Epoxy-Functionalized Isatin Derivative: Synthesis, Computational Evaluation, and Antibacterial Analysis.

IF 4.3 2区 医学 Q1 INFECTIOUS DISEASES
Deepanjali Shukla, Iqbal Azad, Mohd Arsh Khan, Ziaul Husain, Azhar Kamal, Sabahat Yasmeen Sheikh, Ibrahim Alotibi, Varish Ahmad, Firoj Hassan
{"title":"Epoxy-Functionalized Isatin Derivative: Synthesis, Computational Evaluation, and Antibacterial Analysis.","authors":"Deepanjali Shukla, Iqbal Azad, Mohd Arsh Khan, Ziaul Husain, Azhar Kamal, Sabahat Yasmeen Sheikh, Ibrahim Alotibi, Varish Ahmad, Firoj Hassan","doi":"10.3390/antibiotics14060595","DOIUrl":null,"url":null,"abstract":"<p><strong>Background/objectives: </strong>The current need for new antibacterial compounds that target non-classical pathways is highlighted by the emergence of multidrug-resistant <i>Klebsiella pneumoniae</i>. In the development of antibiotics, DNA adenine methyltransferase (Dam), a key regulator of bacterial gene expression and pathogenicity, is still underutilized. Epoxy-functionalized analogues of isatin derivatives have not been adequately investigated for their antibacterial activity, particularly as Dam inhibitors. In the pursuit of antimicrobial agents, this study synthesized an epoxy-functionalized isatin derivative (<b>L3</b>) using a one-pot reaction. The compound was characterized using FT-IR, ¹H-NMR, <sup>13</sup>C-NMR, HR-MS, and UV-Vis spectroscopy.</p><p><strong>Methods: </strong><i>In silico</i> evaluation performed by using ADMETlab3 and SwissADME. While molecular docking studies were achieved by AutoDock and Vina to find <b>L3</b>'s interaction with potential antibacterial target (Dam protein in <i>K. pneumoniae</i>). In addition, the antibacterial potential of <b>L3</b> was evaluated using minimum inhibitory concentration (MIC) assays against <i>Bacillus cereus</i>, <i>Bacillus pumilus</i>, <i>Escherichia coli</i>, and <i>K. pneumoniae</i>.</p><p><strong>Results: </strong>Among these, <b>L3</b> exhibited potential inhibitory activity against <i>K. pneumoniae</i>, with a MIC value of 93.75 μg/mL. <i>In silico</i> evaluations confirmed <b>L3</b>'s favorable drug-like properties, including potential oral bioavailability, blood-brain barrier (BBB) permeability, and low plasma protein binding (PPB). The compound satisfied Lipinski's and other drug-likeness rules as well as getting a quantitative estimate of drug-likeness (QED) score of 0.52. Here, a homology model of Dam protein in <i>K. pneumoniae</i> was generated using the SWISS-MODEL server and validated using computational tools. Targeted docking analysis revealed that <b>L3</b> exhibited significant potential binding affinity against Dam protein, with binding energies of -6.4 kcal/mol and -4.85 kcal/mol, as determined by Vina and AutoDock, respectively. The associated inhibition constant was calculated as 280.35 µM. Further interaction analysis identified the formation of hydrogen bonds with TRP7 and PHE32, along with Van der Waals' interactions involving GLY9, ASP51, and ASP179.</p><p><strong>Conclusions: </strong>These findings highlight <b>L3</b> as a promising scaffold for antimicrobial drug development, particularly in targeting Dam protein in <i>K. pneumoniae</i>. Furthermore, the ADMET profiling and physicochemical properties of <b>L3</b> support its potential as a drug-like candidate.</p>","PeriodicalId":54246,"journal":{"name":"Antibiotics-Basel","volume":"14 6","pages":""},"PeriodicalIF":4.3000,"publicationDate":"2025-06-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12189316/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Antibiotics-Basel","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.3390/antibiotics14060595","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"INFECTIOUS DISEASES","Score":null,"Total":0}
引用次数: 0

Abstract

Background/objectives: The current need for new antibacterial compounds that target non-classical pathways is highlighted by the emergence of multidrug-resistant Klebsiella pneumoniae. In the development of antibiotics, DNA adenine methyltransferase (Dam), a key regulator of bacterial gene expression and pathogenicity, is still underutilized. Epoxy-functionalized analogues of isatin derivatives have not been adequately investigated for their antibacterial activity, particularly as Dam inhibitors. In the pursuit of antimicrobial agents, this study synthesized an epoxy-functionalized isatin derivative (L3) using a one-pot reaction. The compound was characterized using FT-IR, ¹H-NMR, 13C-NMR, HR-MS, and UV-Vis spectroscopy.

Methods: In silico evaluation performed by using ADMETlab3 and SwissADME. While molecular docking studies were achieved by AutoDock and Vina to find L3's interaction with potential antibacterial target (Dam protein in K. pneumoniae). In addition, the antibacterial potential of L3 was evaluated using minimum inhibitory concentration (MIC) assays against Bacillus cereus, Bacillus pumilus, Escherichia coli, and K. pneumoniae.

Results: Among these, L3 exhibited potential inhibitory activity against K. pneumoniae, with a MIC value of 93.75 μg/mL. In silico evaluations confirmed L3's favorable drug-like properties, including potential oral bioavailability, blood-brain barrier (BBB) permeability, and low plasma protein binding (PPB). The compound satisfied Lipinski's and other drug-likeness rules as well as getting a quantitative estimate of drug-likeness (QED) score of 0.52. Here, a homology model of Dam protein in K. pneumoniae was generated using the SWISS-MODEL server and validated using computational tools. Targeted docking analysis revealed that L3 exhibited significant potential binding affinity against Dam protein, with binding energies of -6.4 kcal/mol and -4.85 kcal/mol, as determined by Vina and AutoDock, respectively. The associated inhibition constant was calculated as 280.35 µM. Further interaction analysis identified the formation of hydrogen bonds with TRP7 and PHE32, along with Van der Waals' interactions involving GLY9, ASP51, and ASP179.

Conclusions: These findings highlight L3 as a promising scaffold for antimicrobial drug development, particularly in targeting Dam protein in K. pneumoniae. Furthermore, the ADMET profiling and physicochemical properties of L3 support its potential as a drug-like candidate.

环氧功能化Isatin衍生物:合成、计算评价和抗菌分析。
背景/目的:多药耐药肺炎克雷伯菌的出现突出了当前对针对非经典途径的新型抗菌化合物的需求。在抗生素的开发中,DNA腺嘌呤甲基转移酶(DNA adenine methyltransferase, Dam)作为细菌基因表达和致病性的关键调控因子尚未得到充分利用。环氧功能化的isatin衍生物类似物尚未充分研究其抗菌活性,特别是作为Dam抑制剂。在抗菌药物的研究中,本研究采用一锅反应合成了环氧功能化的isatin衍生物(L3)。采用FT-IR、¹H-NMR、13C-NMR、HR-MS和UV-Vis光谱对化合物进行了表征。方法:采用ADMETlab3和SwissADME进行计算机评价。而通过AutoDock和Vina进行分子对接研究,发现L3与潜在抗菌靶点(肺炎克雷伯菌中的Dam蛋白)的相互作用。此外,通过最低抑菌浓度(MIC)测定对蜡样芽孢杆菌、短小芽孢杆菌、大肠杆菌和肺炎克雷伯菌的抑菌潜力进行了评估。结果:其中L3对肺炎克雷伯菌表现出潜在的抑制活性,MIC值为93.75 μg/mL。计算机评估证实L3具有良好的药物样特性,包括潜在的口服生物利用度、血脑屏障(BBB)渗透性和低血浆蛋白结合(PPB)。该化合物满足Lipinski和其他药物相似规则,并且药物相似度(QED)的定量估计得分为0.52。本文使用SWISS-MODEL服务器生成了肺炎克雷伯菌中Dam蛋白的同源模型,并使用计算工具进行了验证。靶向对接分析表明,L3对Dam蛋白具有显著的潜在结合亲和力,根据Vina和AutoDock测定,L3的结合能分别为-6.4 kcal/mol和-4.85 kcal/mol。计算出相关抑制常数为280.35µM。进一步的相互作用分析发现了与TRP7和PHE32形成的氢键,以及涉及GLY9、ASP51和ASP179的范德华相互作用。结论:这些发现突出了L3作为抗微生物药物开发的有前途的支架,特别是针对肺炎克雷伯菌的Dam蛋白。此外,L3的ADMET分析和物理化学性质支持其作为类似药物的候选药物的潜力。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Antibiotics-Basel
Antibiotics-Basel Pharmacology, Toxicology and Pharmaceutics-General Pharmacology, Toxicology and Pharmaceutics
CiteScore
7.30
自引率
14.60%
发文量
1547
审稿时长
11 weeks
期刊介绍: Antibiotics (ISSN 2079-6382) is an open access, peer reviewed journal on all aspects of antibiotics. Antibiotics is a multi-disciplinary journal encompassing the general fields of biochemistry, chemistry, genetics, microbiology and pharmacology. Our aim is to encourage scientists to publish their experimental and theoretical results in as much detail as possible. Therefore, there is no restriction on the length of papers.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信