1
|
Khan M, Zhang B, Zhang H, Wu J, Gao P, Li J. Ureases in nature: Multifaceted roles and implications for plant and human health - A review. Int J Biol Macromol 2025; 306:141702. [PMID: 40043969 DOI: 10.1016/j.ijbiomac.2025.141702] [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: 12/23/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 05/03/2025]
Abstract
Urease, a multifunctional enzyme that catalyzes the hydrolysis of urea into ammonia, plays a pivotal role in nitrogen metabolism across diverse organisms. While essential for survival, its unregulated activity is implicated in numerous pathologies, including peptic ulcers, nephropathy, and gastric cancer, as well as agricultural challenges such as soil ammonium depletion and reduced nitrogen-use efficiency. Beyond its canonical enzymatic function, urease engages in protein-protein interactions with bioactive counterparts like jaburetox, canatoxin, and soyuretox plant-derived proteins with insecticidal, antifungal, and membranolytic properties. Exploring the relationship between ureases and these proteins, along with their mechanistic synergies, presents novel opportunities to develop targeted inhibitors for urease-related diseases while unlocking broader therapeutic and biotechnological applications. This review delves into the dual roles of ureases in plants and humans, bridging the gap between their ureolytic and non-ureolytic activities. We highlight recent advances in the design of urease inhibitors, which have emerged as critical tools for managing pathologies such as Helicobacter pylori-induced ulcers and urease-mediated kidney stone formation. These inhibitors also hold transformative potential in agriculture, where they mitigate nitrogen loss by stabilizing urea fertilizers, thereby enhancing crop yields and reducing environmental pollution. Furthermore, their utility extends to industrial biotechnology, including biofilm disruption and wastewater treatment, where urease inhibition prevents microbially induced corrosion and ammonia toxicity. The collected information is anticipated to offer insightful guidance and effective strategies for developing novel potent and safe urease inhibitors in the future.
Collapse
Affiliation(s)
- Majid Khan
- College of Chemistry, Fuzhou University, 350116 Fuzhou, China
| | - Bo Zhang
- College of Chemistry, Fuzhou University, 350116 Fuzhou, China
| | - Han Zhang
- College of Chemistry, Fuzhou University, 350116 Fuzhou, China
| | - Juhong Wu
- College of Chemistry, Fuzhou University, 350116 Fuzhou, China
| | - Ping Gao
- College of Chemistry, Fuzhou University, 350116 Fuzhou, China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, 350116 Fuzhou, China; College of Biological and Pharmaceutical Engineering, Jilin Agricultural Science and Technology University, 132101 Jilin, China.
| |
Collapse
|
2
|
Zaman H, Saeed A, Ul Muntaha T, Ismail H, Hashmi SK, E Rubab U, Alsharif A, Soliman MM, Gaber A, Mumtaz A. Development of novel cyclopropyl tethered iminothiazolidinone-isatin hybrids as effective multi target intestinal alkaline phosphatase, urease and α-glucosidase inhibitors. Int J Biol Macromol 2025; 310:143377. [PMID: 40274167 DOI: 10.1016/j.ijbiomac.2025.143377] [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: 02/24/2025] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
In the pursuit of potent enzyme inhibitors to combat metabolic and microbial diseases, here we report the rational design and synthesis of novel cyclopropyl-tethered 2-iminothiazolidin-4-one-isatin hybrids (7a-k), aimed at overcoming limitations of current therapeutics in terms of potency, selectivity, and safety. The structures were confirmed through spectroscopy and compounds were assessed for inhibitory potential against α-glucosidase, urease, and Intestinal Alkaline phosphatase (IALP). Notably, compound (7d) featuring n-heptyl chain exhibited the highest potency against IALP, with an IC₅₀ value of 55.70 ± 0.19 μM "mean ± SEM (n = 2)", surpassing the standard L-phenylalanine (IC₅₀ = 80.7 ± 0.09 μM). For urease inhibition, n-butyl substituted compound (7a) demonstrated the highest effectiveness (IC₅₀ = 56.52 ± 0.14 μM), while n-pentyl substituted compound (7b) showed the pronounced α-glucosidase inhibition (IC₅₀ = 63.80 ± 0.15 μM) however less active than their standard inhibitors. The SAR analysis indicated that variations in aryl and alkyl substituents, particularly alkyl chain length, significantly influenced biological activity. Interestingly compounds (7j) and (7k) were found to be the least active for all enzymes. Computational studies provided further insights into the compounds' electronic properties, binding affinities, and pharmacokinetic profiles. All these findings underscore the therapeutic potential of cyclopropyl-tethered 2-iminothiazolidin-4-one-isatin hybrids as a promising scaffold for multifunctional enzyme inhibition, paving the way for future drug discovery and optimization efforts.
Collapse
Affiliation(s)
- Hina Zaman
- Department of Chemistry, Quaid-I-Azam University, Islamabad 45320, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-I-Azam University, Islamabad 45320, Pakistan.
| | - Tamknat Ul Muntaha
- Department of Chemistry, Quaid-I-Azam University, Islamabad 45320, Pakistan
| | - Hammad Ismail
- Department of Biochemistry and Biotechnology, University of Gujrat, Gujrat 50700, Pakistan
| | - Syeda Kinza Hashmi
- Department of Biochemistry and Biotechnology, University of Gujrat, Gujrat 50700, Pakistan
| | - Umm E Rubab
- Department of Biochemistry and Biotechnology, University of Gujrat, Gujrat 50700, Pakistan
| | - Abdualziz Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Mohamed Mohamed Soliman
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, Taif, Saudi Arabia
| | - Ahmed Gaber
- Department of Biology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Amara Mumtaz
- Department of Chemistry, COMSATS University, Abbottabad 22060, Pakistan
| |
Collapse
|
3
|
Bilal H, Ullah S, Halim SA, Khan M, Avula SK, Alam A, Zayed ES, El-Ghaiesh SH, Ogaly HA, Shah Z, Khan A, Al-Harrasi A. Design and synthesis of terephthalic dihydrazide analogues as dual inhibitors of glycation and urease. RSC Adv 2025; 15:9510-9520. [PMID: 40161525 PMCID: PMC11951093 DOI: 10.1039/d5ra00459d] [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: 01/19/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
The overexpression of urease is the root cause of peptic ulcers and gastritis. Therefore, introducing new inhibitors against urease is a possible therapeutic approach to overcoming the pathogenesis; for instance, limiting the risk of development of urinary calculi. Moreover, glycation is the leading cause of several complications. Thus, in this study, we synthesized novel terephthalic dihydrazide analogues and evaluated their biological importance. These terephthalic dihydrazide analogues were characterized using advanced spectroscopic techniques, such as 1H NMR, 13C NMR, 19F NMR and HRMS (ESI+), and FT-IR. Fortunately, 6 of the 11 synthesized compounds exhibited urease inhibitory capability, and 8 compounds exhibited anti-glycation capability. Compounds 13-14, 20 and 23 showed significant urease inhibition with IC50 values of 63.12 ± 0.28, 65.71 ± 0.40, 49.2 ± 0.49 and 51.45 ± 0.39 μM, respectively. Meanwhile, they exhibited potent anti-glycation activity with IC50 values of 67.53 ± 0.46, 68.06 ± 0.43, 48.32 ± 0.42 and 54.36 ± 0.40 μM, respectively. Molecular docking of active urease inhibitors showed their good binding at the entrance of the active site and good correlation with our in vitro results.
Collapse
Affiliation(s)
- Hazrat Bilal
- Department of Chemistry, Bacha Khan University Charsadda Charsadda-24420 Khyber Pakhtunkhwa Pakistan
- Department of Chemistry, Government Postgraduate College Dargai Malakand Pakistan
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| | - Momin Khan
- Department of Chemistry, Abdul Wali Khan University Mardan Mardan 23200 Pakistan
| | - Satya Kumar Avula
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| | - Aftab Alam
- Department of Biochemistry, Abdul Wali Khan University Mardan Mardan 23200 Pakistan
| | - Eman Serry Zayed
- Department of Clinical Biochemistry, Faculty of Medicine, University of Tabuk Tabuk 71491 Saudi Arabia
| | - Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, University of Tabuk Tabuk 71491 Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Tanta University Tanta Egypt
| | - Hanan A Ogaly
- Chemistry Department, College of Science, King Khalid University Abha 61421 Saudi Arabia
| | - Zarbad Shah
- Department of Chemistry, Bacha Khan University Charsadda Charsadda-24420 Khyber Pakhtunkhwa Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
- Department of Chemical and Biological Engineering, College of Engineering, Korea University Seongbuk-gu 02841 Republic of Korea
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa PO Box 33, 616 Birkat Al Mauz Nizwa Sultanate of Oman +968-98957352 +968 25446328
| |
Collapse
|
4
|
Islam M, Ullah S, Khan A, Batool Z, Mali SN, Gurav SS, Dahlous KA, Mohammad S, Hussain J, Al-Harrasi A, Shafiq Z. Design, synthesis, in vitro, and in silico studies of 4-fluorocinnamaldehyde based thiosemicarbazones as urease inhibitors. Sci Rep 2025; 15:609. [PMID: 39753613 PMCID: PMC11698833 DOI: 10.1038/s41598-024-83386-4] [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: 08/17/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025] Open
Abstract
Clinically significant problems such as kidney stones and stomach ulcers are linked to the activation of the urease enzyme. At low pH, this enzyme gives an ideal environment to Helicobacter pylori in the stomach which is the cause of gastric ulcers and peptic ulcers. In recent work, we have developed a library of 4-fluorocinnamaldehyde base thiosemicarbazones and assessed them for their potential against urease enzyme. The synthesized compounds displayed significant to moderate inhibition potential with IC50 values ranging from 2.7 ± 0.5 µM to 29.0 ± 0.5 µM. compound 3c displayed the highest inhibition potential followed by 3a and 3b. Two compounds of the series 3f and 3 g remained inactive against urease. The kinetic study of compound 3c exhibited a competitive type of inhibition with a Ki value of 3.26 ± 0.0048 µM. SAR analysis was also thoroughly done. Molecular docking was used to analyze the interaction pattern of each derivative, and the outcomes demonstrated that the compounds had excellent binding interactions with the active site.
Collapse
Affiliation(s)
- Muhammad Islam
- Department of Basic Sciences and Humanities (Chemistry), Muhammad Nawaz Sharif University of Engineering and Technology (MNSUET), 60000, Multan, Pakistan
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin, 300072, China
| | - Saeed Ullah
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, P.O. Box 33, Nizwa, PC 616, Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, P.O. Box 33, Nizwa, PC 616, Sultanate of Oman
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Zahra Batool
- Institute of Chemical Sciences, Bahauddin Zakariya University, 60800, Multan, Pakistan
| | - Suraj N Mali
- School of Pharmacy, D.Y. Patil University (Deemed to be University), Sector 7, Nerul, Navi, Mumbai, 400706, India
| | - Shailesh S Gurav
- Department of Chemistry, VIVA College, Virar, Maharashtra, India
| | - Kholood A Dahlous
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Saikh Mohammad
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Javid Hussain
- Department of Biological Sciences and Chemistry, University of Nizwa, Nizwa, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, P.O. Box 33, Nizwa, PC 616, Sultanate of Oman.
| | - Zahid Shafiq
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
5
|
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
|
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
|
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
|
8
|
Emmanuel BN, Peter DA, Peter MO, Adedayo IS, Olaifa K. Helicobacter pylori infection in Africa: comprehensive insight into its pathogenesis, management, and future perspectives. JOURNAL OF UMM AL-QURA UNIVERSITY FOR APPLIED SCIENCES 2024. [DOI: 10.1007/s43994-024-00166-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/22/2024] [Indexed: 01/04/2025]
Abstract
AbstractHelicobacter pylori is a widespread bacterium that has effectively colonized half of the global population, with Africa having over 70% of the total burden of H. pylori infections (HPI). Considering its acknowledged classification of as bacterial carcinogens and their significant contribution to the development of gastrointestinal disorders such as gastritis, peptic ulcers, and gastric neoplasia, together with their growing resistance to antibiotics. Gaining insight into the etiology of this organism is crucial in order to investigate and develop appropriate treatment strategies. Furthermore, the rise of bacteria that are resistant to antibiotics presents an extra danger in managing this detrimental bacterium. Our review focuses on investigating the presence of H. pylori in Africa and analyzing the various factors that contribute to its extensive prevalence. We simplified the complex mechanisms that H. pylori utilizes to flourish in the human body, with a specific emphasis on its virulence factors and antibiotic resistance. These variables pose significant challenges to conventional treatment strategies. In addition, we analyze both conventional and developing diagnostic methods, as well as the current treatment approaches implemented in various African nations. In addition, we tackle the distinct healthcare obstacles of the region and put-up practical remedies. The main goal of this review is to improve the formulation of more efficient methods for the management and treatment of HPI in Africa.
Collapse
|
9
|
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
|
10
|
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
|
11
|
Fath MK, Khalili S, Boojar MMA, Hashemi ZS, Zarei M. Clodronic Acid has Strong Inhibitory Interactions with the Urease Enzyme of Helicobacter pylori: Computer-aided Design and in vitro Confirmation. Curr Comput Aided Drug Des 2024; 20:1100-1112. [PMID: 37957909 DOI: 10.2174/0115734099271837231026064439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Helicobacter pylori (HP) infection could lead to various gastrointestinal diseases. Urease is the most important virulence factor of HP. It protects the bacterium against gastric acid. OBJECTIVE Therefore, we aimed to design urease inhibitors as drugs against HP infection. METHODS The DrugBank-approved library was assigned with 3D conformations and the structure of the urease was prepared. Using a re-docking strategy, the proper settings were determined for docking by PyRx and GOLD software. Virtual screening was performed to select the best inhibitory drugs based on binding affinity, FitnessScore, and binding orientation to critical amino acids of the active site. The best inhibitory drug was then evaluated by IC50 and the diameter of the zone of inhibition for bacterial growth. RESULTS The structures of prepared drugs were screened against urease structure using the determined settings. Clodronic acid was determined to be the best-identified drug, due to higher PyRx binding energy, better GOLD FitnessScore, and interaction with critical amino acids of urease. In vitro results were also in line with the computational data. IC50 values of Clodronic acid and Acetohydroxamic Acid (AHA) were 29.78 ± 1.13 and 47.29 ± 2.06 μg/ml, respectively. Diameters of the zones of inhibition were 18 and 15 mm for Clodronic acid and AHA, respectively. CONCLUSION Clodronic acid has better HP urease inhibition potential than AHA. Given its approved status, the development of a repurposed drug based on Clodronic acid would require less time and cost. Further, in vivo studies would unveil the efficacy of Clodronic acid as a urease inhibitor.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | | | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Mahboubeh Zarei
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
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
|
13
|
Dilshad R, Khan KUR, Ahmad S, Shaik Mohammad AA, Sherif AE, Rao H, Ahmad M, Ghalloo BA, Begum MY. Phytochemical characterization of Typha domingensis and the assessment of therapeutic potential using in vitro and in vivo biological activities and in silico studies. Front Chem 2023; 11:1273191. [PMID: 38025070 PMCID: PMC10663946 DOI: 10.3389/fchem.2023.1273191] [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: 08/05/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Typha domingensis, a medicinal plant with significant traditional importance for curing various human diseases, has potentially bioactive compounds but was less explored previously. Therefore, this study aims to investigate the therapeutic potential of T. domingensis by evaluating the phytochemical profile through high-performance liquid chromatography (HPLC) techniques and its biological activities (in vitro and in vivo) from the methanolic extract derived from the entire plant (TDME). The secondary metabolite profile of TDME regulated by reverse phase ultra-high-performance liquid chromatography-mass spectrometry (RP-UHPLC-MS) revealed some bioactive compounds by -ve and +ve modes of ionization. The HPLC quantification study showed the precise quantity of polyphenols (p-coumaric acid, 207.47; gallic acid, 96.25; and kaempferol, 95.78 μg/g extract). The enzyme inhibition assays revealed the IC50 of TDME as 44.75 ± 0.51, 52.71 ± 0.01, and 67.19 ± 0.68 µgmL-1, which were significant compared to their respective standards (indomethacin, 18.03 ± 0.12; quercetin, 4.11 ± 0.01; and thiourea, 8.97 ± 0.11) for lipoxygenase, α-glucosidase, and urease, respectively. Safety was assessed by in vitro hemolysis (4.25% ± 0.16% compared to triton × 100, 93.51% ± 0.36%), which was further confirmed (up to 10 g/kg) by an in vivo model of rats. TDME demonstrated significant (p < 0.05) potential in analgesic activity by hot plate and tail immersion tests and anti-inflammatory activity by the carrageenan-induced hind paw edema model. Pain latency decreased significantly, and the anti-inflammatory effect increased in a dose-dependent way. Additionally, in silico molecular docking revealed that 1,3,4,5-tetracaffeoylquinic acid and formononetin 7-O-glucoside-6″-O-malonate possibly contribute to enzyme inhibitory activities due to their higher binding affinities compared to standard inhibitors. An in silico absorption, distribution, metabolism, excretion, and toxicological study also predicted the pharmacokinetics and safety of the chosen compounds identified from TDME. To sum up, it was shown that TDME contains bioactive chemicals and has strong biological activities. The current investigations on T. domingensis could be extended to explore its potential applications in nutraceutical industries and encourage the isolation of novel molecules with anti-inflammatory and analgesic effects.
Collapse
Affiliation(s)
- Rizwana Dilshad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Kashif-ur-Rehman Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Saeed Ahmad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | | | - Asmaa E. Sherif
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam bin Abdul Aziz, Al-Khar, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Huma Rao
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Maqsood Ahmad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Bilal Ahmad Ghalloo
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - M. Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
14
|
Manoharan A, Farrell J, Aldilla VR, Whiteley G, Kriel E, Glasbey T, Kumar N, Moore KH, Manos J, Das T. N-acetylcysteine prevents catheter occlusion and inflammation in catheter associated-urinary tract infections by suppressing urease activity. Front Cell Infect Microbiol 2023; 13:1216798. [PMID: 37965267 PMCID: PMC10641931 DOI: 10.3389/fcimb.2023.1216798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/19/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction Proteus mirabilis is a key pathobiont in catheter-associated urinary tract infections (CA-UTIs), which is well known to form crystalline biofilms that occlude catheters. Urease activity alkylates urine through the release of ammonia, consequentially resulting in higher levels of Mg2+ and Ca2+ and formation of crystals. In this study, we showed that N-acetyl cysteine (NAC), a thiol antioxidant, is a potent urease inhibitor that prevents crystalline biofilm formation. Methods To quantify urease activity, Berthelot's method was done on bacterial extracts treated with NAC. We also used an in vitro catheterised glass bladder model to study the effect of NAC treatment on catheter occlusion and biofilm encrustation in P. mirabilis infections. Inductively-coupled plasma mass spectrometry (ICP-MS) was performed on catheter samples to decipher elemental profiles. Results NAC inhibits urease activity of clinical P. mirabilis isolates at concentrations as low as 1 mM, independent of bacterial killing. The study also showed that NAC is bacteriostatic on P. mirabilis, and inhibited biofilm formation and catheter occlusion in an in vitro. A significant 4-8log10 reduction in viable bacteria was observed in catheters infected in this model. Additionally, biofilms in NAC treated catheters displayed a depletion of calcium, magnesium, or phosphates (>10 fold reduction), thus confirming the absence of any urease activity in the presence of NAC. Interestingly, we also showed that not only is NAC anti-inflammatory in bladder epithelial cells (BECs), but that it mutes its inflammatory response to urease and P. mirabilis infection by reducing the production of IL-6, IL-8 and IL-1b. Discussion Using biochemical, microbiological and immunological techniques, this study displays the functionality of NAC in preventing catheter occlusion by inhibiting urease activity. The study also highlights NAC as a strong anti-inflammatory antibiofilm agent that can target both bacterial and host factors in the treatment of CA-UTIs.
Collapse
Affiliation(s)
- Arthika Manoharan
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Institute of Infectious Disease, The University of Sydney, Sydney, NSW, Australia
| | - Jessica Farrell
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Whiteley Corporation, Tomago, NSW, Australia
| | - Vina R. Aldilla
- School of Chemistry, The University of New South Wales, Sydney, NSW, Australia
| | - Greg Whiteley
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Whiteley Corporation, Tomago, NSW, Australia
- School of Medicine, Western Sydney University, NSW, Australia
| | - Erik Kriel
- Whiteley Corporation, Tomago, NSW, Australia
| | | | - Naresh Kumar
- School of Chemistry, The University of New South Wales, Sydney, NSW, Australia
| | - Kate H. Moore
- Department of Urogynaecology, St George Hospital, University of New South Wales, Sydney, NSW, Australia
| | - Jim Manos
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Institute of Infectious Disease, The University of Sydney, Sydney, NSW, Australia
| | - Theerthankar Das
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Institute of Infectious Disease, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
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: 15] [Impact Index Per Article: 7.5] [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
|
18
|
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]
|
19
|
Shahin AI, Zaib S, Zaraei SO, Kedia RA, Anbar HS, Younas MT, Al-Tel TH, Khoder G, El-Gamal MI. Design and synthesis of novel anti-urease imidazothiazole derivatives with promising antibacterial activity against Helicobacter pylori. PLoS One 2023; 18:e0286684. [PMID: 37267378 DOI: 10.1371/journal.pone.0286684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/21/2023] [Indexed: 06/04/2023] Open
Abstract
Urease enzyme is a known therapeutic drug target for treatment of Helicobacter pylori infection due to its role in settlement and growth in gastric mucosa. In this study, we designed a new series of sulfonates and sulfamates bearing imidazo[2,1-b]thiazole scaffold that exhibit a potent inhibitory activity of urease enzyme. The most potent compound 2c inhibited urease with an IC50 value of 2.94 ± 0.05 μM, which is 8-fold more potent than the thiourea positive control (IC50 = 22.3 ± 0.031 μM). Enzyme kinetics study showed that compound 2c is a competitive inhibitor of urease. Molecular modeling studies of the most potent inhibitors in the urease active site suggested multiple binding interactions with different amino acid residues. Phenotypic screening of the developed compounds against H. pylori delivered molecules of that possess high potency (1a, 1d, 1h, 2d, and 2f) in comparison to the positive control, acetohydroxamic acid. Additional studies to investigate the selectivity of these compounds against AGS gastric cell line and E. coli were performed. Permeability of the most promising derivatives (1a, 1d, 1h, 2d, and 2f) in Caco-2 cell line, was investigated. As a result, compound 1d presented itself as a lead drug candidate since it exhibited a promising inhibition against urease with an IC50 of 3.09 ± 0.07 μM, MIC value against H. pylori of 0.031 ± 0.011 mM, and SI against AGS of 6.05. Interestingly, compound 1d did not show activity against urease-negative E. coli and exhibited a low permeability in Caco-2 cells which supports the potential use of this compound for GIT infection without systemic effect.
Collapse
Affiliation(s)
- Afnan I Shahin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Sumera Zaib
- Faculty of Science and Technology, Department of Basic and Applied Chemistry, University of Central Punjab, Lahore, Pakistan
| | - Seyed-Omar Zaraei
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Reena A Kedia
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hanan S Anbar
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Muhammad Tayyab Younas
- Faculty of Science and Technology, Department of Basic and Applied Chemistry, University of Central Punjab, Lahore, Pakistan
| | - Taleb H Al-Tel
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ghalia Khoder
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohammed I El-Gamal
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Faculty of Pharmacy, Department of Medicinal Chemistry, Mansoura University, Mansoura, Egypt
| |
Collapse
|
20
|
Hina S, Zaib S, Uroos M, Zia-ur-Rehman M, Munir R, Riaz H, Syed Q, Abidi SHI. N-Arylacetamide derivatives of methyl 1,2-benzothiazine-3-carboxylate as potential drug candidates for urease inhibition. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230104. [PMID: 37035287 PMCID: PMC10073911 DOI: 10.1098/rsos.230104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 03/14/2023] [Indexed: 06/19/2023]
Abstract
Urease enzyme is an infectious factor that provokes the growth and colonization of virulence pathogenic bacteria in humans. To overcome the deleterious effects of bacterial infections, inhibition of urease enzyme is one of the promising approaches. The current study is designed to synthesize new 1,2-benzothiazine-N-arylacetamide derivatives 5(a-n) that can effectively provide a new drug candidate to avoid bacterial infections by urease inhibition. After structural elucidation by FT-IR, proton and carbon-13 NMR and mass spectroscopy, the synthesized compounds 5(a-n) were investigated to evaluate their inhibitory potential against urease enzyme. In vitro analysis against positive control of thiourea indicated that all the synthesized compounds have strong inhibitory strengths as compared to the reference drug. Compound 5k, being the most potent inhibitor, strongly inhibited the urease enzymes and revealed an IC50 value of 9.8 ± 0.023 µM when compared with the IC50 of thiourea (22.3 ± 0.031 µM)-a far more robust inhibitory potential. Docking studies of 5k within the urease active site revealed various significant interactions such as H-bond, π-alkyl with amino acid residues like Val744, Lys716, Ala16, Glu7452, Ala37 and Asp730.
Collapse
Affiliation(s)
- Sajila Hina
- Centre for Research in Ionic Liquids, School of Chemistry, University of the Punjab, Quaid e Azam Campus, Lahore 54590, Pakistan
- Applied Chemistry Research Centre, PCSIR Laboratories Complex, Lahore 54600, Pakistan
| | - Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan
| | - Maliha Uroos
- Centre for Research in Ionic Liquids, School of Chemistry, University of the Punjab, Quaid e Azam Campus, Lahore 54590, Pakistan
| | | | - Rubina Munir
- Department of Chemistry, Kinnaird College for Women, Lahore 54000, Pakistan
| | - Huma Riaz
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore 54590, Pakistan
| | - Quratulain Syed
- Applied Chemistry Research Centre, PCSIR Laboratories Complex, Lahore 54600, Pakistan
| | - Syed Hussain Imam Abidi
- Pakistan Council of Scientific and Industrial Research, 01-Constitution Avenue, G-5/2, Islamabad 44050, Pakistan
| |
Collapse
|
21
|
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
|
22
|
In Vivo Role of Two-Component Regulatory Systems in Models of Urinary Tract Infections. Pathogens 2023; 12:pathogens12010119. [PMID: 36678467 PMCID: PMC9861413 DOI: 10.3390/pathogens12010119] [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: 11/24/2022] [Revised: 12/23/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023] Open
Abstract
Two-component signaling systems (TCSs) are finely regulated mechanisms by which bacteria adapt to environmental conditions by modifying the expression of target genes. In bacterial pathogenesis, TCSs play important roles in modulating adhesion to mucosal surfaces, resistance to antibiotics, and metabolic adaptation. In the context of urinary tract infections (UTI), one of the most common types infections causing significant health problems worldwide, uropathogens use TCSs for adaptation, survival, and establishment of pathogenicity. For example, uropathogens can exploit TCSs to survive inside bladder epithelial cells, sense osmolar variations in urine, promote their ascension along the urinary tract or even produce lytic enzymes resulting in exfoliation of the urothelium. Despite the usefulness of studying the function of TCSs in in vitro experimental models, it is of primary necessity to study bacterial gene regulation also in the context of host niches, each displaying its own biological, chemical, and physical features. In light of this, the aim of this review is to provide a concise description of several bacterial TCSs, whose activity has been described in mouse models of UTI.
Collapse
|
23
|
Khaliq S, Khan MA, Ahmad I, Ahmad I, Ahmed J, Ullah F. Synthesis, antimicrobial and molecular docking study of structural analogues of 3-((5-(dimethylcarbamoyl)pyrrolidin-3-yl)thio)-6-(1-hydroxyethyl)-4-methyl-7-oxo-1-azabicyclo[3.2.0]heptane-2-carboxylic acid. PLoS One 2022; 17:e0278684. [PMID: 36574404 PMCID: PMC9794083 DOI: 10.1371/journal.pone.0278684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/21/2022] [Indexed: 12/28/2022] Open
Abstract
The goal of the current work was to create structural analogues of a beta lactam antibiotic that might be possibly effective against bacterial resistant strains. FTIR, 1H NMR, 13C NMR, and CHNS analyses were used to perform the spectroscopic study on the compounds M1-8. The effects of the aforementioned substances on gram-positive and gram-negative bacterial strains were investigated. Most of the eight compounds had antibacterial activity that was lower than or equivalent to that of the original medication, but two molecules, M2 and M3, surprisingly, had stronger antibacterial activity. The findings of synthesized analogues against alpha-glucosidase and DPPH inhibition were found to be modest, whereas M2, M3, and M7 strongly inhibited the urease. To comprehend the potential mode of action, a molecular docking research was conducted against urease and -amylase. The research may help in the quest for novel chemical compounds that would be effective against bacteria that are resistant to antibiotics.
Collapse
Affiliation(s)
- Saharish Khaliq
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
- * E-mail: (SK); (MAK)
| | - Mohsin Abbas Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
- * E-mail: (SK); (MAK)
| | - Irshad Ahmad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Imtiaz Ahmad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Javed Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Farhat Ullah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
24
|
Enzyme Inhibitory Activities of Extracts and Carpachromene from the Stem of Ficus benghalensis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7053655. [PMID: 36582600 PMCID: PMC9794428 DOI: 10.1155/2022/7053655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Ficus benghalensis is one of the potential medicinal plants which is used locally for the treatment of various ailments such as diabetes, antiasthmatic, and wound healing. To provide a scientific background to these folklores, the current study was designed to evaluate the extract and isolated compound against various enzymes such as ureases, tyrosinase, and phosphodiesterase. The methanolic extract and carpachromene demonstrated a significant urease inhibition effect with maximum percent inhibition of 72.09 and 92.87%, respectively. Regarding the tyrosinase inhibition, the percent antagonist effect of carpachromene and the methanolic extract was 84.80 and 70.98%, respectively. The phosphodiesterase was also significantly antagonized by crude extract and carpachromene with a maximum percent inhibition of 82.98% and 89.54%, respectively. The docking study demonstrated that the carpachromene fits well into the active site of all three enzymes with significant interactions. Carpachromene might possess the potential to inhibit all three enzymes and can effectively treat different diseases associated with the hyperactivity of these enzymes. In conclusion, the crude extract and carpachromene exhibit significant urease, tyrosinase, and phosphodiesterase inhibitory activity which might be used against various diseases. In conclusion, the crude extract and carpachromene exhibit significant urease, tyrosinase, and phosphodiesterase inhibitory activity which might be used against diabetes and bronchoconstriction. Further, the current study provides scientific backup to the folklore (antidiabetic and antiasthmatic) of Ficus benghalensis.
Collapse
|
25
|
Amini M, Abdel-Jalil R, Moghadam ES, Al-Sadi AM, Talebi M, Amanlou M, Shongwe M. Piperazine-based Semicarbazone Derivatives as Potent Urease Inhibitors:
Design, Synthesis, and Bioactivity Screening. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220405234009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
An enzyme called urease assists highly pathogenic bacteria in colonizing and
maintaining themselves. Accordingly, inhibiting urease enzymes has been shown to be a promising strategy
for preventing ureolytic bacterial infections.
Objective:
This study aimed to synthesize and evaluate the bioactivity of a series of semicarbazone derivatives.
Methods:
A series of piperazine-based semicarbazone derivatives 5a-o were synthesized and isolated, and
their structures were elucidated by 1H-NMR and 13C-NMR spectroscopic techniques besides MS and
elemental analysis. The urease inhibition activity of these compounds was evaluated using the standard
urease enzyme inhibition kit. An MTT assay was performed on two different cell lines (NIH-3T3 and
MCF-7) to investigate the cytotoxicity profile.
Results:
All semicarbazone 5a-o exhibited higher urease inhibition activity (3.95–6.62 μM) than the reference
standards thiourea and hydroxyurea (IC50: 22 and 100 μM, respectively). Derivatives 5m and 5o
exhibited the best activity with the IC50 values of 3.95 and 4.05 μM, respectively. Investigating the cytotoxicity
profile of the target compound showed that all compounds 5a-o have IC50 values higher than 50
μM for both tested cell lines.
Conclusion:
The results showed that semicarbazone derivatives could be highly effective as urease inhibitors.
Collapse
Affiliation(s)
- Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical
Sciences, Tehran 1417614411, Iran
- Drug Design and Development Research Center, The Institute of Pharmaceutical
Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Raid Abdel-Jalil
- Department of Chemistry, College of Science, Sultan Qaboos University, Al-Khod 123, Muscat, Sultanate of Oman
| | - Ebrahim Saeedian Moghadam
- Department of Chemistry, College of Science, Sultan Qaboos University, Al-Khod 123, Muscat, Sultanate of Oman
| | - Abdullah Mohammed Al-Sadi
- Department of Crop Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al-Khod 123,
Muscat, Sultanate of Oman
| | - Meysam Talebi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical
Sciences, Tehran 1417614411, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical
Sciences, Tehran 1417614411, Iran
- Drug Design and Development Research Center, The Institute of Pharmaceutical
Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Musa Shongwe
- Department of Chemistry, College of Science, Sultan Qaboos University, Al-Khod 123, Muscat, Sultanate of Oman
| |
Collapse
|
26
|
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
|
27
|
Naseer A, Osra FA, Awan AN, Imran A, Hameed A, Ali Shah SA, Iqbal J, Zakaria ZA. Exploring Novel Pyridine Carboxamide Derivatives as Urease Inhibitors: Synthesis, Molecular Docking, Kinetic Studies and ADME Profile. Pharmaceuticals (Basel) 2022; 15:1288. [PMID: 36297400 PMCID: PMC9609714 DOI: 10.3390/ph15101288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 09/07/2024] Open
Abstract
The rapid development of resistance by ureolytic bacteria which are involved in various life-threatening conditions such as gastric and duodenal cancer has induced the need to develop a new line of therapy which has anti-urease activity. A series of pyridine carboxamide and carbothioamide derivatives which also have some novel structures were synthesized via condensation reaction and investigated against urease for their inhibitory action. Among the series, 5-chloropyridine-2 yl-methylene hydrazine carbothioamide (Rx-6) and pyridine 2-yl-methylene hydrazine carboxamide (Rx-7) IC50 = 1.07 ± 0.043 µM, 2.18 ± 0.058 µM both possessed significant activity. Furthermore, molecular docking and kinetic studies were performed for the most potent inhibitors to demonstrate the binding mode of the active pyridine carbothioamide with the enzyme urease and its mode of interaction. The ADME profile also showed that all the synthesized molecules present oral bioavailability and high GI absorption.
Collapse
Affiliation(s)
- Ayesha Naseer
- Research Institute of Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
| | | | - Asia Naz Awan
- Research Institute of Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Aqeel Imran
- Center for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Abdul Hameed
- Department of Chemistry, University of Sahiwal, Sahiwal 57000, Pakistan
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam 42300, Selangor, Malaysia
- Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, Bandar Puncak Alam 42300, Selangor, Malaysia
| | - Jamshed Iqbal
- Center for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Zainul Amiruddin Zakaria
- Borneo Research on Algesia Inflammation and Neurodegeneration (BRAIN) Group, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Jalan UMS, Kota Kinabalu 88400, Sabah, Malaysia
| |
Collapse
|
28
|
Wu Y, Zhao S, Liu C, Hu L. Development of urease inhibitors by fragment-based dynamic combinatorial chemistry. ChemMedChem 2022; 17:e202200307. [PMID: 35975876 DOI: 10.1002/cmdc.202200307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/12/2022] [Indexed: 11/07/2022]
Abstract
In this study, fragment-based dynamic combinatorial chemistry (DCC) was explored for the development of novel urease inhibitors. Based on a rationally designed fragment, two iteratively evolved dynamic combinatorial libraries (DCLs) were generated and screened in the presence of urease template. The best ligand identified revealed not only strong urease inhibition but also low cytotoxicity. Additionally, possible inhibitory mechanism was elucidated in the binding kinetic study and docking simulation.
Collapse
Affiliation(s)
- Yao Wu
- Jiangsu University School of Pharmacy, College of Pharmacy, CHINA
| | - Shuang Zhao
- Jiangsu University School of Pharmacy, College of Pharmacy, CHINA
| | - Changming Liu
- Jiangsu University School of Pharmacy, College of Pharmacy, CHINA
| | - Lei Hu
- Jiangsu University School of Pharmacy, College of pharmacy, 301 Xuefu Rd., Zhenjiang, China, 212013, Zhenjiang, CHINA
| |
Collapse
|
29
|
Design and synthesis of new N-thioacylated ciprofloxacin derivatives as urease inhibitors with potential antibacterial activity. Sci Rep 2022; 12:13827. [PMID: 35970866 PMCID: PMC9378659 DOI: 10.1038/s41598-022-17993-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 08/03/2022] [Indexed: 01/06/2023] Open
Abstract
A new series of N-thioacylated ciprofloxacin 3a-n were designed and synthesized based on Willgerodt-Kindler reaction. The results of in vitro urease inhibitory assay indicated that almost all the synthesized compounds 3a-n (IC50 = 2.05 ± 0.03-32.49 ± 0.32 μM) were more potent than standard inhibitors, hydroxyurea (IC50 = 100 ± 2.5 μM) and thiourea (IC50 = 23 ± 0.84 μM). The study of antibacterial activity against Gram-positive species (S. aureus and S. epidermidis) revealed that the majority of compounds were more active than ciprofloxacin as the standard drug, and 3h derivative bearing 3-fluoro group had the same effect as ciprofloxacin against Gram-negative bacteria (P. aeruginosa and E. coli). Based on molecular dynamic simulations, compound 3n exhibited pronounced interactions with the critical residues of the urease active site and mobile flap pocket so that the quinolone ring coordinated toward the metal bi-nickel center and the essential residues at the flap site like His593, His594, and Arg609. These interactions caused blocking the active site and stabilized the movement of the mobile flap at the entrance of the active site channel, which significantly reduced the catalytic activity of urease. Noteworthy, 3n also exhibited IC50 values of 5.59 ± 2.38 and 5.72 ± 1.312 µg/ml to inhibit urease enzyme against C. neoformans and P. vulgaris in the ureolytic assay.
Collapse
|
30
|
Farooq U, Khan S, Naz S, Wani TA, Bukhari SM, Aborode AT, Shahzad SA, Zargar S. Three New Acrylic Acid Derivatives from Achillea mellifolium as Potential Inhibitors of Urease from Jack Bean and α-Glucosidase from Saccharomyces cerevisiae. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155004. [PMID: 35956953 PMCID: PMC9370616 DOI: 10.3390/molecules27155004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022]
Abstract
(1) Background: Achillea mellifolium belongs to a highly reputed family of medicinal plants, with plant extract being used as medicine in indigenous system. However, limited data is available regarding the exploitation of the medicinal potential of isolated pure compounds from this family; (2) Methods: A whole plant extract was partitioned into fractions and on the basis of biological activity, an ethyl acetate fraction was selected for isolation of pure compounds. Isolated compounds were characterized using different spectroscopic techniques. The compounds isolated from this study were tested for their medicinal potential using in-vitro enzyme assay, coupled with in-silico studies; (3) Results: Three new acrylic acid derivatives (1–3) have been isolated from the ethyl acetate fraction of Achillea mellifolium. The characterization of these compounds (1–3) was carried out using UV/Vis, FT-IR, 1D and 2D-NMR spectroscopy (1H-NMR, 13C-NMR, HMBC, NOESY) and mass spectrometry. These acrylic acid derivatives were further evaluated for their enzyme inhibition potential against urease from jack bean and α glucosidase from Saccharomyces cerevisiae, using both in-silico and in-vitro approaches. In-vitro studies showed that compound 3 has the highest inhibition against urease enzyme (IC50 =10.46 ± 0.03 μΜ), followed by compound 1 and compound 2 with percent inhibition and IC50 value of 16.87 ± 0.02 c and 13.71 ± 0.07 μΜ, respectively, compared to the standard (thiourea-IC50 = 21.5 ± 0.01 μΜ). The investigated IC50 value of compound 3 against the urease enzyme is two times lower compared to thiourea, suggesting that this compound is twice as active compared to the standard drug. On the other hand, all three compounds (1–3) revealed mild inhibition potential against α-glucosidase. In-silico molecular docking studies, in combination with MD simulations and free energy, calculations were also performed to rationalize their time evolved mode of interaction inside the active pocket. Binding energies were computed using a MMPBSA approach, and the role of individual residues to overall binding of the inhibitors inside the active pockets were also computed; (4) Conclusions: Together, these studies confirm the inhibitory potential of isolated acrylic acid derivatives against both urease and α-glucosidase enzymes; however, their inhibition potential is better for urease enzyme even when compared to the standard.
Collapse
Affiliation(s)
- Umar Farooq
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
- Correspondence: (U.F.); (S.K.); Tel.: +92-992383591 (U.F. & S.K.)
| | - Sara Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
- Correspondence: (U.F.); (S.K.); Tel.: +92-992383591 (U.F. & S.K.)
| | - Sadia Naz
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
| | - Tanveer A. Wani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Syed Majid Bukhari
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
| | | | - Sohail Anjum Shahzad
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, KPK, Pakistan
| | - Seema Zargar
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
31
|
Ahangarzadeh N, Shakour N, Rezvanpoor S, Bakherad H, Pakdel MH, Farhadi G, Sepehri S. Design, synthesis, and in silico studies of tetrahydropyrimidine analogs as urease enzyme inhibitors. Arch Pharm (Weinheim) 2022; 355:e2200158. [PMID: 35833485 DOI: 10.1002/ardp.202200158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/07/2022]
Abstract
The urease enzyme, a metalloenzyme having Ni2+ ions, is recognized in some bacteria, fungi, and plants. Particularly, it is vital to the progress of infections induced by pathogenic microbes, such as Proteus mirabilis and Helicobacter pylori. Herein, we reported the synthesis of a series of tetrahydropyrimidine derivatives and evaluated their antiurease activity. Finally, quantitative and qualitative analyses of the derivatives were performed via in silico studies. Urease inhibitory activity was determined as the reaction of H. pylori urease with different concentrations of compounds, and thiourea was used as a standard compound. Docking and dynamics methodologies were applied to study the interactions of the best compounds with the amino acids in the active site. All compounds showed good to excellent antiurease activity. The potent compounds were not cytotoxic against the HUVEC normal cell line. Based on the docking study, compound 4e with the highest urease inhibitory activity (IC50 = 6.81 ± 1.42 µM) showed chelates with both Ni2+ ions of the urease active site. Further, compound 4f displayed a very good inhibitory activity (IC50 = 8.45 ± 1.64 μM) in comparison to thiourea (IC50 = 22.03 ± 1.24 μM). The molecular docking and dynamics simulation results were correlated with the in vitro assay results. Moreover, the derivatives 4a-n followed Lipinski's rule-of-five and had drug-likeness properties.
Collapse
Affiliation(s)
- Nazli Ahangarzadeh
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sadaf Rezvanpoor
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamid Bakherad
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad H Pakdel
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghazaleh Farhadi
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saghi Sepehri
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.,Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
32
|
Moghadam ES, Al‐Sadi AM, Talebi M, Amanlou M, Stoll R, Amini M, Abdel‐Jalil R. Thiosemicarbazone Derivatives Act as Potent Urease Inhibitors; Synthesis, Bioactivity Screening and Molecular Docking Study. ChemistrySelect 2022. [DOI: 10.1002/slct.202200860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Ebrahim Saeedian Moghadam
- Department of Chemistry College of Science Sultan Qaboos University Muscat, P.O. Box 36, P.C. 123, Sultanate of Oman
| | - Abdullah Mohammed Al‐Sadi
- Department of Crop Sciences College of Agricultural and Marine Sciences Sultan Qaboos University Muscat Oman
| | - Meysam Talebi
- Department of Medicinal Chemistry Faculty of Pharmacy Tehran University of Medical Sciences Tehran 1417614411 Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry Faculty of Pharmacy Tehran University of Medical Sciences Tehran 1417614411 Iran
- Drug Design and Development Research Center The Institute of Pharmaceutical Sciences (TIPS) Tehran University of Medical Sciences Tehran Iran
| | - Raphael Stoll
- Biomolecular NMR Ruhr University of Bochum D 44780 Bochum Germany
| | - Mohsen Amini
- Department of Medicinal Chemistry Faculty of Pharmacy Tehran University of Medical Sciences Tehran 1417614411 Iran
- Drug Design and Development Research Center The Institute of Pharmaceutical Sciences (TIPS) Tehran University of Medical Sciences Tehran Iran
| | - Raid Abdel‐Jalil
- Department of Chemistry College of Science Sultan Qaboos University Muscat, P.O. Box 36, P.C. 123, Sultanate of Oman
| |
Collapse
|
33
|
Duran Ramirez JM, Gomez J, Obernuefemann CLP, Gualberto NC, Walker JN. Semi-Quantitative Assay to Measure Urease Activity by Urinary Catheter-Associated Uropathogens. Front Cell Infect Microbiol 2022; 12:859093. [PMID: 35392611 PMCID: PMC8980526 DOI: 10.3389/fcimb.2022.859093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Catheter-associated urinary tract infections (CAUTIs) are one of the most common healthcare-associated infections in the US, accounting for over 1 million cases annually and totaling 450 million USD. CAUTIs have high morbidity and mortality rates and can be caused by a wide range of pathogens, making empiric treatment difficult. Furthermore, when urease-producing uropathogens cause symptomatic CAUTI or asymptomatic catheter colonization, the risk of catheter failure due to blockage increases. The enzyme urease promotes catheter blockage by hydrolyzing urea in urine into ammonia and carbon dioxide, which results in the formation of crystals that coat the catheter surface. If CAUTI is left untreated, the crystals can grow until they block the urinary catheter. Catheter blockage and subsequent failure reduces the quality of life for the chronically catheterized, as it requires frequent catheter exchanges and can promote more severe disease, including dissemination of the infection to the kidneys or bloodstream. Thus, understanding how urease contributes to catheter blockages and/or more severe disease among the broad range of urease-producing microbes may provide insights into better prevention or treatment strategies. However, clinical assays that detect urease production among clinical isolates are qualitative and prioritize the detection of urease from Proteus mirabilis, the most well-studied uropathogenic urease producer. While urease from other known urease producers, such as Morganella morganii, can also be detected with these methods, other uropathogens, including Staphylococcus aureus and Klebsiella pneumonia, are harder to detect. In this study, we developed a high throughput, semiquantitative assay capable of testing multiple uropathogens in a rapid and efficient way. We validated the assay using Jack Bean urease, the urease producing species: Proteus spp., M. morganii, K. pneumonia, and S. aureus strains, and the non-urease producer: Escherichia coli. This modified assay more rapidly detected urease-producing strains compared to the current clinical test, Christensen Urea Agar, and provided semiquantitative values that may be used to further investigate different aspects of urease regulation, production, or activity in these diverse species. Furthermore, this assay can be easily adapted to account for different environmental stimuli affecting urease production, including bacterial concentration, aeration, or addition of anti-urease compounds.
Collapse
Affiliation(s)
- Jesus M. Duran Ramirez
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, United States
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Center for Infectious Diseases, School of Public Health, The University of Texas Health Science Center, Houston, TX, United States
| | - Jana Gomez
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, United States
| | - Chloe L. P. Obernuefemann
- The Center for Women’s Infectious Disease Research, Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Nathaniel C. Gualberto
- The Center for Women’s Infectious Disease Research, Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jennifer N. Walker
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, United States
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Center for Infectious Diseases, School of Public Health, The University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
34
|
Yaqoob S, Hameed A, Ahmed M, Imran M, Qadir MA, Ramzan M, Yousaf N, Iqbal J, Muddassar M. Antiurease screening of alkyl chain-linked thiourea derivatives: in vitro biological activities, molecular docking, and dynamic simulations studies. RSC Adv 2022; 12:6292-6302. [PMID: 35424581 PMCID: PMC8981555 DOI: 10.1039/d1ra08694d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/08/2022] [Indexed: 12/23/2022] Open
Abstract
Urease has become an important therapeutic target because it stimulates the pathogenesis of many human health conditions, such as pyelonephritis, the development of urolithiasis, hepatic encephalopathy, peptic ulcers, gastritis and gastric cancer. A series of alkyl chain-linked thiourea derivatives were synthesized to screen for urease inhibition activity. Structure elucidation of these compounds was done by spectral studies, such as IR, 1H NMR and 13C NMR, and MS analysis. In vitro urease enzyme inhibition assay revealed that compound 3c was the most potent thiourea derivative among the series with IC50 values of 10.65 ± 0.45 μM, while compound 3g also exhibited good activity with an IC50 value of 15.19 ± 0.58 μM compared to standard thiourea with an IC50 value of 15.51 ± 0.11 μM. The other compounds in the series possessed moderate to weak urease inhibition activity with IC50 values ranging from 20.16 ± 0.48 to 60.11 ± 0.78 μM. The most potent compounds 3c and 3g were docked to jack bean urease (PDB ID: 4H9M) to evaluate their binding affinities and to find the plausible binding poses. The docked complexes were refined through 100 ns-long MD simulations. The simulation results revealed that the average RMSD of 3c was less than that of the 3g compound. Furthermore, the radius of gyration plots for both complexes showed that 3c and 3g docking predicted binding modes did not induce any conformational change in the urease structure.
Collapse
Affiliation(s)
- Sana Yaqoob
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi Karachi Pakistan
| | - Abdul Hameed
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi Karachi Pakistan
- Department of Chemistry, University of Sahiwal Sahiwal Pakistan
| | - Mahmood Ahmed
- Department of Chemistry, Division of Science and Technology, University of Education College Road Lahore Pakistan
| | - Muhammad Imran
- KAM-School of Life Sciences, FC College (A Chartered University) Lahore Pakistan
| | | | - Mahwish Ramzan
- Department of Biosciences, COMSATS University Islamabad Park Road Islamabad Pakistan
| | - Numan Yousaf
- Department of Biosciences, COMSATS University Islamabad Park Road Islamabad Pakistan
| | - Jamshed Iqbal
- Center for Advanced Drug Research, COMSATS Institute of Information Technology Abbottabad 22060 Pakistan
| | - Muhammad Muddassar
- Department of Biosciences, COMSATS University Islamabad Park Road Islamabad Pakistan
| |
Collapse
|
35
|
Abbasova G, Medjidov A. One-pot synthesis of the new Hydroxamic acid and its complexes with metals. LETT ORG CHEM 2022. [DOI: 10.2174/1570178619666220111121743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
A one-pot conversion of 2-hydroxy-1-naphthoic aldehyde to hydroxamic acid was described. An efficient photoorganocatalytic method of synthesis was developed. The obtained hydroxamic acid was identified by various physicochemical methods such as IR, UV- and NMR-spectroscopy. Solid colored complexes of copper (II) and iron (II), respectively, green and brown colours with the obtained hydroxamic acid were synthesized in ethanol medium for the first time. The molar ratio of ligand and metal in the complex was 2:1. Their structures were established using IR, UV- spectroscopy and thermogravimetric analysis.
Collapse
Affiliation(s)
- Gulu Abbasova
- ANAS Institute of Catalysis and Inorganic Chemistry is named after Acad. M. F. Nagiyev
АZ 1143 Baku, H. Cavid Avenue, 113, Azerbaijan
| | - Ajdar Medjidov
- ANAS Institute of Catalysis and Inorganic Chemistry is named after Acad. M. F. Nagiyev
АZ 1143 Baku, H. Cavid Avenue, 113, Azerbaijan
| |
Collapse
|
36
|
Design, synthesis, in vitro evaluation, and docking studies on ibuprofen derived 1,3,4-oxadiazole derivatives as dual α-glucosidase and urease inhibitors. Med Chem Res 2022. [DOI: 10.1007/s00044-021-02814-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Kalatuwawege IP, Gunaratna MJ, Udukala DN. Synthesis, In Silico Studies, and Evaluation of Syn and Anti Isomers of N-Substituted Indole-3-carbaldehyde Oxime Derivatives as Urease Inhibitors against Helicobacter pylori. Molecules 2021; 26:molecules26216658. [PMID: 34771067 PMCID: PMC8588131 DOI: 10.3390/molecules26216658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal tract infection caused by Helicobacter pylori is a common virulent disease found worldwide, and the infection rate is much higher in developing countries than in developed ones. In the pathogenesis of H. pylori in the gastrointestinal tract, the secretion of the urease enzyme plays a major role. Therefore, inhibition of urease is a better approach against H. pylori infection. In the present study, a series of syn and anti isomers of N-substituted indole-3-carbaldehyde oxime derivatives was synthesized via Schiff base reaction of appropriate carbaldehyde derivatives with hydroxylamine hydrochloride. The in vitro urease inhibitory activities of those derivatives were evaluated against that of Macrotyloma uniflorum urease using the modified Berthelot reaction. Out of the tested compounds, compound 8 (IC50 = 0.0516 ± 0.0035 mM) and compound 9 (IC50 = 0.0345 ± 0.0008 mM) were identified as the derivatives with potent urease inhibitory activity with compared to thiourea (IC50 = 0.2387 ± 0.0048 mM). Additionally, in silico studies for all oxime compounds were performed to investigate the binding interactions with the active site of the urease enzyme compared to thiourea. Furthermore, the drug-likeness of the synthesized oxime compounds was also predicted.
Collapse
Affiliation(s)
- Ishani P. Kalatuwawege
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya 10100, Sri Lanka; (I.P.K.); (D.N.U.)
| | - Medha J. Gunaratna
- Department of Chemistry, University of Kelaniya, Kelaniya 10300, Sri Lanka
- Correspondence:
| | - Dinusha N. Udukala
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya 10100, Sri Lanka; (I.P.K.); (D.N.U.)
| |
Collapse
|
38
|
Elbastawesy MA, Aly AA, El-Shaier YA, Brown AB, Abuo-Rahma GEDA, Ramadan M. New 4-thiazolidinone/quinoline-2-ones scaffold: Design, synthesis, docking studies and biological evaluation as potential urease inhibitors. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
39
|
Ali Redha A, Valizadenia H, Siddiqui SA, Maqsood S. A state-of-art review on camel milk proteins as an emerging source of bioactive peptides with diverse nutraceutical properties. Food Chem 2021; 373:131444. [PMID: 34717085 DOI: 10.1016/j.foodchem.2021.131444] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023]
Abstract
The generation of camel milk derived bioactive peptides (CM-BAPs) have started to grab keen interest of many researchers during the past decade. CM-BAPs have shown more significant bioactive properties in comparison to camel milk intact proteins. CM-BAPs can be obtained using enzyme hydrolysis to form hydrolysates, or by the fermentation process. In this systematic review, 46 research articles exploring the health-related bioactive properties of CM-BAPs through in-vitro and in-vivo studies have been included. CM-BAPs have been reported for their antioxidant, anti-diabetic, anti-obesity, antihypertensive, antibacterial, antibiofilm, anticancer, anti-inflammatory, anti-haemolytic, and anti-hyperpigmentation activities. The effects of factors such as molecular weight of peptides, type of enzyme, enzyme to substrate ratio, hydrolysis temperature and duration have been analysed. The in-vitro studies have provided enough evidence on certain aspects of the pharmacological actives of camel milk bioactive peptides. Nevertheless, the in-vivo studies are very limited, and no clinical studies on CM-BAPs have been reported.
Collapse
Affiliation(s)
- Ali Ali Redha
- Chemistry Department, School of Science, Loughborough University, Loughborough LE11 3TU, United Kingdom.
| | - Hamidreza Valizadenia
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Semnan Province, Iran
| | - Shahida Anusha Siddiqui
- Technical University of Munich Campus Straubing for Biotechnology and Sustainability, Essigberg 3, 94315 Straubing, Germany; DIL e.V. - German Institute of Food Technologies, D-Quakenbrück, Germany
| | - Sajid Maqsood
- Department of Food Science, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al-Ain 15551, United Arab Emirates; Zayed Centre of Health Science, United Arab Emirates University, Al-Ain 15551, United Arab Emirates.
| |
Collapse
|
40
|
Chelerythrine Chloride: A Potential Rumen Microbial Urease Inhibitor Screened by Targeting UreG. Int J Mol Sci 2021; 22:ijms22158212. [PMID: 34360977 PMCID: PMC8347364 DOI: 10.3390/ijms22158212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 01/17/2023] Open
Abstract
Inhibition of ruminal microbial urease is of particular interest due to its crucial role in regulating urea-N utilization efficiency and nitrogen pollution in the livestock industry. Acetohydroxamic acid (AHA) is currently the only commercially available urease inhibitor, but it has adverse side effects. The urease accessory protein UreG, which facilitates the functional incorporation of the urease nickel metallocentre, has been proposed in developing urease inhibitor through disrupting urease maturation. The objective of this study was to screen natural compounds as potential urease inhibitors by targeting UreG in a predominant ruminal microbial urease. In silico screening and in vitro tests for potential inhibitors were performed using molecular docking and an assay for the GTPase activity of UreG. Chelerythrine chloride was selected as a potential urease inhibitor of UreG with an inhibition concentration IC50 value of 18.13 μM. It exhibited mixed inhibition, with the Ki value being 26.28 μM. We further explored its inhibition mechanism using isothermal titration calorimetry (ITC) and circular dichroism (CD) spectroscopy, and we found that chelerythrine chloride inhibited the binding of nickel to UreG and induced changes in the secondary structure, especially the α-helix and β-sheet of UreG. Chelerythrine chloride formed a pi-anion interaction with the Asp41 residue of UreG, which is an important residue in initiating the conformational changes of UreG. In conclusion, chelerythrine chloride exhibited a potential inhibitory effect on urease, which provided new evidence for strategies to develop novel urease inhibitors targeting UreG to reduce nitrogen excretion from ruminants.
Collapse
|
41
|
Tirmazi SAAS, Qadir MA, Ahmed M, Imran M, Hussain R, Sharif M, Yousaf M, Muddassar M. Levofloxacin and sulfa drugs linked via Schiff bases: Exploring their urease inhibition, enzyme kinetics and in silico studies. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
42
|
Mamidala R, Bhimathati SRS, Vema A. Discovery of Novel Dihydropyrimidine and hydroxamic acid hybrids as potent Helicobacter pylori Urease inhibitors. Bioorg Chem 2021; 114:105010. [PMID: 34102519 DOI: 10.1016/j.bioorg.2021.105010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/25/2022]
Abstract
Two novel series of Dihydropyrimidine-hydroxamic acid hybrids (4a-4l and 5a-5l) were designed, synthesized and evaluated for in vitro Helicobacter pylori urease inhibition. In vitro enzyme inhibition screening led to the discovery of three potent urease inhibitors 2-[[4-(4-hydroxy phenyl)-6-oxo-1,6-dihydropyrimidine-2-yl]-amino]-N-hydroxy acetamide (4g), 2-[[4-(4-chloro phenyl)-6-oxo-1,6-dihydropyrimidine-2-yl]-amino]-N-hydroxy acetamide (4b) and 3-[[4-(3-methoxy phenyl)-6-oxo-1,6-dihydropyrimidine-2-yl]-amino]-N-hydroxy propanamide (5l). Compound 4g showed excellent urease inhibition with IC50 value of 14 ± 1 nM, indicated by its strong interactions with both metallic Ni++ ions, Gly279, His221, Ala365, Asp362, Asn168, Arg338 and His322 residues of the active site of urease. Further, compounds 4b and 5l displayed very good activity with IC50 value of 0.082 ± 0.004 µM and 0.14 ± 0.013 µM respectively compared to standard Acetohydroxamic acid (IC50 - 27.4 ± 1.2 µM). Kinetic studies revealed that a mixed inhibition with both competitive and non-competitive aspects is involved in the urease inhibition mechanism. The in vitro urease inhibition results were supported by molecular docking studies. Collectively, this study indicates that 4g could be considered as promising lead molecule that can be further developed as a potent drug molecule for the treatment of Helicobacter pylori caused gastritis for further studies.
Collapse
Affiliation(s)
- Ravinder Mamidala
- Department of Medicinal Chemistry, St. Peter's Institute of Pharmaceutical Sciences, Hanamkonda, Warangal, Telangana 506001, India; Department of Pharmaceutical Chemistry, Institute of Science and Technology, Jawaharlal Nehru Technological University, Hyderabad, Telangana 500085, India
| | - Solomon Raj S Bhimathati
- Department of Pharmacology, Gland Institute of Pharmaceutical Sciences, Medak, Telangana 502220, India
| | - Aparna Vema
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India.
| |
Collapse
|
43
|
Talebi M, Hamidian E, Niasari-Naslaji F, Rahmani S, Hosseini FS, Boumi S, Montazer MN, Asadi M, Amanlou M. Synthesis, molecular docking, and biological evaluation of nitroimidazole derivatives as potent urease inhibitors. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02727-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
44
|
In vitro study of the antioxidant, photoprotective, anti-tyrosinase, and anti-urease effects of methanolic extracts from leaves of six Moroccan Lamiaceae. JOURNAL OF FOOD MEASUREMENT AND CHARACTERIZATION 2021. [DOI: 10.1007/s11694-020-00759-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
45
|
Affiliation(s)
- Gülay Akyüz
- Department of Chemistry, Art and Science Faculty Recep Tayyip Erdoğan University Rize Turkey
| |
Collapse
|
46
|
Nazari Montazer M, Asadi M, Bahadorikhalili S, Hosseini FS, Amanlou A, Biglar M, Amanlou M. Design, synthesis, docking study and urease inhibitory activity evaluation of novel 2-((5-amino-1,3,4-thiadiazol-2-yl)thio)-N-arylacetamide derivatives. Med Chem Res 2021. [DOI: 10.1007/s00044-020-02683-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
47
|
Cunha ES, Chen X, Sanz-Gaitero M, Mills DJ, Luecke H. Cryo-EM structure of Helicobacter pylori urease with an inhibitor in the active site at 2.0 Å resolution. Nat Commun 2021; 12:230. [PMID: 33431861 PMCID: PMC7801526 DOI: 10.1038/s41467-020-20485-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/04/2020] [Indexed: 02/08/2023] Open
Abstract
Infection of the human stomach by Helicobacter pylori remains a worldwide problem and greatly contributes to peptic ulcer disease and gastric cancer. Without active intervention approximately 50% of the world population will continue to be infected with this gastric pathogen. Current eradication, called triple therapy, entails a proton-pump inhibitor and two broadband antibiotics, however resistance to either clarithromycin or metronidazole is greater than 25% and rising. Therefore, there is an urgent need for a targeted, high-specificity eradication drug. Gastric infection by H. pylori depends on the expression of a nickel-dependent urease in the cytoplasm of the bacteria. Here, we report the 2.0 Å resolution structure of the 1.1 MDa urease in complex with an inhibitor by cryo-electron microscopy and compare it to a β-mercaptoethanol-inhibited structure at 2.5 Å resolution. The structural information is of sufficient detail to aid in the development of inhibitors with high specificity and affinity.
Collapse
Affiliation(s)
- Eva S. Cunha
- grid.5510.10000 0004 1936 8921Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
| | - Xiaorui Chen
- grid.266093.80000 0001 0668 7243Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697 USA ,grid.506938.10000 0004 0633 8088Present Address: Genomics Research Center, Academia Sinica, 128 Academia Road, Sect. 2, Nankang District, Taipei, Taiwan
| | - Marta Sanz-Gaitero
- grid.5510.10000 0004 1936 8921Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, 0318 Oslo, Norway
| | - Deryck J. Mills
- grid.419494.50000 0001 1018 9466Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Hartmut Luecke
- Structural Biology and Drug Discovery Group, Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, 0318, Oslo, Norway. .,Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, USA. .,Department of Medical Biochemistry, University of Oslo and Oslo University Hospital, 0372, Oslo, Norway. .,Department of Physiology and Biophysics, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
48
|
Pagoni A, Grabowiecka A, Tabor W, Mucha A, Vassiliou S, Berlicki Ł. Covalent Inhibition of Bacterial Urease by Bifunctional Catechol-Based Phosphonates and Phosphinates. J Med Chem 2020; 64:404-416. [PMID: 33369409 DOI: 10.1021/acs.jmedchem.0c01143] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this study, a new class of bifunctional inhibitors of bacterial ureases, important molecular targets for antimicrobial therapies, was developed. The structures of the inhibitors consist of a combination of a phosphonate or (2-carboxyethyl)phosphinate functionality with a catechol-based fragment, which are designed for complexation of the catalytic nickel ions and covalent bonding with the thiol group of Cys322, respectively. Compounds with three types of frameworks, including β-3,4-dihydroxyphenyl-, α-3,4-dihydroxybenzyl-, and α-3,4-dihydroxybenzylidene-substituted derivatives, exhibited complex and varying structure-dependent kinetics of inhibition. Among irreversible binders, methyl β-(3,4-dihydroxyphenyl)-β-(2-carboxyethyl)phosphorylpropionate was observed to be a remarkably reactive inhibitor of Sporosarcina pasteurii urease (kinact/KI = 10 420 s-1 M-1). The high potential of this group of compounds was also confirmed in Proteus mirabilis whole-cell-based inhibition assays. Some compounds followed slow-binding and reversible kinetics, e.g., methyl β-(3,4-dihydroxyphenyl)-β-phosphonopropionate, with Ki* = 0.13 μM, and an atypical low dissociation rate (residence time τ = 205 min).
Collapse
Affiliation(s)
- Aikaterini Pagoni
- Laboratory of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, Zografou, 15701 Athens, Greece
| | - Agnieszka Grabowiecka
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Wojciech Tabor
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Artur Mucha
- 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, 15701 Athens, Greece
| | - Łukasz Berlicki
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| |
Collapse
|
49
|
Rashid M, Rafique H, Roshan S, Shamas S, Iqbal Z, Ashraf Z, Abbas Q, Hassan M, Qureshi ZUR, Asad MHHB. Enzyme Inhibitory Kinetics and Molecular Docking Studies of Halo-Substituted Mixed Ester/Amide-Based Derivatives as Jack Bean Urease Inhibitors. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8867407. [PMID: 33426080 PMCID: PMC7775144 DOI: 10.1155/2020/8867407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/28/2020] [Accepted: 12/15/2020] [Indexed: 12/22/2022]
Abstract
A series of halo-substituted mixed ester/amide-based analogues 4a-l have been prepared as jack bean urease inhibitor, which showed good to excellent inhibition of enzyme activity. The role of halo-substituted benzoyl moieties and alkyl substituted anilines in urease inhibitory kinetics was also investigated. The alkyl-substituted anilines 1a-b reacted with chloroacetyl chloride to afford intermediates 2a-b, which were then reacted with different halo-substituted benzoic acids 3a-f to prepare the title compounds 4a-l. The chemical structures of final products 4a-l were ascertained by FTIR, 1H NMR, 13C NMR, and mass spectra. The compound 4b showed remarkable activity with IC501.6 ± 0.2 nM, better than the standard thiourea having IC50472.1 ± 135.1 nM. The 2-chloro-substituted phenyl ring on one side of compound 4b and 4-isopropyl-substituted benzene on the other side play an essential role in inhibition of urease activity. Lineweaver-Burk plots (kinetics study) indicated about 4b derivative as a mixed type of inhibitor. The virtual screening performed against urease enzyme (PDBID 4H9M) showed that compounds 4b and 4e have binding energies of -7.8 and -7.9 Kcal/mol, respectively. Based upon our results, it was found that derivative 4b is a highly potent urease inhibitor, better than the standard thiourea.
Collapse
Affiliation(s)
- Muhammad Rashid
- Department of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan
| | - Hummera Rafique
- Department of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | - Sadia Roshan
- Department of Zoology, University of Gujrat, Gujrat 50700, Pakistan
| | - Shazia Shamas
- Department of Zoology, University of Gujrat, Gujrat 50700, Pakistan
| | - Zafar Iqbal
- Department of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan
| | - Zaman Ashraf
- Department of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan
| | - Qamar Abbas
- Department of Physiology, University of Sindh, Jamshoro, Pakistan
| | - Mubashir Hassan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | | | - Muhammad Hassham Hassan Bin Asad
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Pakistan
- Institute of Fundamental Medicine, Department of Genetics, Kazan Federal University, Russia
| |
Collapse
|
50
|
Wang Y, Wang G, Moitessier N, Mittermaier AK. Enzyme Kinetics by Isothermal Titration Calorimetry: Allostery, Inhibition, and Dynamics. Front Mol Biosci 2020; 7:583826. [PMID: 33195429 PMCID: PMC7604385 DOI: 10.3389/fmolb.2020.583826] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Isothermal titration calorimetry (ITC) involves accurately measuring the heat that is released or absorbed in real time when one solution is titrated into another. This technique is usually used to measure the thermodynamics of binding reactions. However, there is mounting interest in using it to measure reaction kinetics, particularly enzymatic catalysis. This application of ITC has been steadily growing for the past two decades, and the method is proving to be sensitive, generally applicable, and capable of providing information on enzyme activity that is difficult to obtain using traditional biochemical assays. This review aims to give a broad overview of the use of ITC to measure enzyme kinetics. It describes several different classes of ITC experiment, their strengths and weaknesses, and recent methodological advancements. A summary of applications in the literature is given and several examples where ITC has been used to investigate challenging aspects of enzyme behavior are presented in more detail. These include examples of allostery, where small-molecule binding outside the active site modulates activity. We describe the use of ITC to measure the strength, mode (i.e., competitive, uncompetitive, or mixed), and association and dissociation kinetics of enzyme inhibitors. Further, we provide examples of ITC applied to complex, heterogeneous mixtures, such as insoluble substrates and live cells. These studies exemplify the wide range of problems where ITC can provide answers, and illustrate the versatility of the technique and potential for future development and applications.
Collapse
Affiliation(s)
- Yun Wang
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | - Guanyu Wang
- Department of Chemistry, McGill University, Montreal, QC, Canada
| | | | | |
Collapse
|