Fareed
- Pharmaceutical Research Centre PCSIR Laboratories Complex Karachi Pakistan
| | - Hamna Rafiq
- Pharmaceutical Research Centre PCSIR Laboratories Complex Karachi Pakistan
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Centre for Chemical and Biological Sciences, University of Karachi Pakistan
| | - Mehreen Lateef
- Department of Biochemistry, Bahria University Medical and Dental College, Bahria University Karachi Pakistan
| | - Nazia Fareed
- Department of Chemistry, Federal Urdu University of Arts, Science and Technology Gulshan-e-Iqbal Karachi Pakistan
| | - Shafqat Hussain
- Department of Chemistry, University of Baltistan Skardu Gilgit-Baltistan 1600 Pakistan
| | - Sikander Khan Sherwani
- Department of Microbiology, Federal Urdu University of Arts, Science and Technology Gulshan-e-Iqbal Karachi Pakistan
| |
Collapse
|
21
|
Afkhamipour M, Kaviani F, Dalali S, Piri-Gharaghie T, Doosti A. Potential Gastric Cancer Immunotherapy: Stimulating the Immune System with Helicobacter pylori pIRES2-DsRed-Express- ureF DNA Vaccines. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0004. [PMID: 38346161 DOI: 10.2478/aite-2024-0004] [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: 02/26/2023] [Accepted: 09/20/2023] [Indexed: 02/15/2024]
Abstract
Most gastric cancers (GC) are thought to be caused by Helicobacter pylori (H. pylori) infections. However, there is mounting evidence that GC patients with positive H. pylori status have improved prognoses. The H. pylori-induced cellular immune reaction may inhibit cancer. In this study, BALB/c mice were immunized using recombinant plasmids that encode the ureF gene of H. pylori. Purified functional splenic CD3+ T lymphocytes are used to study the anticancer effects in vitro and in vivo. The immunological state of GC patients with ongoing H. pylori infection is mimicked by the H. pylori DNA vaccines, which cause a change in the reaction from Th1 to Th2. Human GC cells grow more slowly when stimulated CD3+ T lymphocytes are used as adoptive infusions because they reduce GC xenograft development in vivo. The more excellent ratios of infiltrating CD8+/CD4+ T cells, the decreased invasion of regulatory FOXP3+ Treg lymphocytes, and the increased apoptosis brought on by Caspase9/Caspase-3 overexpression and Survivin downregulation may all contribute to the consequences. Our findings suggest that in people with advanced GC, H. pylori pIRES2-DsRed-Express-ureF DNA vaccines may have immunotherapeutic utility.
Collapse
Affiliation(s)
- Mahsa Afkhamipour
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fatemeh Kaviani
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Samaneh Dalali
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Tohid Piri-Gharaghie
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
- Biotechnology Research Center, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Abbas Doosti
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
22
|
Montazer MN, Asadi M, Moradkhani F, Omrany ZB, Mahdavi M, Amanlou M. Design, synthesis, and biological studies of the new cysteine-N-arylacetamide derivatives as a potent urease inhibitor. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:305-315. [PMID: 37436497 DOI: 10.1007/s00210-023-02596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 06/20/2023] [Indexed: 07/13/2023]
Abstract
Inhibition of Helicobacter pylori urease is an effective method in the treatment of several gastrointestinal diseases in humans. This bacterium plays an important role in the pathogenesis of gastritis and peptic ulceration. Considering the presence of cysteine and N-arylacetamide derivatives in potent urease inhibitors, here, we designed hybrid derivatives of these pharmacophores. Therefore, cysteine-N-arylacetamide derivatives 5a-l were synthesized through simple nucleophilic reactions with good yield. In vitro urease inhibitory activity assay of these compounds demonstrated that all newly synthesized compounds exhibited high inhibitory activity (IC50 values = 0.35-5.83 μM) when compared with standard drugs (thiourea: IC50 = 21.1 ± 0.11 μM and hydroxyurea: IC50 = 100.0 ± 0.01 μM). Representatively, compound 5e with IC50 = 0.35 μM was 60 times more potent than strong urease inhibitor thiourea. Enzyme kinetic study of this compound revealed that compound 5e is a competitive urease inhibitor. Moreover, a docking study of compound 5e was performed to explore crucial interactions at the urease active site. This study revealed that compound 5e is capable to inhibit urease by interactions with two crucial residues at the active site: Ni and CME592. Furthermore, a molecular dynamics study confirmed the stability of the 5e-urease complex and Ni chelating properties of this compound. It should be considered that, in the following study, the focus was placed on jack bean urease instead of H. pylori urease, and this was acknowledged as a limitation.
Collapse
Affiliation(s)
- Mohammad Nazari Montazer
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Asadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradkhani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zinat Bahrampour Omrany
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
da Fonseca AB, Santos C, Nunes APP, Oliveira DP, de Melo MEA, Takayama T, Mansur BL, de Jesus Fernandes T, do Carmo Alexandrino G, Dias MAN, Guelfi D. Urease inhibitors technologies as strategy to mitigate agricultural ammonia emissions and enhance the use efficiency of urea-based fertilizers. Sci Rep 2023; 13:22739. [PMID: 38123658 PMCID: PMC10733344 DOI: 10.1038/s41598-023-50061-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Experiments were conducted to evaluate the stability and degradation of NBPT under storage conditions and to quantify urease activity, ammonia losses by volatilization, and agronomic efficiency of urea treated with different urease inhibitors, measured in the field. Experiments included urea treated with 530 mg NBPT kg-1 (UNBPT) in contact with six P-sources (monoammonium phosphate-MAP; single superphosphate; triple superphosphate; P-Agrocote; P-Phusion; P-Policote), with two P-concentrations (30; 70%); the monitoring four N-technologies (SoILC; Limus; Nitrain; Anvol); and the application of conventional urea (UGRAN) or urea treated with urease inhibitors as topdressing in three maize fields, at three N rates. It is concluded that: the mixture of UNBPT and P-fertilizers is incompatible. When MAP granules were coated to control P-release (P-Agrocote), the degradation of NBPT was moderate (approximately 400 mg kg-1 at the end of the storage test). SoILC and Limus solvent technologies extended the NBPT half-life by up to 3.7 and 4.7 months, respectively. Under field, each inhibition technology reduced urease activity, and lowered the intensity of ammonia emission compared to UGRAN by 50-62%. Our results show that the concentration of NBPT is reduced by up to 53.7% for mixing with phosphates. In addition, even with coatings, the storage of mixtures of urea with NBPT and phosphates should be for a time that does not reduce the efficiency of the inhibitor after application, and this time under laboratory conditions was 168 h. The reduction of NBPT concentration in urea is reduced even in isolated storage, our results showed that the half-life time is variable according to the formulation used, being 4.7, 3.7, 2.8 and 2.7 days for Limus, SoILC, Nitrain and Anvol, respectively. The results of these NBPT formulations in the field showed that the average losses by volatilization in the three areas were: 15%, 16%, 17%, 19% and 39% of the N applied, for SoILC, Anvol, Nitrain, Limus and urea, respectively. The rate of nitrogen application affected all agronomic variables, with varied effects in Ingaí. Even without N, yields were higher than 9200 kg ha-1 of grains. The increase in nitrogen rates resulted in linear increases in production and N removal in Luminárias and Ingaí, but in Lavras, production decreased above 95.6 kg ha-1 of N. The highest production in Lavras (13,772 kg ha-1 of grains) occurred with 100 kg ha-1 of N. The application of Anvol reduced the removal of N in Ingaí.
Collapse
Affiliation(s)
- Adrianne Braga da Fonseca
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | - César Santos
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | - Ana Paula Pereira Nunes
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | - Damiany Pádua Oliveira
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | - Maria Elisa Araújo de Melo
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | - Thalita Takayama
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | - Bethânia Leite Mansur
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | | | - Gilson do Carmo Alexandrino
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil
| | | | - Douglas Guelfi
- Laboratory of Fertilizers Technologies-INNOVA FERT, Department of Soil Science, Federal University of Lavras-UFLA, P.O. Box 3037, Lavras, MG, 37203-202, Brazil.
| |
Collapse
|
24
|
Governa P, Romagnoli G, Albanese P, Rossi F, Manetti F, Biagi M. Effect of in vitro simulated digestion on the anti- Helicobacter Pylori activity of different Propolis extracts. J Enzyme Inhib Med Chem 2023; 38:2183810. [PMID: 36916299 PMCID: PMC10026752 DOI: 10.1080/14756366.2023.2183810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
Helicobacter pylori (HP) is among the most common pathogens causing infection in humans worldwide. Oxidative stress and gastric inflammation are involved in the progression of HP-related gastric diseases, and they can be targeted by integrating conventional antibiotic treatment with polyphenol-enriched natural products. In this work, we characterised three different propolis extracts and evaluated their stability under in vitro simulated gastric digestion, compared to their main constituents alone. The extract with the highest stability to digestion (namely, the dark propolis extract, DPE) showed a minimum bactericidal concentration (MBC) lower than 1 mg/mL on HP strains with different virulence factors. Finally, since urease is one of the virulence factors contributing to the establishment of a microenvironment that promotes HP infection, we evaluated the possible inhibition of this enzyme by using molecular docking simulations and in vitro colorimetric assay, showing that galangin and pinocembrin may be involved in this activity.
Collapse
Affiliation(s)
- Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulia Romagnoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Paola Albanese
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| | - Federico Rossi
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Marco Biagi
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| |
Collapse
|
25
|
Liu S, Guan L, Peng C, Cheng Y, Cheng H, Wang F, Ma M, Zheng R, Ji Z, Cui P, Ren Y, Li L, Shi C, Wang J, Huang X, Cai X, Qu D, Zhang H, Mao Z, Liu H, Wang P, Sha W, Yang H, Wang L, Ge B. Mycobacterium tuberculosis suppresses host DNA repair to boost its intracellular survival. Cell Host Microbe 2023; 31:1820-1836.e10. [PMID: 37848028 DOI: 10.1016/j.chom.2023.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/19/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Mycobacterium tuberculosis (Mtb) triggers distinct changes in macrophages, resulting in the formation of lipid droplets that serve as a nutrient source. We discover that Mtb promotes lipid droplets by inhibiting DNA repair responses, resulting in the activation of the type-I IFN pathway and scavenger receptor-A1 (SR-A1)-mediated lipid droplet formation. Bacterial urease C (UreC, Rv1850) inhibits host DNA repair by interacting with RuvB-like protein 2 (RUVBL2) and impeding the formation of the RUVBL1-RUVBL2-RAD51 DNA repair complex. The suppression of this repair pathway increases the abundance of micronuclei that trigger the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway and subsequent interferon-β (IFN-β) production. UreC-mediated activation of the IFN-β pathway upregulates the expression of SR-A1 to form lipid droplets that facilitate Mtb replication. UreC inhibition via a urease inhibitor impaired Mtb growth within macrophages and in vivo. Thus, our findings identify mechanisms by which Mtb triggers a cascade of cellular events that establish a nutrient-rich replicative niche.
Collapse
Affiliation(s)
- Shanshan Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Liru Guan
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Cheng Peng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Yuanna Cheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Hongyu Cheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Fei Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Mingtong Ma
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Ruijuan Zheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Zhe Ji
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Pengfei Cui
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Yefei Ren
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Liru Li
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Chenyue Shi
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Jie Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Xiaochen Huang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Xia Cai
- Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Di Qu
- Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Haiping Zhang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Zhiyong Mao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, P.R. China
| | - Haipeng Liu
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Peng Wang
- Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Wei Sha
- Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Hua Yang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China.
| | - Lin Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China.
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China; Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China; Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China.
| |
Collapse
|
26
|
Akhtar M, Gul S, Shamim S, Naeem S, Khan A. Moxifloxacin Amide Analogs as Antibacterial and Antifungal Agents: Synthesis, Characterization, and Enzyme Inhibition Studies. Pharm Chem J 2023; 57:1008-1017. [DOI: 10.1007/s11094-023-02978-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Indexed: 07/04/2024]
|
27
|
Uddin J, Ullah S, Halim SA, Waqas M, Ibrar A, Khan I, Bin Muhsinah A, Khan A, Al-Harrasi A. Triazolothiadiazoles and Triazolothiadiazines as New and Potent Urease Inhibitors: Insights from In Vitro Assay, Kinetics Data, and In Silico Assessment. ACS OMEGA 2023; 8:31890-31898. [PMID: 37692208 PMCID: PMC10483676 DOI: 10.1021/acsomega.3c03546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023]
Abstract
Hyperactivity of the urease enzyme induces the pathogenesis of peptic ulcers and gastritis. The identification of new urease inhibitors can reduce the activity of urease. Therefore, in the current study, we have evaluated 28 analogues of triazolothiadiazole and triazolothiadiazine heteroaromatics for their in vitro urease inhibitory efficacy. All the tested compounds displayed a remarkable inhibitory potential ranging from 3.33 to 46.83 μM. Among them, compounds 5k and 5e emerged as lead inhibitors with IC50 values of 3.33 ± 0.11 and 3.51 ± 0.49 μM, respectively. The potent inhibitory potential of these compounds was ∼6.5-fold higher than that of the marketed drug thiourea (IC50 = 22.45 ± 0.30 μM). The mechanistic insights from kinetics experiments of the highest potent inhibitors (4g, 5e, and 5k) revealed a competitive type of inhibition with ki values 2.25 ± 0.0028, 3.11 ± 0.0031, and 3.62 ± 0.0034 μM, respectively. In silico modeling was performed to investigate the binding interactions of potent inhibitors with the enzyme active site residues, which strongly supported our experimental results. Furthermore, ADME analysis also showed good druglikeness properties demonstrating the potential of these compounds to be developed as lead antiurease agents.
Collapse
Affiliation(s)
- Jalal Uddin
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Kingdom
of Saudi Arabia
| | - Saeed Ullah
- Natural
and Medical Sciences Research Center, University
of Nizwa, Birkat-ul-Mouz 616 Nizwa, Sultanate of Oman
| | - Sobia Ahsan Halim
- Natural
and Medical Sciences Research Center, University
of Nizwa, Birkat-ul-Mouz 616 Nizwa, Sultanate of Oman
| | - Muhammad Waqas
- Natural
and Medical Sciences Research Center, University
of Nizwa, Birkat-ul-Mouz 616 Nizwa, Sultanate of Oman
| | - Aliya Ibrar
- Department
of Chemistry, Faculty of Natural Sciences, The University of Haripur, Haripur, KPK 22620, Pakistan
| | - Imtiaz Khan
- Department
of Chemistry and Manchester Institute of Biotechnology,The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Abdullatif Bin Muhsinah
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Kingdom
of Saudi Arabia
| | - Ajmal Khan
- Natural
and Medical Sciences Research Center, University
of Nizwa, Birkat-ul-Mouz 616 Nizwa, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural
and Medical Sciences Research Center, University
of Nizwa, Birkat-ul-Mouz 616 Nizwa, Sultanate of Oman
| |
Collapse
|
28
|
Sepehri S, Khedmati M. An overview of the privileged synthetic heterocycles as urease enzyme inhibitors: Structure-activity relationship. Arch Pharm (Weinheim) 2023; 356:e2300252. [PMID: 37401193 DOI: 10.1002/ardp.202300252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 07/05/2023]
Abstract
Urease is a metalloenzyme including two Ni2+ ions, found in some plants, bacteria, fungi, microorganisms, invertebrate animals, and animal tissues. Urease acts as a significant virulence factor, mainly in catheter blockage and infective urolithiasis as well as in the pathogenesis of gastric infection. Therefore, studies on urease lead to novel synthetic inhibitors. In this review, the synthesis and antiurease activities of a collection of privileged synthetic heterocycles such as (thio)barbiturate, (thio)urea, dihydropyrimidine, and triazol derivatives were described and discussed according to structure-activity relationship findings in search of the best moieties and substituents that are answerable for encouraging the desired activity even more potent than the standard. It was found that linking substituted phenyl and benzyl rings to the heterocycles led to potent urease inhibitors.
Collapse
Affiliation(s)
- Saghi Sepehri
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mina Khedmati
- Students Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
29
|
Fatima M, Aslam S, Zafar AM, Irfan A, Khan MA, Ashraf M, Faisal S, Noreen S, Shazly GA, Shah BR, Bin Jardan YA. Exploring the Synthetic Chemistry of Phenyl-3-(5-aryl-2-furyl)- 2-propen-1-ones as Urease Inhibitors: Mechanistic Approach through Urease Inhibition, Molecular Docking and Structure-Activity Relationship. Biomedicines 2023; 11:2428. [PMID: 37760869 PMCID: PMC10525509 DOI: 10.3390/biomedicines11092428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Furan chalcone scaffolds belong to the most privileged and promising oxygen-containing heterocyclic class of compounds, which have a wide spectrum of therapeutic applications in the field of pharmaceutics, pharmacology, and medicinal chemistry. This research described the synthesis of a series of twelve novel and seven reported furan chalcone (conventional synthetic approach) analogues 4a-s through the application of microwave-assisted synthetic methodology and evaluated for therapeutic inhibition potential against bacterial urease enzyme. In the first step, a series of nineteen substituted 5-aryl-2-furan-2-carbaldehyde derivatives 3a-s were achieved in moderate to good yields (40-70%). These substituted 5-aryl-2-furan-2-carbaldehyde derivatives 3a-s were condensed with acetophenone via Claisen-Schmidt condensation to furnish 19 substituted furan chalcone scaffolds 4a-s in excellent yields (85-92%) in microwave-assisted synthetic approach, while in conventional methodology, these furan chalcone 4a-s were furnished in good yield (65-90%). Furan chalcone structural motifs 4a-s were characterized through elemental analysis and spectroscopic techniques. These nineteen (19)-afforded furan chalcones 4a-s were screened for urease inhibitory chemotherapeutic efficacy and most of the furan chalcones displayed promising urease inhibition activity. The most active urease inhibitors were 1-phenyl-3-[5-(2',5'-dichlorophenyl)-2-furyl]-2-propen-1-one 4h with an IC50 value of 16.13 ± 2.45 μM, and 1-phenyl- 3-[5-(2'-chlorophenyl)-2-furyl] -2-propen-1-one 4s with an IC50 value of 18.75 ± 0.85 μM in comparison with reference drug thiourea (IC50 = 21.25 ± 0.15 μM). These furan chalcone derivatives 4h and 4s are more efficient urease inhibitors than reference drug thiourea. Structure-activity relationship (SAR) revealed that the 2,5-dichloro 4h and 2-chloro 4s moiety containing furan chalcone derivatives may be considered as potential lead reagents for urease inhibition. The in silico molecular docking study results are in agreement with the experimental biological findings. The results of this study may be helpful in the future drug discovery and designing of novel efficient urease inhibitory agents from this biologically active class of furan chalcones.
Collapse
Affiliation(s)
- Miraj Fatima
- Department of Chemistry, The Women University, Multan 66000, Pakistan
| | - Samina Aslam
- Department of Chemistry, The Women University, Multan 66000, Pakistan
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Ansa Madeeha Zafar
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Chemistry, Government Sadiq Women University, Bahawalpur 63100, Pakistan
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Misbahul Ain Khan
- Department of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Muhammad Ashraf
- Department of Biotechnology and Biochemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Shah Faisal
- Department of Chemistry, Islamia College University Peshawar, Peshawar 25120, Pakistan
| | - Sobia Noreen
- Institute of Chemistry, University of Sargodha, Sargodha 40100, Pakistan
| | - Gamal A. Shazly
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Bakht Ramin Shah
- Skin Barrier Research Group, Faculty of Pharmacy in Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
30
|
Zahra FT, Saeed A, Ahmed A, Ismail H, Ijaz MU, Albericio F. Synthesis of amantadine clubbed N-aryl amino thiazoles as potent urease, α-amylase & α-glucosidase inhibitors, kinetic and molecular docking studies. RSC Adv 2023; 13:24988-25001. [PMID: 37614781 PMCID: PMC10442672 DOI: 10.1039/d3ra05330j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023] Open
Abstract
A series of ten novel compounds were synthesized by incorporating a 1,3 thiazole core into amantadine and their structures were validated using different analytical and spectral methods such as FTIR, EI-MS, 1H NMR, and 13C NMR. The antibacterial and enzyme inhibitory properties of these newly synthesized compounds were evaluated. Remarkably, the compounds exhibited significant antibacterial activity against Escherichia coli and Bacillus subtilis. Additionally, the in vitro inhibitory activities of the synthesized compounds, against α-amylase, α-glucosidase, and urease were investigated. Among the tested compounds, compound 6d demonstrated potent and selective inhibition of α-amylase IC50 = 97.37 ± 1.52 μM, while acarbose was used as positive control and exhibited IC50 = 5.17 ± 0.25 μM. Compound 6d and 6e exhibited prominent inhibition against α-glucosidase IC50 = 38.73 ± 0.80 μM and 41.63 ± 0.26 μM respectively. Furthermore, compound 6d inhibited urease with exceptional efficacy IC50 = 32.76 μM, while positive control thiourea showed more prominent activity having IC50 = 1.334 μM. Molecular docking studies disclosed the binding mechanism and affinity of these new inhibitors within the binding sites of various amino acids. To investigate the association between molecular structural characteristics and inhibitory actions of synthesized derivatives, preliminary structure-activity relationship (SAR) studies were performed. These findings indicated that compounds 6a, 6c, 6d and 6e are potential candidates for hit-to-lead follow-up in the drug-discovery process for treating diabetes and hyperglycemia.
Collapse
Affiliation(s)
- Fatima Tuz Zahra
- Department of Chemistry, Quaid-i-Azam University 45320 Islamabad Pakistan +92-51-9064-2241 +92-51-9064-2128
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University 45320 Islamabad Pakistan +92-51-9064-2241 +92-51-9064-2128
| | - Atteeque Ahmed
- Department of Chemistry, Quaid-i-Azam University 45320 Islamabad Pakistan +92-51-9064-2241 +92-51-9064-2128
| | - Hammad Ismail
- Department of Biochemistry and Biotechnology, University of Gujrat Gujrat 50700 Pakistan
| | - Muhammad Umar Ijaz
- Department of Zoology, Wildlife and Fisheries, University of Agriculture Faisalabad 38040 Pakistan
| | - Fernando Albericio
- Peptides Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal Westville Durban 4000 South Africa
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, Department of Organic Chemistry, University of Barcelona 08028 Barcelona Spain
| |
Collapse
|
31
|
Tabor W, Katsogiannou A, Karta D, Andrianopoulou E, Berlicki Ł, Vassiliou S, Grabowiecka A. Exploration of Thiourea-Based Scaffolds for the Construction of Bacterial Ureases Inhibitors. ACS OMEGA 2023; 8:28783-28796. [PMID: 37576686 PMCID: PMC10413841 DOI: 10.1021/acsomega.3c03702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023]
Abstract
A series of 32 thiourea-based urease inhibitors were synthesized and evaluated against native bacterial enzyme and whole cells of Sporosarcina pasteurii and Proteus mirabilis strains. The proposed inhibitors represented structurally diverse thiosemicarbazones and thiocarbohydrazones, benzyl-substituted thiazolyl thioureas, 1H-pyrazole-1-carbothioamides, and dihydropirimidine-2(1H)-thiones. Kinetic characteristics with purified S. pasteurii enzyme determined low micromolar inhibitors within each structural group. (E)-2-(1-Phenylethylidene)hydrazine-1-carbothioamide 19 (Ki = 0.39 ± 0.01 μM), (E)-2-(4-methylbenzylidene)hydrazine-1-carbothioamide 16 (Ki = 0.99 ± 0.04 μM), and N'-((1E,2E)-1,3-diphenylallylidene)hydrazinecarbothiohydrazide 29 (Ki = 2.23 ± 0.19 μM) were used in modeling studies that revealed sulfur ion coordination of the active site nickel ion and hydrogen bonds between the amide group and the side chain of Asp363 and Ala366 carbonyl moiety. Whole-cell studies proved the activity of compounds in Gram-positive and Gram-negative microorganisms. Ureolysis control observed in P. mirabilis PCM 543 (e.g., IC50 = 304 ± 14 μM for 1-benzyl-3-(4-(4-hydroxyphenyl)thiazol-2-yl)thiourea 52) is a valuable achievement, as urease is recognized as a major virulence factor of this urinary tract pathogen.
Collapse
Affiliation(s)
- Wojciech Tabor
- Department
of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego
27, 50-370 Wrocław, Poland
| | - Aikaterini Katsogiannou
- Laboratory
of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Danai Karta
- Laboratory
of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Evgenia Andrianopoulou
- Laboratory
of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Łukasz Berlicki
- Department
of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego
27, 50-370 Wrocław, Poland
| | - Stamatia Vassiliou
- Laboratory
of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Agnieszka Grabowiecka
- Department
of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego
27, 50-370 Wrocław, Poland
| |
Collapse
|
32
|
Ahmad R, Khan M, Alam A, Elhenawy AA, Qadeer A, AlAsmari AF, Alharbi M, Alasmari F, Ahmad M. Synthesis, molecular structure and urease inhibitory activity of novel bis-Schiff bases of benzyl phenyl ketone: A combined theoretical and experimental approach. Saudi Pharm J 2023; 31:101688. [PMID: 37457366 PMCID: PMC10345485 DOI: 10.1016/j.jsps.2023.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Background Urease belongs to the family of amid hydrolases with two nickel atoms in their core structure. On the basis of literature survey, this research work is mainly focused on the study of bis-Schiff base derivatives of benzyl phenyl ketone nucleus. Objective Synthesis of benzyl phenyl ketone based bis-Schiff bases in search of potent urease inhibitors. Method In the current work, bis-Schiff bases were synthesized through two steps reaction by reacting benzyl phenyl ketone with excess of hydrazine hydrate in ethanol solvent in the first step to get the desired hydrazone. In last, different substituted aromatic aldehydes were refluxed in catalytic amount of acetic acid with the desired hydrazone to obtain bis-Schiff base derivatives in tremendous yields. Using various spectroscopic techniques including FTIR, HR-ESI-MS, and 1H NMR spectroscopy were used to clarify the structures of the created bis-Schiff base derivatives. Results The prepared compounds were finally screened for their in-vitro urease inhibition activity. All the synthesized derivatives (3-9) showed excellent to less inhibitory activity when compared with standard thiourea (IC50 = 21.15 ± 0.32 µM). Compounds 3 (IC50 = 22.21 ± 0.42 µM), 4 (IC50 = 26.11 ± 0.22 µM) and 6 (IC50 = 28.11 ± 0.22 µM) were found the most active urease inhibitors near to standard thiourea among the synthesized series. Similarly, compound 5 having IC50 value of 34.32 ± 0.65 µM showed significant inhibitory activity against urease enzyme. Furthermore, three compounds 7, 8, and 9 exhibited less activity with IC50 values of 45.91 ± 0.14, 47.91 ± 0.14, and 48.33 ± 0.72 µM respectively. DFT used to calculate frontier molecular orbitals including; HOMO and LUMO to indicate the charge transfer from molecule to biological transfer, and MEP map to indicate the chemically reactive zone suitable for drug action. The electron localization function (ELF), non-bonding orbitals, AIM charges are also calculated. The docking study contributed to the analysis of urease protein binding.
Collapse
Affiliation(s)
- Rashid Ahmad
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
- Department of Chemistry, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Momin Khan
- Department of Chemistry, Abdul Wali Khan University, Mardan 23200, Khyber Pakhtunkhwa, Pakistan
| | - Aftab Alam
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| | - Ahmed A. Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Abdul Qadeer
- Key Laboratory of Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Abdullah F. AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Manzoor Ahmad
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
33
|
Zhuravleva OI, Chingizova EA, Oleinikova GK, Starnovskaya SS, Antonov AS, Kirichuk NN, Menshov AS, Popov RS, Kim NY, Berdyshev DV, Chingizov AR, Kuzmich AS, Guzhova IV, Yurchenko AN, Yurchenko EA. Anthraquinone Derivatives and Other Aromatic Compounds from Marine Fungus Asteromyces cruciatus KMM 4696 and Their Effects against Staphylococcus aureus. Mar Drugs 2023; 21:431. [PMID: 37623712 PMCID: PMC10455474 DOI: 10.3390/md21080431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
New anthraquinone derivatives acruciquinones A-C (1-3), together with ten known metabolites, were isolated from the obligate marine fungus Asteromyces cruciatus KMM 4696. Acruciquinone C is the first member of anthraquinone derivatives with a 6/6/5 backbone. The structures of isolated compounds were established based on NMR and MS data. The absolute stereoconfigurations of new acruciquinones A-C were determined using ECD and quantum chemical calculations (TDDFT approach). A plausible biosynthetic pathway of the novel acruciquinone C was proposed. Compounds 1-4 and 6-13 showed a significant antimicrobial effects against Staphylococcus aureus growth, and acruciquinone A (1), dendryol B (4), coniothyrinone B (7), and ω-hydroxypachybasin (9) reduced the activity of a key staphylococcal enzyme, sortase A. Moreover, the compounds, excluding 4, inhibited urease activity. We studied the effects of anthraquinones 1, 4, 7, and 9 and coniothyrinone D (6) in an in vitro model of skin infection when HaCaT keratinocytes were cocultivated with S. aureus. Anthraquinones significantly reduce the negative impact of S. aureus on the viability, migration, and proliferation of infected HaCaT keratinocytes, and acruciquinone A (1) revealed the most pronounced effect.
Collapse
Affiliation(s)
- Olesya I. Zhuravleva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
- Institute of High Technologies and Advanced Materials, Far Eastern Federal University, 10 Ajax Bay, Russky Island, Vladivostok 690922, Russia
| | - Ekaterina A. Chingizova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Galina K. Oleinikova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Sofya S. Starnovskaya
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Alexandr S. Antonov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Natalia N. Kirichuk
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Alexander S. Menshov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Roman S. Popov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Natalya Yu. Kim
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Dmitrii V. Berdyshev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Artur R. Chingizov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Alexandra S. Kuzmich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Irina V. Guzhova
- Institute of Cytology Russian Academy of Sciences, Tikhoretskiy Ave. 4, St. Petersburg 194064, Russia;
| | - Anton N. Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| | - Ekaterina A. Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Prospect 100-Letiya Vladivostoka, 159, Vladivostok 690022, Russia; (O.I.Z.); (E.A.C.)
| |
Collapse
|
34
|
Islam M, Khan A, Khan M, Halim SA, Ullah S, Hussain J, Al-Harrasi A, Shafiq Z, Tasleem M, El-Gokha A. Synthesis and biological evaluation of 2-nitrocinnamaldehyde derived thiosemicarbazones as urease inhibitors. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
35
|
Maślanka M, Tabor W, Krzyżek P, Grabowiecka A, Berlicki Ł, Mucha A. Inhibitory activity of catecholic phosphonic and phosphinic acids against Helicobacter pylori ureolysis. Eur J Med Chem 2023; 257:115528. [PMID: 37290184 DOI: 10.1016/j.ejmech.2023.115528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023]
Abstract
Catechols have been reported to be potent covalent inhibitors of ureases, and they exhibit activity by modifying cysteine residues at the entrance to enzymatic active sites. Following these principles, we designed and synthesized novel catecholic derivatives that contained carboxylate and phosphonic/phosphinic functionalities and assumed expanded specific interactions. When studying the chemical stability of the molecules, we found that their intrinsic acidity catalyzes spontaneous esterification/hydrolysis reactions in methanol or water solutions, respectively. Regarding biological activity, the most promising compound, 2-(3,4-dihydroxyphenyl)-3-phosphonopropionic acid (15), exhibited significant anti-urease potential (Ki = 2.36 μM, Sporosarcinia pasteurii urease), which was reflected in the antiureolytic effect in live Helicobacter pylori cells at a submicromolar concentration (IC50 = 0.75 μM). As illustrated by molecular modeling, this compound was bound in the active site of urease through a set of concerted electrostatic and hydrogen bond interactions. The antiureolytic activity of catecholic phosphonic acids could be specific because these compounds were chemically inert and not cytotoxic to eukaryotic cells.
Collapse
Affiliation(s)
- Marta Maślanka
- Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| | - Wojciech Tabor
- Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| | - Paweł Krzyżek
- Department of Microbiology, Faculty of Medicine, Wrocław Medical University, Wybrzeże L. Pasteura 1, 50-367, Wrocław, Poland
| | - Agnieszka Grabowiecka
- Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| | - Łukasz Berlicki
- Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland
| | - Artur Mucha
- Department of Bioorganic Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370, Wrocław, Poland.
| |
Collapse
|
36
|
Tavares MC, Dos Santos Nascimento IJ, de Aquino TM, de Oliveira Brito T, Macedo F, Modolo LV, de Fátima Â, Santos JCC. The influence of N-alkyl chains in benzoyl-thiourea derivatives on urease inhibition: Soil studies and biophysical and theoretical investigations on the mechanism of interaction. Biophys Chem 2023; 299:107042. [PMID: 37263179 DOI: 10.1016/j.bpc.2023.107042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023]
Abstract
Ureases are enzymes produced by fungi, plants, and bacteria associated with agricultural and clinical problems. The urea hydrolysis in NH3 and CO2 leads to the loss of N-urea fertilizers in soils and changes the human stomach microenvironment, favoring the colonization of H. pylori. In this sense, it is necessary to evaluate potential enzyme inhibitors to mitigate the effects of their activities and respond to scientific and market demands to produce fertilizers with enhanced efficiency. Thus, biophysical and theoretical studies were carried out to evaluate the influence of the N-alkyl chain in benzoyl-thiourea derivatives on urease enzyme inhibition. A screening based on IC50, binding constants, and theoretical studies demonstrated that BTU1 without the N-alkyl chain (R = H) was more active than other compounds, so the magnitude of the interaction was determined as BTU1 > BTU2 > BTU3 > BTU4 > BTU5, corresponding to progressively increased chain length. Thus, BTU1 was selected for interaction and soil application essays. The binding constants (Kb) for the supramolecular urease-BTU1 complex ranged from 7.95 to 5.71 × 103 M-1 at different temperatures (22, 30, and 38 °C), indicating that the preferential forces responsible for the stabilization of the complex are hydrogen bonds and van der Waals forces (ΔH = -15.84 kJ mol-1 and ΔS = -36.61 J mol-1 K-1). Theoretical and experimental results (thermodynamics, synchronous fluorescence, and competition assay) agree and indicate that BTU1 is a mixed inhibitor. Finally, urease inhibition was evaluated in the four soil samples, where BTU1 was as efficient as NBPT (based on ANOVA two-way and Tukey test with 95% confidence), with an average inhibition of 20% of urease activity. Thus, the biophysics and theoretical studies are strategies for evaluating potential inhibitors and showed that increasing the N-alkyl chain in benzoyl-thiourea derivatives did not favor urease inhibition.
Collapse
Affiliation(s)
- Maria Célia Tavares
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil; Instituto Federal de Educação, Ciência e Tecnologia de Alagoas, Campus Batalha, AL, Brazil
| | | | | | - Tiago de Oliveira Brito
- Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | - Fernando Macedo
- Departamento de Química, Centro de Ciências Exatas, Universidade Estadual de Londrina, Londrina, PR, Brazil
| | - Luzia Valentina 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.
| | | |
Collapse
|
37
|
Wu H, Sun Q, Dong H, Qiao J, Lin Y, Yu C, Li Y. Gastroprotective action of the extract of Corydalis yanhusuo in Helicobacter pylori infection and its bioactive component, dehydrocorydaline. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116173. [PMID: 36681166 DOI: 10.1016/j.jep.2023.116173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/01/2023] [Accepted: 01/13/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Helicobacter pylori (H. pylori) infection is a frequent chronic infection. Persistent infection is the strongest risk factor for developing gastric complications leading to gastric cancer. The antibiotic resistance of current anti-H. pylori drugs lead to the search for novel candidates from medicinal plants. Traditionally, Corydalis yanhusuo (Y.H. Chou & Chun C.Hsu) W.T. Wang ex Z.Y. Su & C.Y. Wu (Papaveraceae) has been used for the treatment of digestive system diseases in China. So, it's essential to explore and confirm the anti-H. pylori activity of C. yanhusuo and characterize the pharmacologically active compounds. AIM OF THE STUDY This study aims to evaluate the efficacy of C. yanhusuo as complementary or alternative modes of treatment against H. pylori-related diseases and ascertain the active substances of C. yanhusuo to develop non-toxic, natural, and inexpensive products. MATERIALS AND METHODS C. yanhusuo was subjected to solid-liquid extraction with water (WECY), ethanol EECY), and chloroform (CECY). The extracts were screened by agar diffusion assay, the minimum inhibitory concentrations (MIC), the minimum bactericidal (MBC) for their in vitro antimicrobial activity, and by Berthelot reaction for urease inhibition. To assess the in vivo action, H. pylori-induced C57BL/6 mice were used to detect RUT biopsy, perform visual and histopathological analyses and evaluate IgG expression. Furthermore, we compared the anti-H. pylori activities of major alkaloids in CECY to identify the bioactive constituents. RESULTS Among the three C. yanhusuo extracts, CECY showed the maximum in vitro antibacterial activity. Administration of CECY significantly inhibited the survival of H. pylori colonized in the gastric mucosa and alleviated gastric damage along with a reduction in the expression levels of IgG in H. pylori-infected mice. Berberine and dehydrocorydaline exhibited obvious anti-H. pylori activity with MIC of 25 and 12.5 μg/mL, respectively. CONCLUSION C. yanhusuo extracts showed anti-H. pylori activity in different degrees. Among them, CECY showed significant anti-H. pylori, gastroprotective and anti-inflammatory activities in vivo and in vitro. Dehydrocorydalmine, an active alkaloid compound isolated from C. yanhusuo, warranted further investigation for its potential anti-H. pylori activity.
Collapse
Affiliation(s)
- Hao Wu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Qingyue Sun
- No. one Clinical Medicine School of Binzhou Medical University, Binzhou Medical University, Yantai, 264003, China
| | - Huirong Dong
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Jiasen Qiao
- No. one Clinical Medicine School of Binzhou Medical University, Binzhou Medical University, Yantai, 264003, China
| | - Ying Lin
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Chen Yu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| | - Yanni Li
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
38
|
Aniceto N, Albuquerque TS, Bonifácio VDB, Guedes RC, Martinho N. Using Machine Learning and Molecular Docking to Leverage Urease Inhibition Data for Virtual Screening. Int J Mol Sci 2023; 24:8180. [PMID: 37175889 PMCID: PMC10179503 DOI: 10.3390/ijms24098180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Urease is a metalloenzyme that catalyzes the hydrolysis of urea, and its modulation has an important role in both the agricultural and medical industry. Even though numerous molecules have been tested against ureases of different species, their clinical translation has been limited due to chemical and metabolic stability as well as side effects. Therefore, screening new compounds against urease would be of interest in part due to rising concerns regarding antibiotic resistance. In this work, we collected and curated a diverse set of 2640 publicly available small-molecule inhibitors of jack bean urease and developed a classifier using a random forest machine learning method with high predictive performance. In addition, the physicochemical features of compounds were paired with molecular docking and protein-ligand fingerprint analysis to gather insight into the current activity landscape. We observed that the docking score could not differentiate active from inactive compounds within each chemical family, but scores were correlated with compound activity when all compounds were considered. Additionally, a decision tree model was built based on 2D and 3D Morgan fingerprints to mine patterns of the known active-class compounds. The final machine learning model showed good prediction performance against the test set (81% and 77% precision for active and inactive compounds, respectively). Finally, this model was employed, as a proof-of-concept, on an in-house library to predict new hits that were then tested against urease and found to be active. This is, to date, the largest, most diverse dataset of compounds used to develop predictive in silico models. Overall, the results highlight the usefulness of using machine learning classifiers and molecular docking to predict novel urease inhibitors.
Collapse
Affiliation(s)
- Natália Aniceto
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Tânia S. Albuquerque
- iBB—Institute for Bioengineering and Biosciences, and Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Vasco D. B. Bonifácio
- iBB—Institute for Bioengineering and Biosciences, and Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Bioengineering Department, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Rita C. Guedes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Nuno Martinho
- iBB—Institute for Bioengineering and Biosciences, and Associate Laboratory i4HB—Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
39
|
Malfertheiner P, Camargo MC, El-Omar E, Liou JM, Peek R, Schulz C, Smith SI, Suerbaum S. Helicobacter pylori infection. Nat Rev Dis Primers 2023; 9:19. [PMID: 37081005 PMCID: PMC11558793 DOI: 10.1038/s41572-023-00431-8] [Citation(s) in RCA: 228] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 04/22/2023]
Abstract
Helicobacter pylori infection causes chronic gastritis, which can progress to severe gastroduodenal pathologies, including peptic ulcer, gastric cancer and gastric mucosa-associated lymphoid tissue lymphoma. H. pylori is usually transmitted in childhood and persists for life if untreated. The infection affects around half of the population in the world but prevalence varies according to location and sanitation standards. H. pylori has unique properties to colonize gastric epithelium in an acidic environment. The pathophysiology of H. pylori infection is dependent on complex bacterial virulence mechanisms and their interaction with the host immune system and environmental factors, resulting in distinct gastritis phenotypes that determine possible progression to different gastroduodenal pathologies. The causative role of H. pylori infection in gastric cancer development presents the opportunity for preventive screen-and-treat strategies. Invasive, endoscopy-based and non-invasive methods, including breath, stool and serological tests, are used in the diagnosis of H. pylori infection. Their use depends on the specific individual patient history and local availability. H. pylori treatment consists of a strong acid suppressant in various combinations with antibiotics and/or bismuth. The dramatic increase in resistance to key antibiotics used in H. pylori eradication demands antibiotic susceptibility testing, surveillance of resistance and antibiotic stewardship.
Collapse
Affiliation(s)
- Peter Malfertheiner
- Medical Department II, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany.
- Medical Department Klinik of Gastroenterology, Hepatology and Infectiology, Otto-von-Guericke Universität, Magdeburg, Germany.
| | - M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Emad El-Omar
- Microbiome Research Centre, St George & Sutherland Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jyh-Ming Liou
- Department of Internal Medicine, National Taiwan University Cancer Center, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Richard Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christian Schulz
- Medical Department II, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- DZIF Deutsches Zentrum für Infektionsforschung, Partner Site Munich, Munich, Germany
| | - Stella I Smith
- Department of Molecular Biology and Biotechnology, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Sebastian Suerbaum
- DZIF Deutsches Zentrum für Infektionsforschung, Partner Site Munich, Munich, Germany
- Max von Pettenkofer Institute, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- National Reference Center for Helicobacter pylori, Munich, Germany
| |
Collapse
|
40
|
Mollazadeh M, Azizian H, Fakhrioliaei A, Iraji A, Avizheh L, Valizadeh Y, Zomorodian K, Elahi F, Moazzam A, Kazemzadeh H, Amanlou M, Garmciri F, Hamidian E, Biglar M, Larijani B, Mahdavi M. Different barbiturate derivatives linked to aryl hydrazone moieties as urease inhibitors; design, synthesis, urease inhibitory evaluations, and molecular dynamic simulations. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03050-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
41
|
Ata A, Salar U, Saleem F, Lateef M, Khan SA, Khan KM, Taha M, Haider SM, Ul-Haq Z. Identification of potential urease inhibitors and antioxidants based on saccharin derived analogs: Synthesis, in vitro, and in silico studies. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
42
|
Hosseinzadeh N, Nazari Montazer M, Mohammadi‐Khanaposhtani M, Valizadeh Y, Amanlou M, Mahdavi M. Rational Design, Synthesis, Docking Simulation, and ADMET Prediction of Novel Barbituric‐hydrazine‐phenoxy‐1,2,3‐triazole‐acetamide Derivatives as Potent Urease Inhibitors. ChemistrySelect 2023. [DOI: 10.1002/slct.202203297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Nouraddin Hosseinzadeh
- Laboratory of Organic Synthesis and Natural Products Department of Chemistry Sharif University of Technology Tehran Iran
| | - Mohammad Nazari Montazer
- Department of Medicinal Chemistry Faculty of Pharmacy, Tehran University of Medical Sciences Tehran Iran
| | - Maryam Mohammadi‐Khanaposhtani
- Cellular and Molecular Biology Research Center Health Research Institute Babol University of Medical Sciences Babol Iran
| | - Yousef Valizadeh
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry Faculty of Pharmacy, Tehran University of Medical Sciences Tehran Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center Endocrinology and Metabolism Clinical Sciences Institute Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
43
|
Macegoniuk K, Tabor W, Mazzei L, Cianci M, Giurg M, Olech K, Burda-Grabowska M, Kaleta R, Grabowiecka A, Mucha A, Ciurli S, Berlicki Ł. Optimized Ebselen-Based Inhibitors of Bacterial Ureases with Nontypical Mode of Action. J Med Chem 2023; 66:2054-2063. [PMID: 36661843 PMCID: PMC9923736 DOI: 10.1021/acs.jmedchem.2c01799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Screening of 25 analogs of Ebselen, diversified at the N-aromatic residue, led to the identification of the most potent inhibitors of Sporosarcina pasteurii urease reported to date. The presence of a dihalogenated phenyl ring caused exceptional activity of these 1,2-benzisoselenazol-3(2H)-ones, with Ki value in a low picomolar range (<20 pM). The affinity was attributed to the increased π-π and π-cation interactions of the dihalogenated phenyl ring with αHis323 and αArg339 during the initial step of binding. Complementary biological studies with selected compounds on the inhibition of ureolysis in whole Proteus mirabilis cells showed a very good potency (IC50 < 25 nM in phosphate-buffered saline (PBS) buffer and IC90 < 50 nM in a urine model) for monosubstituted N-phenyl derivatives. The crystal structure of S. pasteurii urease inhibited by one of the most active analogs revealed the recurrent selenation of the Cys322 thiolate, yielding an unprecedented Cys322-S-Se-Se chemical moiety.
Collapse
Affiliation(s)
- Katarzyna Macegoniuk
- Department
of Bioorganic Chemistry, Wrocław University
of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Wojciech Tabor
- Department
of Bioorganic Chemistry, Wrocław University
of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Luca Mazzei
- Laboratory
of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology
(FaBiT), University of Bologna, Viale Giuseppe Fanin 40, 40138 Bologna, Italy
| | - Michele Cianci
- Department
of Agricultural, Food and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche 10, 60131 Ancona, Italy
| | - Mirosław Giurg
- Department
of Organic and Medicinal Chemistry, Wrocław
University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Kamila Olech
- Department
of Organic and Medicinal Chemistry, Wrocław
University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Małgorzata Burda-Grabowska
- Department
of Organic and Medicinal Chemistry, Wrocław
University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Rafał Kaleta
- Department
of Organic and Medicinal Chemistry, Wrocław
University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Agnieszka Grabowiecka
- Department
of Bioorganic Chemistry, Wrocław University
of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Artur Mucha
- Department
of Bioorganic Chemistry, Wrocław University
of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Stefano Ciurli
- Laboratory
of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology
(FaBiT), University of Bologna, Viale Giuseppe Fanin 40, 40138 Bologna, Italy
| | - Łukasz Berlicki
- Department
of Bioorganic Chemistry, Wrocław University
of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland,. Phone: +48 71 320
3344. Fax: +48 71 320 2427
| |
Collapse
|
44
|
Al-Rooqi MM, Mughal EU, Raja QA, Hussein EM, Naeem N, Sadiq A, Asghar BH, Moussa Z, Ahmed SA. Flavonoids and related privileged scaffolds as potential urease inhibitors: a review. RSC Adv 2023; 13:3210-3233. [PMID: 36756398 PMCID: PMC9869662 DOI: 10.1039/d2ra08284e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Infections caused by bacteria are a significant issue on a global scale, and imperative action is required to discover novel or improved therapeutic agents. Flavonoids are a class of plant-derived compounds that have a variety of potentially useful bioactivities. These activities include immediate antimicrobial properties, synergistic effect with antimicrobials, ferocious repression of pathogenicity, anti-urease activity etc. This review summarizes current studies concerning anti-urease actions of flavonoids as well as structural-activity correlation investigations of the flavonoid core structure. It is possible that if researchers investigate the many structural changes that may be made in flavonoid rings, they'll be able to build up novel compounds that have powerful and effective anti-urease properties.
Collapse
Affiliation(s)
- Munirah M Al-Rooqi
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
| | | | | | - Essam M Hussein
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
- Chemistry Department, Faculty of Science, Assiut University 71516 Assiut Egypt
| | - Nafeesa Naeem
- Department of Chemistry, University of Gujrat Gujrat-50700 Pakistan
| | - Amina Sadiq
- Department of Chemistry, Government College Women University Sialkot-51300 Pakistan
| | - Basim H Asghar
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
| | - Ziad Moussa
- Department of Chemistry, College of Science, United Arab Emirates University P.O. Box 15551, Al Ain Abu Dhabi United Arab Emirates
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University 21955 Makkah Saudi Arabia
- Chemistry Department, Faculty of Science, Assiut University 71516 Assiut Egypt
| |
Collapse
|
45
|
Patamia V, Floresta G, Zagni C, Pistarà V, Punzo F, Rescifina A. 1,2-Dibenzoylhydrazine as a Multi-Inhibitor Compound: A Morphological and Docking Study. Int J Mol Sci 2023; 24:1425. [PMID: 36674938 PMCID: PMC9864281 DOI: 10.3390/ijms24021425] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/24/2022] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
In the framework of the multitarget inhibitor study, we report an in silico analysis of 1,2-dibenzoylhydrazine (DBH) with respect to three essential receptors such as the ecdysone receptor (EcR), urease, and HIV-integrase. Starting from a crystallographic structural study of accidentally harvested crystals of this compound, we performed docking studies to evaluate the inhibitory capacity of DBH toward three selected targets. A crystal morphology prediction was then performed. The results of our molecular modeling calculations indicate that DBH is an excellent candidate as a ligand to inhibit the activity of EcR receptors and urease. Docking studies also revealed the activity of DBH on the HIV integrase receptor, providing an excellent starting point for developing novel inhibitors using this molecule as a starting lead compound.
Collapse
Affiliation(s)
| | | | | | | | | | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
46
|
Veronica rosea biomolecule profiling, antioxidant potential, dermoprotective effect, anti-inflammatory and hemostatic activities and enzyme inhibitory action. Eur J Integr Med 2022. [DOI: 10.1016/j.eujim.2022.102198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Yakan H, Omer HHS, Buruk O, Çakmak Ş, Marah S, Veyisoğlu A, Muğlu H, Ozen T, Kütük H. Synthesis, structure elucidation, biological activity, enzyme inhibition and molecular docking studies of new Schiff bases based on 5-nitroisatin-thiocarbohydrazone. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
48
|
In Vitro and In Silico Assessment of Bioactivity Properties and Pharmacokinetic Studies of New 3,5-Disubstituted-1,2,4-Triazoles. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
49
|
Freitas T, Bartelega L, Santos C, Dutra MP, Sarkis LF, Guimarães RJ, Dominghetti AW, Zito PC, Fernandes TJ, Guelfi D. Technologies for Fertilizers and Management Strategies of N-Fertilization in Coffee Cropping Systems to Reduce Ammonia Losses by Volatilization. PLANTS (BASEL, SWITZERLAND) 2022; 11:3323. [PMID: 36501362 PMCID: PMC9741429 DOI: 10.3390/plants11233323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 06/17/2023]
Abstract
The aim of this study was to quantify NH3-N losses from conventional, stabilized, slow-release, and controlled-release N fertilizers in a coffee field. The N fertilizers analyzed were prilled urea, prilled urea dissolved in water, ammonium sulfate (AS), ammonium nitrate (AN), urea + Cu + B, urea + adhesive + CaCO3, and urea + NBPT (all with three split applications), as well as blended N fertilizer, urea + elastic resin, urea-formaldehyde, and urea + polyurethane (all applied only once). NH3-N losses (mean of two crop seasons) were statistically higher for urea + adhesive + CaCO3 (27.9% of applied N) in comparison with the other treatments. Loss from prilled urea (23.7%) was less than from urea + adhesive + CaCO3. Losses from urea + NBPT (14.5%) and urea + Cu + B (13.5%) were similar and lower than those from prilled urea. Urea dissolved in water (4.2%) had even lower losses than those treatments, and the lowest losses were observed for AS (0.6%) and AN (0.5%). For the single application fertilizers, higher losses occurred for urea + elastic resin (5.8%), blended N fertilizer (5.5%), and urea + polyurethane (5.2%); and urea-formaldehyde had a lower loss (0.5%). Except for urea + adhesive + CaCO3, all N-fertilizer technologies reduced NH3-N losses compared to prilled urea.
Collapse
Affiliation(s)
- Tainah Freitas
- Department of Agriculture, Federal University of Lavras, Lavras 37203-202, Brazil
| | - Lucas Bartelega
- Department of Soil Science, Federal University of Lavras, Lavras 37203-202, Brazil
| | - César Santos
- Department of Soil Science, Federal University of Lavras, Lavras 37203-202, Brazil
| | - Mateus Portes Dutra
- Department of Soil Science, Federal University of Lavras, Lavras 37203-202, Brazil
| | | | | | | | | | | | - Douglas Guelfi
- Department of Soil Science, Federal University of Lavras, Lavras 37203-202, Brazil
| |
Collapse
|
50
|
Synthesis and Identification of New N, N-Disubstituted Thiourea, and Thiazolidinone Scaffolds Based on Quinolone Moiety as Urease Inhibitor. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27207126. [PMID: 36296723 PMCID: PMC9608620 DOI: 10.3390/molecules27207126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/15/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022]
Abstract
Synthesis of thiazolidinone based on quinolone moiety was established starting from 4-hydroxyquinol-2-ones. The strategy started with the reaction of ethyl bromoacetate with 4-hydroxyquinoline to give the corresponding ethyl oxoquinolinyl acetates, which reacted with hydrazine hydrate to afford the hydrazide derivatives. Subsequently, hydrazides reacted with isothiocyanate derivatives to give the corresponding N,N-disubstituted thioureas. Finally, on subjecting the N,N-disubstituted thioureas with dialkyl acetylenedicarboxylates, cyclization occurred, and thiazolidinone derivatives were obtained in good yields. The two series based on quinolone moiety, one containing N,N-disubstituted thioureas and the other containing thiazolidinone functionalities, were screened for their in vitro urease inhibition properties using thiourea and acetohydroxamic acid as standard inhibitors. The inhibition values of the synthesized thioureas and thiazolidinones exhibited moderate to good inhibitory effects. The structure-activity relationship revealed that N-methyl quinolonyl moiety exhibited a superior effect, since it was proved to be the most potent inhibitor in the present series achieving (IC50 = 1.83 ± 0.79 µM). The previous compound exhibited relatively much greater activity, being approximately 12-fold more potent than thiourea and acetohydroxamic acid as references. Molecular docking analysis showed a good protein-ligand interaction profile against the urease target (PDBID: 4UBP), emphasizing the electronic and geometric effect of N,N-disubstituted thiourea.
Collapse
|