首页 > 最新文献

Journal of Biomolecular Structure & Dynamics最新文献

英文 中文
In-silico investigation of E8 surface protein of the monkeypox virus to identify potential therapeutic agents. 通过对猴痘病毒 E8 表面蛋白的分子内研究,确定潜在的治疗药物。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-09 DOI: 10.1080/07391102.2023.2245041
Shourya Podduturi, Divya Vemula, Siva Singothu, Vasundhra Bhandari

The re-emergence of the monkeypox virus (MPXV) in 2022 has become a global issue. The virus was first found in Denmark in 1958. The first human MPXV disease was reported in 1980 in Congo, caused by a rare zoonotic virus belonging to the genus Orthopoxvirus and the family Poxviridae. Like SARS-CoV, there are no specific drugs to treat this infection. Taking cues from the successful implementation of drug repositioning for the Covid-19 pandemic using in silico drug discovery. We employed structure-based drug design in the study to repurpose the existing drug and natural product derivatives libraries against MPXV. The E8 protein was chosen as a therapeutic target because it is a surface membrane protein involved in viral entry and adhesion to the host cell surface membrane. Our study was bifurcated into the following steps; determining and analyzing the structure of the E8, followed by structure-based virtual screening of different datasets (natural products obtained from bacteria and fungi and FDA-approved drugs) to identify the hits. Based on the best binding affinities and protein-ligand interactions, we further proceeded for molecular dynamic (MD) studies of the identified hits, which revealed Gabosine D (docking score = -8.469 kcal/mol, MM/GBSA dG bind = -41.6729 kcal/mol) and Edoxudine (docking score = -6.372 kcal/mol, MM/GBSA dG bind = -35.8291 kcal/mol) as the best lead molecules. MD simulation for 100 ns was performed in triplicate, and post MM/GBSA analysis was conducted, which proves the stability of the identified leads. In addition, their ADME profiles also confirmed their suitability as therapeutic options for the treatment of monkeypox.Communicated by Ramaswamy H. Sarma.

2022 年再次出现的猴痘病毒(MPXV)已成为一个全球性问题。该病毒于 1958 年首次在丹麦发现。1980 年,刚果报告了第一例人类 MPXV 疾病,它是由一种罕见的人畜共患病毒引起的,这种病毒属于正痘病毒属和痘病毒科。与 SARS-CoV 一样,目前也没有治疗这种感染的特效药物。我们借鉴了针对 Covid-19 大流行病成功实施药物重新定位的经验,使用了硅库药物发现技术。我们在研究中采用了基于结构的药物设计,将现有的药物和天然产物衍生物库重新用于抗击 MPXV。之所以选择 E8 蛋白作为治疗靶点,是因为它是一种表面膜蛋白,参与病毒的进入和与宿主细胞表面膜的粘附。我们的研究分为以下几个步骤:确定和分析 E8 的结构,然后对不同的数据集(从细菌和真菌中获得的天然产物以及美国食品与药物管理局批准的药物)进行基于结构的虚拟筛选,以确定命中目标。根据最佳结合亲和力和蛋白质-配体相互作用,我们进一步对确定的命中物进行了分子动力学(MD)研究,结果发现加博新 D(对接得分 = -8.469 kcal/mol,MM/GBSA dG bind = -41.6729 kcal/mol)和埃多库定(对接得分 = -6.372 kcal/mol,MM/GBSA dG bind = -35.8291 kcal/mol)是最佳先导分子。一式三份进行了 100 ns 的 MD 模拟,并进行了 MM/GBSA 后分析,这证明了所确定的先导分子的稳定性。此外,它们的 ADME 图谱也证实了它们适合作为治疗猴痘的治疗方案。
{"title":"<i>In-silico</i> investigation of E8 surface protein of the monkeypox virus to identify potential therapeutic agents.","authors":"Shourya Podduturi, Divya Vemula, Siva Singothu, Vasundhra Bhandari","doi":"10.1080/07391102.2023.2245041","DOIUrl":"10.1080/07391102.2023.2245041","url":null,"abstract":"<p><p>The re-emergence of the monkeypox virus (MPXV) in 2022 has become a global issue. The virus was first found in Denmark in 1958. The first human MPXV disease was reported in 1980 in Congo, caused by a rare zoonotic virus belonging to the genus Orthopoxvirus and the family Poxviridae. Like SARS-CoV, there are no specific drugs to treat this infection. Taking cues from the successful implementation of drug repositioning for the Covid-19 pandemic using in silico drug discovery. We employed structure-based drug design in the study to repurpose the existing drug and natural product derivatives libraries against MPXV. The E8 protein was chosen as a therapeutic target because it is a surface membrane protein involved in viral entry and adhesion to the host cell surface membrane. Our study was bifurcated into the following steps; determining and analyzing the structure of the E8, followed by structure-based virtual screening of different datasets (natural products obtained from bacteria and fungi and FDA-approved drugs) to identify the hits. Based on the best binding affinities and protein-ligand interactions, we further proceeded for molecular dynamic (MD) studies of the identified hits, which revealed Gabosine D (docking score = -8.469 kcal/mol, MM/GBSA dG bind = -41.6729 kcal/mol) and Edoxudine (docking score = -6.372 kcal/mol, MM/GBSA dG bind = -35.8291 kcal/mol) as the best lead molecules. MD simulation for 100 ns was performed in triplicate, and post MM/GBSA analysis was conducted, which proves the stability of the identified leads. In addition, their ADME profiles also confirmed their suitability as therapeutic options for the treatment of monkeypox.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9960709","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A computational investigation of thymidylate synthase inhibitors through a combined approach of 3D-QSAR and pharmacophore modelling. 通过3D-QSAR和药效团建模的联合方法对胸苷酸合成酶抑制剂进行的计算研究。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-10-23 DOI: 10.1080/07391102.2023.2270752
Sonu Benny, Prayaga Rajappan Krishnendu, Sunil Kumar, Vaishnav Bhaskar, Deepthi S Manisha, Mohamed A Abdelgawad, Mohammed M Ghoneim, Ibrahim A Naguib, Leena K Pappachen, Subin Mary Zachariah, Bijo Mathew, Aneesh Tp

Thymidylate synthase (TS) is a crucial target of cancer drug discovery and is mainly involved in the De novo synthesis of the DNA precursor thymine. In the present study, to generate reliable models and identify a few promising molecules, we combined QSAR modelling with the pharmacophore hypothesis-generating technique. Input molecules were clustered on their similarity, and a cluster of 74 molecules with a pyrimidine moiety was chosen as the set for 3D-QSAR and pharmacophore modelling. Atom-based and field-based 3D-QSAR models were generated and statistically validated with R2 > 0.90 and Q2 > 0.75. The common pharmacophore hypothesis(CPH) generation identified the best six-point model ADHRRR. Using these best models, a library of FDA-approved drugs was screened for activity and filtered via molecular docking, ADME profiling, and molecular dynamics simulations. The top ten promising TS-inhibiting candidates were identified, and their chemical features profitable for TS inhibitors were explored.Communicated by Ramaswamy H. Sarma.

胸苷酸合成酶(TS)是癌症药物发现的重要靶点,主要参与DNA前体胸腺嘧啶的从头合成。在本研究中,为了生成可靠的模型并鉴定一些有前景的分子,我们将QSAR建模与药效团假说生成技术相结合。根据输入分子的相似性对其进行聚类,并选择具有嘧啶部分的74个分子的聚类作为3D-QSAR和药效团建模的集合。生成了基于原子和基于场的3D-QSAR模型,并在R2>0.90和Q2>0.75的情况下进行了统计验证。共同药效团假说(CPH)生成确定了最佳的六点模型ADHRRR。使用这些最佳模型,对美国食品药品监督管理局批准的药物库进行活性筛选,并通过分子对接、ADME图谱和分子动力学模拟进行过滤。确定了十大有前景的TS抑制剂候选物,并探索了其对TS抑制剂有益的化学特征。Ramaswamy H.Sarma通讯。
{"title":"A computational investigation of thymidylate synthase inhibitors through a combined approach of 3D-QSAR and pharmacophore modelling.","authors":"Sonu Benny, Prayaga Rajappan Krishnendu, Sunil Kumar, Vaishnav Bhaskar, Deepthi S Manisha, Mohamed A Abdelgawad, Mohammed M Ghoneim, Ibrahim A Naguib, Leena K Pappachen, Subin Mary Zachariah, Bijo Mathew, Aneesh Tp","doi":"10.1080/07391102.2023.2270752","DOIUrl":"10.1080/07391102.2023.2270752","url":null,"abstract":"<p><p>Thymidylate synthase (TS) is a crucial target of cancer drug discovery and is mainly involved in the <i>De novo</i> synthesis of the DNA precursor thymine. In the present study, to generate reliable models and identify a few promising molecules, we combined QSAR modelling with the pharmacophore hypothesis-generating technique. Input molecules were clustered on their similarity, and a cluster of 74 molecules with a pyrimidine moiety was chosen as the set for 3D-QSAR and pharmacophore modelling. Atom-based and field-based 3D-QSAR models were generated and statistically validated with R<sup>2</sup> > 0.90 and Q<sup>2</sup> > 0.75. The common pharmacophore hypothesis(CPH) generation identified the best six-point model ADHRRR. Using these best models, a library of FDA-approved drugs was screened for activity and filtered <i>via</i> molecular docking, ADME profiling, and molecular dynamics simulations. The top ten promising TS-inhibiting candidates were identified, and their chemical features profitable for TS inhibitors were explored.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49690693","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Suppression of human lysozyme aggregation by a novel copper-based complex of 3,4-dimethoxycinnamic acid. 3,4-二甲氧基肉桂酸的新型铜基复合物抑制人类溶菌酶的聚集。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-14 DOI: 10.1080/07391102.2023.2246567
Sumra Dilshad, Shabnam, Arif Ali, Shama Firdaus, Musheer Ahmad, Aiman Ahmad

In this work, a new Cu(II)-based complex as a chemotherapeutic drug agent, formulated as[Cu(DCA)4(H2O)2]⋅4H2O⋅4MeOH, (DCA = 3,4-dimethoxycinnamic acid), namely 1 was successfully synthesized utilizing DCA as a ligand to arrest fibrillation in Human lysozyme. The 1 was thoroughly characterized by single crystal X-ray diffraction (SC-XRD), spectroscopic (UV-Vis and FTIR) techniques, PXRD, and TGA analysis. Its crystal structure reveals a paddle wheel network around central copper metal ions. The Cu(II) metal ions exhibit a distorted square pyramidal configuration. The fluorescence titration studies showed moderate binding interaction of 1 with HuL with Ka of 6.3x105 M-1 at pH-2, 25 °C due to its interaction withAsp53, Tyr63, Val110, and Ala111 as shown by docking and simulation studies. 1suppresses the HuL fibrillation in a concentration-dependent manner, as demonstrated by ThT assay. At 200 µM concentration, it leads to the formation of smaller species of the protein in comparison to the control sample, as suggested by Light Scattering studies. The species formed are less hydrophobic and retain their native α-helix structure compared to the control samples, which are hydrophobic and form β-sheet rich amyloids as shown by ANS hydrophobicity assay and CD spectroscopy, respectively. Furthermore, morphological analysis of the species by AFM has demonstrated that, unlike mature amyloid fibrils in the control sample, HuL forms small-size aggregates in the presence of 1 under similar fibrillation conditions. It can be concluded that 1 effectively suppresses HuL fibrillation due to moderate binding to the protein.Communicated by Ramaswamy H. Sarma.

在这项工作中,成功合成了一种新的铜(II)基配合物作为化疗药物,其配体为[Cu(DCA)4(H2O)2]⋅4H2O⋅4MeOH(DCA = 3,4-二甲氧基肉桂酸),即利用 DCA 作为配体来抑制人类溶菌酶的纤维化。通过单晶 X 射线衍射 (SC-XRD)、光谱(紫外可见光和傅立叶变换红外光谱)技术、PXRD 和 TGA 分析,对 1 进行了全面的表征。其晶体结构显示出围绕中心铜金属离子的桨轮网络。铜(II)金属离子呈扭曲的方形金字塔构型。荧光滴定研究表明,在 pH 值为 2、温度为 25 ℃ 时,1 与 HuL 的结合力中等,其 Ka 值为 6.3x105 M-1,这是由于对接和模拟研究表明 1 与 Asp53、Tyr63、Val110 和 Ala111 发生了相互作用。ThT 试验表明,1 能以浓度依赖性方式抑制 HuL 纤维化。光散射研究表明,在 200 µM 浓度下,与对照样品相比,它能导致形成更小的蛋白质种类。ANS 疏水性检测法和 CD 光谱法分别显示,与对照样品相比,所形成的物种疏水性较低,并保留了其原生的 α-helix 结构;与对照样品相比,所形成的物种疏水性较高,并形成了富含 β-sheet 的淀粉样。此外,用原子力显微镜对这些物种进行的形态分析表明,与对照样本中成熟的淀粉样纤维不同,在类似的纤化条件下,HuL 在 1 的存在下会形成小尺寸的聚集体。由此可以得出结论,由于 1 与蛋白质的结合适度,它能有效抑制 HuL 的纤维化。
{"title":"Suppression of human lysozyme aggregation by a novel copper-based complex of 3,4-dimethoxycinnamic acid.","authors":"Sumra Dilshad, Shabnam, Arif Ali, Shama Firdaus, Musheer Ahmad, Aiman Ahmad","doi":"10.1080/07391102.2023.2246567","DOIUrl":"10.1080/07391102.2023.2246567","url":null,"abstract":"<p><p>In this work, a new Cu(II)-based complex as a chemotherapeutic drug agent, formulated as[Cu(DCA)<sub>4</sub>(H<sub>2</sub>O)<sub>2</sub>]⋅4H<sub>2</sub>O⋅4MeOH, (DCA = 3,4-dimethoxycinnamic acid), namely <b>1</b> was successfully synthesized utilizing DCA as a ligand to arrest fibrillation in Human lysozyme. The <b>1</b> was thoroughly characterized by single crystal X-ray diffraction (SC-XRD), spectroscopic (UV-Vis and FTIR) techniques, PXRD, and TGA analysis. Its crystal structure reveals a paddle wheel network around central copper metal ions. The Cu(II) metal ions exhibit a distorted square pyramidal configuration. The fluorescence titration studies showed moderate binding interaction of <b>1</b> with HuL with Ka of 6.3x10<sup>5</sup> M<sup>-1</sup> at pH-2, 25 °C due to its interaction withAsp53, Tyr63, Val110, and Ala111 as shown by docking and simulation studies. <b>1</b>suppresses the HuL fibrillation in a concentration-dependent manner, as demonstrated by ThT assay. At 200 µM concentration, it leads to the formation of smaller species of the protein in comparison to the control sample, as suggested by Light Scattering studies. The species formed are less hydrophobic and retain their native α-helix structure compared to the control samples, which are hydrophobic and form β-sheet rich amyloids as shown by ANS hydrophobicity assay and CD spectroscopy, respectively. Furthermore, morphological analysis of the species by AFM has demonstrated that, unlike mature amyloid fibrils in the control sample, HuL forms small-size aggregates in the presence of <b>1</b> under similar fibrillation conditions. It can be concluded that <b>1</b> effectively suppresses HuL fibrillation due to moderate binding to the protein.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10343668","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fragment-based structural exploration and chemico-biological interaction study of HDAC3 inhibitors through non-linear pattern recognition, chemical space, and binding mode of interaction analysis. 通过非线性模式识别、化学空间和相互作用结合模式分析,对 HDAC3 抑制剂进行基于片段的结构探索和化学生物相互作用研究。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-23 DOI: 10.1080/07391102.2023.2248509
Suvankar Banerjee, Shraddha Dumawat, Tarun Jha, Goverdhan Lanka, Nilanjan Adhikari, Balaram Ghosh

HDAC3 is an emerging target for the identification and discovery of novel drug candidates against several disease conditions including cancer. Here, a fragment-based non-linear machine learning (ML) method along with chemical space exploration followed by a structure-based binding mode of interaction analysis study was carried out on some HDAC3 inhibitors to obtain the key structural features modulating HDAC3 inhibition. Both the ML and chemical space analysis identified several physicochemical and structural properties namely lipophilicity, polar and relative polar surface area, arylcarboxamide moiety, bulky fused aromatic group, n-alkyl, and cinnamoyl moieties, the higher number of oxygen atoms, π-electrons for the substituted tetrahydrofuronaphthodioxolone moiety favorable for higher HDAC3 inhibition. Moreover, hydrogen bond forming capabilities, the length and substitution position of the linker moiety, the importance of phenyl ring in the linker motif, the contribution of heterocyclic cap moieties for effective inhibitor binding at the HDAC3 catalytic site that correspondingly affects the HDAC3 inhibitory potency. Again, macrocyclic ring structure and cyclohexyl cap moiety are responsible for lower HDAC3 inhibition. The MD simulation study of selected compounds explained strong binding patterns at the HDAC3 active site as evidenced by the lower RMSD and RMSF values. Nevertheless, it also explained the importance of the crucial structural fragments derived from the fragment-based analysis during ligand-enzyme interactions. Therefore, the outcomes of this current structural analysis will be a useful tool for fragment-based drug discovery of effective HDAC3 inhibitors for clinical therapeutics in the future.Communicated by Ramaswamy H. Sarma.

HDAC3 是针对包括癌症在内的多种疾病鉴定和发现新型候选药物的新兴靶点。在此,我们对一些HDAC3抑制剂进行了基于片段的非线性机器学习(ML)方法和化学空间探索,以及基于结构的相互作用结合模式分析研究,以获得调节HDAC3抑制作用的关键结构特征。通过 ML 分析和化学空间分析,确定了几种物理化学和结构特性,即亲脂性、极性和相对极性表面积、芳基甲酰胺分子、膨大的融合芳基、正烷基和肉桂酰基、较多的氧原子数目、取代的四氢呋喃萘并二氧戊环分子的 π 电子,这些特性有利于提高 HDAC3 抑制作用。此外,氢键形成能力、连接分子的长度和取代位置、苯基环在连接基团中的重要性、杂环盖分子对抑制剂在 HDAC3 催化位点有效结合的贡献,都会相应地影响 HDAC3 的抑制效力。同样,大环环结构和环己基帽子分子对 HDAC3 的抑制作用较低。对所选化合物进行的 MD 模拟研究解释了 HDAC3 活性位点的强结合模式,较低的 RMSD 值和 RMSF 值证明了这一点。不过,这也说明了基于片段分析得出的关键结构片段在配体与酶相互作用过程中的重要性。因此,目前的结构分析结果将成为基于片段的药物发现有效 HDAC3 抑制剂的有用工具,在未来用于临床治疗。
{"title":"Fragment-based structural exploration and chemico-biological interaction study of HDAC3 inhibitors through non-linear pattern recognition, chemical space, and binding mode of interaction analysis.","authors":"Suvankar Banerjee, Shraddha Dumawat, Tarun Jha, Goverdhan Lanka, Nilanjan Adhikari, Balaram Ghosh","doi":"10.1080/07391102.2023.2248509","DOIUrl":"10.1080/07391102.2023.2248509","url":null,"abstract":"<p><p>HDAC3 is an emerging target for the identification and discovery of novel drug candidates against several disease conditions including cancer. Here, a fragment-based non-linear machine learning (ML) method along with chemical space exploration followed by a structure-based binding mode of interaction analysis study was carried out on some HDAC3 inhibitors to obtain the key structural features modulating HDAC3 inhibition. Both the ML and chemical space analysis identified several physicochemical and structural properties namely lipophilicity, polar and relative polar surface area, arylcarboxamide moiety, bulky fused aromatic group, <i>n</i>-alkyl, and cinnamoyl moieties, the higher number of oxygen atoms, π-electrons for the substituted tetrahydrofuronaphthodioxolone moiety favorable for higher HDAC3 inhibition. Moreover, hydrogen bond forming capabilities, the length and substitution position of the linker moiety, the importance of phenyl ring in the linker motif, the contribution of heterocyclic cap moieties for effective inhibitor binding at the HDAC3 catalytic site that correspondingly affects the HDAC3 inhibitory potency. Again, macrocyclic ring structure and cyclohexyl cap moiety are responsible for lower HDAC3 inhibition. The MD simulation study of selected compounds explained strong binding patterns at the HDAC3 active site as evidenced by the lower RMSD and RMSF values. Nevertheless, it also explained the importance of the crucial structural fragments derived from the fragment-based analysis during ligand-enzyme interactions. Therefore, the outcomes of this current structural analysis will be a useful tool for fragment-based drug discovery of effective HDAC3 inhibitors for clinical therapeutics in the future.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10051377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In silico identification of α-bisabolol and letestuianin C as potential inhibitors of Trypanosoma brucei trypanothione reductase. 硅学鉴定α-双香叶醇和letestuianin C作为布氏锥虫硫胺素还原酶的潜在抑制剂。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-16 DOI: 10.1080/07391102.2023.2247084
Prisca Baah Nketia, Edward Ntim Gasu, Jehoshaphat Oppong Mensah, Lawrence Sheringham Borquaye

Despite the recent advances in drug research, finding a safe, effective, and easy to use chemotherapy for human African trypanosomiasis (HAT) remains a challenging task. Trypanosomatids have developed resistance mechanisms towards melarsoprol (the current drug of choice), and the fact that it is poisonous is problematic. Therefore, a search for alternative therapeutics against the parasite is urgently needed. Natural products offer potential for drug discovery, but little or nothing is known about the target of inhibition or possible mode of inhibition. Therefore, this study aimed to use molecular docking and molecular dynamics simulations to evaluate 30 antitrypanosomal natural products as potential inhibitors of trypanothione reductase, a key protein necessary for the survival of the Trypanosoma brucei. The study also assessed the pharmacokinetic properties of the most promising compounds. Of the compounds evaluated, α-bisabolol, letestuianin C, waltherione, and mexicanin E were found to bind at the active site of TR and interact with Met115, Tyr112, and Trp23, which are essential for enzyme functioning. Molecular dynamic simulations revealed the sustained binding of α-bisabolol and letestuianin C throughout the simulation period, potentially obstructing the binding of the substrate (T[S]2) and impeding catalysis. The binding of these compounds to TR led to the presence of solvent molecules in the enzyme's active site, and this could potentially lead to protein aggregation. Furthermore, α-bisabolol and letestuianin C exhibited promising safety profiles. Consequently, α-bisabolol and letestuianin C have been shown to be viable candidates for targeting trypanothione reductase in the fight against human African trypanosomiasis.Communicated by Ramaswamy H. Sarma.

尽管近来药物研究取得了进展,但找到一种安全、有效且易于使用的化疗方法来治疗人类非洲锥虫病(HAT)仍然是一项具有挑战性的任务。锥虫对美拉索洛尔(目前的首选药物)产生了抗药性,而且美拉索洛尔有毒,这也是一个问题。因此,迫切需要寻找针对寄生虫的替代疗法。天然产品为药物发现提供了潜力,但人们对其抑制靶点或可能的抑制模式知之甚少或一无所知。因此,本研究旨在利用分子对接和分子动力学模拟来评估 30 种抗锥虫天然产物作为锥硫蛋白还原酶潜在抑制剂的潜力,锥硫蛋白还原酶是布氏锥虫生存所必需的关键蛋白。研究还评估了最有前景的化合物的药代动力学特性。在所评估的化合物中,α-bisabolol、letestuianin C、waltherione 和 mexicanin E 被发现能与 TR 的活性位点结合,并与 Met115、Tyr112 和 Trp23 相互作用,而 Met115、Tyr112 和 Trp23 对酶的运作至关重要。分子动态模拟显示,α-双羟基苯酚和 letestuianin C 在整个模拟期间持续结合,有可能阻碍底物(T[S]2)的结合并妨碍催化。这些化合物与 TR 的结合导致酶的活性位点出现溶剂分子,这有可能导致蛋白质聚集。此外,α-双羟基苯乙醇和 Letestuianin C 表现出良好的安全性。因此,α-双羟基苯乙醇和来曲霉素 C 被证明是针对非洲锥虫病的锥硫还原酶的可行候选药物。
{"title":"<i>In silico</i> identification of α-bisabolol and letestuianin C as potential inhibitors of <i>Trypanosoma brucei</i> trypanothione reductase.","authors":"Prisca Baah Nketia, Edward Ntim Gasu, Jehoshaphat Oppong Mensah, Lawrence Sheringham Borquaye","doi":"10.1080/07391102.2023.2247084","DOIUrl":"10.1080/07391102.2023.2247084","url":null,"abstract":"<p><p>Despite the recent advances in drug research, finding a safe, effective, and easy to use chemotherapy for human African trypanosomiasis (HAT) remains a challenging task. Trypanosomatids have developed resistance mechanisms towards melarsoprol (the current drug of choice), and the fact that it is poisonous is problematic. Therefore, a search for alternative therapeutics against the parasite is urgently needed. Natural products offer potential for drug discovery, but little or nothing is known about the target of inhibition or possible mode of inhibition. Therefore, this study aimed to use molecular docking and molecular dynamics simulations to evaluate 30 antitrypanosomal natural products as potential inhibitors of trypanothione reductase, a key protein necessary for the survival of the <i>Trypanosoma brucei</i>. The study also assessed the pharmacokinetic properties of the most promising compounds. Of the compounds evaluated, α-bisabolol, letestuianin C, waltherione, and mexicanin E were found to bind at the active site of TR and interact with Met115, Tyr112, and Trp23, which are essential for enzyme functioning. Molecular dynamic simulations revealed the sustained binding of α-bisabolol and letestuianin C throughout the simulation period, potentially obstructing the binding of the substrate (T[S]<sub>2</sub>) and impeding catalysis. The binding of these compounds to TR led to the presence of solvent molecules in the enzyme's active site, and this could potentially lead to protein aggregation. Furthermore, α-bisabolol and letestuianin C exhibited promising safety profiles. Consequently, α-bisabolol and letestuianin C have been shown to be viable candidates for targeting trypanothione reductase in the fight against human African trypanosomiasis.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10009675","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phytochemicals-based β-amyloid cleaving enzyme-1 and MAO-B inhibitors for the treatment of Alzheimer's disease: molecular simulations-based predictions. 基于植物化学物质的β-淀粉样蛋白裂解酶-1和MAO-B抑制剂治疗阿尔茨海默病:基于分子模拟的预测。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-10-10 DOI: 10.1080/07391102.2023.2265494
Mater H Mahnashi, Muhammad Ayaz, Mehreen Ghufran, Ibrahim Abdullah Almazni, Omaish Alqahtani, Bandar A Alyami, Yahya S Alqahtani, Haider Ali Khan, Abdul Sadiq, Muhammad Waqas

Alzheimer's disease (AD) is among the highly prevalent neurodegenerative disorder of the aging brain and is allied with cognitive and behavioral abnormalities. Unfortunately, there is very limited drug discovery for the effective management of AD, and the clinically approved drugs have limited efficacy. Consequently, there is an immediate demand for the development of new compounds that have the ability to act as multitarget-directed ligands (MTDLs). As major pathological targets of the disease, the current study aimed to investigate lead natural bioactive compounds including apigenin, epigallocatechin-3-gallate, berberine, curcumin, genistein, luteolin, quercetin, resveratrol for their inhibitory potentials against β-amyloid cleaving enzyme-1 (BACE1) and monoamine oxidase-B (MAO-B) enzymes. The study compounds were docked against the target enzymes (MAO-B and BACE1) using MOE software and subsequent molecular dynamics simulations (MDS) studies. The molecular docking analysis revealed that these phytochemicals (MTDLs) showed good interactions with the target enzymes as compared to the reference inhibitors. Among these eight phytocompounds, the epigallocatechin-3-gallate compound was an active inhibitor against both drug targets, with the highest docking scores and good interactions with the active residues of the enzymes. Furthermore, the docking result of the active one inhibitor in complex with the target enzymes (epigallocatechin-3-gallate/BACE1, epigallocatechin-3-gallate/MAO-B, reference/BACE1 and reference/MAO-B) were further validated by MDS. According to the findings of our study, epigallocatechin-3-gallate has the potential to be a candidate for use in the treatment of neurological illnesses like AD. This compound has MTDL potential and may be exploited to create new compounds with disease-modifying features.Communicated by Ramaswamy H. Sarma.

阿尔茨海默病(AD)是老年大脑中高度流行的神经退行性疾病之一,与认知和行为异常有关。不幸的是,有效治疗AD的药物发现非常有限,临床批准的药物疗效有限。因此,迫切需要开发具有多靶点定向配体(MTDL)能力的新化合物。作为该疾病的主要病理靶点,本研究旨在研究主要的天然生物活性化合物,包括芹菜素、表没食子儿茶素-3-没食子酸盐、黄连素、姜黄素、染料木素、木犀草素、槲皮素、白藜芦醇对β-淀粉样蛋白裂解酶-1(BACE1)和单胺氧化酶-B(MAO-B)的抑制潜力。使用MOE软件和随后的分子动力学模拟(MDS)研究,将研究化合物与靶酶(MAO-B和BACE1)对接。分子对接分析表明,与参考抑制剂相比,这些植物化学物质(MTDL)与靶酶表现出良好的相互作用。在这八种植物化合物中,表没食子儿茶素-3-没食子酸盐化合物是对两种药物靶点的活性抑制剂,具有最高的对接得分和与酶的活性残基的良好相互作用。此外,MDS进一步验证了活性一种抑制剂与靶酶(表没食子儿茶素-3-没食子酸盐/BACE1、表没食子子茶素3-没食子酸盐/MAO-B、参考/BACE1和参考/MAO-B)复合物的对接结果。根据我们的研究结果,表没食子儿茶素-3-没食子酸盐有可能成为治疗AD等神经系统疾病的候选药物。该化合物具有MTDL潜力,可用于开发具有疾病修饰特征的新化合物。Ramaswamy H.Sarma通讯。
{"title":"Phytochemicals-based β-amyloid cleaving enzyme-1 and MAO-B inhibitors for the treatment of Alzheimer's disease: molecular simulations-based predictions.","authors":"Mater H Mahnashi, Muhammad Ayaz, Mehreen Ghufran, Ibrahim Abdullah Almazni, Omaish Alqahtani, Bandar A Alyami, Yahya S Alqahtani, Haider Ali Khan, Abdul Sadiq, Muhammad Waqas","doi":"10.1080/07391102.2023.2265494","DOIUrl":"10.1080/07391102.2023.2265494","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is among the highly prevalent neurodegenerative disorder of the aging brain and is allied with cognitive and behavioral abnormalities. Unfortunately, there is very limited drug discovery for the effective management of AD, and the clinically approved drugs have limited efficacy. Consequently, there is an immediate demand for the development of new compounds that have the ability to act as multitarget-directed ligands (MTDLs). As major pathological targets of the disease, the current study aimed to investigate lead natural bioactive compounds including apigenin, epigallocatechin-3-gallate, berberine, curcumin, genistein, luteolin, quercetin, resveratrol for their inhibitory potentials against β-amyloid cleaving enzyme-1 (BACE1) and monoamine oxidase-B (MAO-B) enzymes. The study compounds were docked against the target enzymes (MAO-B and BACE1) using MOE software and subsequent molecular dynamics simulations (MDS) studies. The molecular docking analysis revealed that these phytochemicals (MTDLs) showed good interactions with the target enzymes as compared to the reference inhibitors. Among these eight phytocompounds, the epigallocatechin-3-gallate compound was an active inhibitor against both drug targets, with the highest docking scores and good interactions with the active residues of the enzymes. Furthermore, the docking result of the active one inhibitor in complex with the target enzymes (epigallocatechin-3-gallate/BACE1, epigallocatechin-3-gallate/MAO-B, reference/BACE1 and reference/MAO-B) were further validated by MDS. According to the findings of our study, epigallocatechin-3-gallate has the potential to be a candidate for use in the treatment of neurological illnesses like AD. This compound has MTDL potential and may be exploited to create new compounds with disease-modifying features.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41182651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular analysis of dUTPase of Helicobacter pylori for identification of novel inhibitors using in silico studies. 对幽门螺杆菌 dUTP 酶进行分子分析,利用硅学研究鉴定新型抑制剂。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-17 DOI: 10.1080/07391102.2023.2247080
Rinki Sisodia, Debapriyo Sarmadhikari, Pooja Anjali Mazumdar, Shailendra Asthana, Chaithanya Madhurantakam

The human gastric pathogen Helicobacter pylori chronically affects the gastric mucosal layer of approximately half of world's population. The emergence of resistant strains urges the need for identification of novel and selective drug against new molecular targets. A ubiquitous enzyme, Deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase), is considered as first line of defense against uracil mis-incorporation into DNA, and essential for genome integrity. Lack of dUTPase triggers an elevated recombination frequency, DNA breaks and ultimately cell death. Hence, dUTPase can be considered as a promising target for development of novel lead inhibitor compounds in H. pylori treatment. Herein, we report the generation of three-dimensional model of the target protein using comparative modelling and its validation. To identify dUTPase inhibitors, a high throughput virtual screening approach utilizing Knowledge-based inhibitors and DrugBank database was implemented. Top ranked compounds were scrutinized based on investigations of the protein-ligand interaction fingerprints, molecular interaction maps and binding affinities and the drug potentiality. The best ligands were studied further for complex stability and intermolecular interaction profiling with respect to time under 100 ns classical molecular dynamic stimulation, establishing significant stability in dynamic states as observed from RMSD and RMSF parameters and interactions with the catalytic site residues. The binding free energy calculation computed using MM-GBSA method from the MD simulation trajectories demonstrated that our molecules possess strong binding affinity towards the Helicobacter pylori dUTPase protein. We conclude that our proposed molecules may be potential lead molecules for effective inhibition against the H. pylori dUTPase protein subject to experimental validation.Communicated by Ramaswamy H. Sarma.

人类胃部病原体幽门螺旋杆菌长期影响着全球约一半人口的胃黏膜层。耐药菌株的出现促使人们需要针对新的分子靶点寻找新型选择性药物。一种无处不在的酶--脱氧尿苷-5'-三磷酸核苷酸水解酶(dUTPase),被认为是防止尿嘧啶错并入 DNA 的第一道防线,对基因组的完整性至关重要。缺乏 dUTPase 会导致重组频率升高、DNA 断裂,最终导致细胞死亡。因此,dUTPase 可被视为开发治疗幽门螺杆菌的新型先导抑制剂化合物的理想靶点。在此,我们报告了利用比较建模生成目标蛋白的三维模型及其验证。为了确定 dUTPase 抑制剂,我们利用基于知识的抑制剂和 DrugBank 数据库实施了高通量虚拟筛选方法。根据对蛋白质配体相互作用指纹、分子相互作用图谱和结合亲和力以及药物潜力的研究,对排名靠前的化合物进行了仔细研究。根据 RMSD 和 RMSF 参数以及与催化位点残基的相互作用,确定了动态状态下的显著稳定性。利用 MM-GBSA 方法对 MD 模拟轨迹进行的结合自由能计算表明,我们的分子与幽门螺杆菌 dUTP 酶蛋白具有很强的结合亲和力。我们的结论是,我们提出的分子可能是有效抑制幽门螺杆菌 dUTP 酶蛋白的潜在先导分子,但还有待于实验验证。
{"title":"Molecular analysis of dUTPase of <i>Helicobacter pylori</i> for identification of novel inhibitors using <i>in silico</i> studies.","authors":"Rinki Sisodia, Debapriyo Sarmadhikari, Pooja Anjali Mazumdar, Shailendra Asthana, Chaithanya Madhurantakam","doi":"10.1080/07391102.2023.2247080","DOIUrl":"10.1080/07391102.2023.2247080","url":null,"abstract":"<p><p>The human gastric pathogen <i>Helicobacter pylori</i> chronically affects the gastric mucosal layer of approximately half of world's population. The emergence of resistant strains urges the need for identification of novel and selective drug against new molecular targets. A ubiquitous enzyme, Deoxyuridine 5'-triphosphate nucleotidohydrolase (dUTPase), is considered as first line of defense against uracil mis-incorporation into DNA, and essential for genome integrity. Lack of dUTPase triggers an elevated recombination frequency, DNA breaks and ultimately cell death. Hence, dUTPase can be considered as a promising target for development of novel lead inhibitor compounds in <i>H. pylori</i> treatment. Herein, we report the generation of three-dimensional model of the target protein using comparative modelling and its validation. To identify dUTPase inhibitors, a high throughput virtual screening approach utilizing Knowledge-based inhibitors and DrugBank database was implemented. Top ranked compounds were scrutinized based on investigations of the protein-ligand interaction fingerprints, molecular interaction maps and binding affinities and the drug potentiality. The best ligands were studied further for complex stability and intermolecular interaction profiling with respect to time under 100 ns classical molecular dynamic stimulation, establishing significant stability in dynamic states as observed from RMSD and RMSF parameters and interactions with the catalytic site residues. The binding free energy calculation computed using MM-GBSA method from the MD simulation trajectories demonstrated that our molecules possess strong binding affinity towards the <i>Helicobacter pylori</i> dUTPase protein. We conclude that our proposed molecules may be potential lead molecules for effective inhibition against the <i>H. pylori</i> dUTPase protein subject to experimental validation.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10005499","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Substrate-like novel inhibitors of prolyl specific oligo peptidase for neurodegenerative disorders. 治疗神经退行性疾病的脯氨酰特异性寡肽酶底物样新型抑制剂。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-22 DOI: 10.1080/07391102.2023.2246577
Majid Khan, Sobia Ahsan Halim, Muhammad Waqas, Farhad Golmohammadi, Saeed Balalaie, Rene Csuk, Jalal Uddin, Ajmal Khan, Ahmed Al-Harrasi

Prolyl specific oligopeptidase (POP), is one of the highly expressed enzymes in the brain and is a prime target to treat disorders related to the central nervous system. Here, we describe the structure-based design of the tacrine derivatives, selective, and brain-permeable POP inhibitors. These compounds inactivate POP in-vitro specifically and sustainably at very low concentrations (nano molar). Among this series, compound 6b (IC50 = 0.81 ± 0.04 µM) exhibited most potent inhibition. Furthermore, kinetic study revealed that these molecules target active site of POP which is further confirmed by in-silico molecular interaction analysis. The computational docking results indicates that the compounds are well fitted in the active site with high binding score (i.e., > -7 to > -4 kcal/mol) where Trp595, Arg643, Tyr473, and Ser554 plays important role in binding with the active compounds. The molecular dynamic simulation of most active compounds (6a, 6b, 6d, and 6f) displayed higher free energy binding, when compared to the standard drug in MM-PBSA based binding free energy calculation. In addition, the predicted pharmacokinetic profile suggests that these compounds can serve as excellent inhibitors upon additional optimization which makes them prime choice for further investigation.Communicated by Ramaswamy H. Sarma.

脯氨酰特异性寡肽酶(POP)是大脑中高表达的酶之一,也是治疗中枢神经系统相关疾病的主要靶点。在此,我们介绍了基于结构设计的他克林衍生物、选择性和脑渗透性 POP 抑制剂。这些化合物能在体外以极低的浓度(纳摩尔)特异性地持续灭活持久性有机污染物。在这一系列化合物中,化合物 6b(IC50 = 0.81 ± 0.04 µM)的抑制作用最强。此外,动力学研究表明,这些分子以持久性有机污染物的活性位点为靶点,这一点在室内分子相互作用分析中得到了进一步证实。计算对接结果表明,这些化合物与活性位点的结合得分很高(即 > -7 至 > -4 kcal/mol),其中 Trp595、Arg643、Tyr473 和 Ser554 在与活性化合物的结合中发挥了重要作用。在基于 MM-PBSA 的结合自由能计算中,与标准药物相比,大多数活性化合物(6a、6b、6d 和 6f)的分子动力学模拟显示出更高的结合自由能。此外,预测的药代动力学特征表明,这些化合物经进一步优化后可作为出色的抑制剂,因此成为进一步研究的首选。
{"title":"Substrate-like novel inhibitors of prolyl specific oligo peptidase for neurodegenerative disorders.","authors":"Majid Khan, Sobia Ahsan Halim, Muhammad Waqas, Farhad Golmohammadi, Saeed Balalaie, Rene Csuk, Jalal Uddin, Ajmal Khan, Ahmed Al-Harrasi","doi":"10.1080/07391102.2023.2246577","DOIUrl":"10.1080/07391102.2023.2246577","url":null,"abstract":"<p><p>Prolyl specific oligopeptidase (POP), is one of the highly expressed enzymes in the brain and is a prime target to treat disorders related to the central nervous system. Here, we describe the structure-based design of the tacrine derivatives, selective, and brain-permeable POP inhibitors. These compounds inactivate POP in-vitro specifically and sustainably at very low concentrations (nano molar). Among this series, compound <b>6b</b> (IC<sub>50</sub> = 0.81 ± 0.04 µM) exhibited most potent inhibition. Furthermore, kinetic study revealed that these molecules target active site of POP which is further confirmed by <i>in-silico</i> molecular interaction analysis. The computational docking results indicates that the compounds are well fitted in the active site with high binding score (i.e., > -7 to > -4 kcal/mol) where Trp595, Arg643, Tyr473, and Ser554 plays important role in binding with the active compounds. The molecular dynamic simulation of most active compounds (<b>6a</b>, <b>6b</b>, <b>6d</b>, and <b>6f</b>) displayed higher free energy binding, when compared to the standard drug in MM-PBSA based binding free energy calculation. In addition, the predicted pharmacokinetic profile suggests that these compounds can serve as excellent inhibitors upon additional optimization which makes them prime choice for further investigation.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10050920","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of a novel apigenin prodrug programmed for alkaline-phosphatase instructed self-inhibition to combat cancer. 开发出一种新型芹菜素原药,可用于抗击癌症的碱性磷酸酶自我抑制。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-28 DOI: 10.1080/07391102.2023.2247083
Dimitrios Diamantis, Antonios D Tsiailanis, Christina Papaemmanouil, Maria-Christina Nika, Zoi Kanaki, Simona Golic Grdadolnik, Andrej Babic, Eleftherios Paraskevas Tzakos, Isabelle Fournier, Michel Salzet, Prem Prakash Kushwaha, Nikolaos S Thomaidis, Theodoros Rampias, Eswar Shankar, Serdar Karakurt, Sanjay Gupta, Andreas G Tzakos

Elevated levels of alkaline phosphatase (ALP) in the tumor microenvironment (TME) are a hallmark of cancer progression and thus inhibition of ALP could serve as an effective approach against cancer. Herein, we developed a novel prodrug approach to tackle cancer that bears self-inhibiting alkaline phosphatase-responsiveness properties that can enhance at the same time the solubility of the parent compound. To probe this novel concept, we selected apigenin as the cytotoxic agent since we first unveiled, that it directly interacts and inhibits ALP activity. Consequently, we rationally designed and synthesized, using a self-immolative linker, an ALP responsive apigenin-based phosphate prodrug, phospho-apigenin. Phospho-apigenin markedly increased the stability of the parent compound apigenin. Furthermore, the prodrug exhibited enhanced antiproliferative effect in malignant cells with elevated ALP levels, compared to apigenin. This recorded potency of the developed prodrug was further confirmed in vivo where phospho-apigenin significantly suppressed by 52.8% the growth of PC-3 xenograft tumors.Communicated by Ramaswamy H. Sarma.

肿瘤微环境(TME)中碱性磷酸酶(ALP)水平的升高是癌症进展的标志,因此抑制 ALP 可作为一种有效的抗癌方法。在此,我们开发了一种新的原药方法来治疗癌症,这种方法具有自我抑制碱性磷酸酶的特性,可以同时提高母体化合物的溶解度。为了探究这个新概念,我们选择了芹菜素作为细胞毒剂,因为我们首次发现芹菜素能直接与碱性磷酸酶相互作用并抑制其活性。因此,我们利用自巯基连接体合理地设计和合成了一种对 ALP 有反应的芹菜素磷酸盐原药--磷酸芹菜素。磷酸化芹菜素显著提高了母体化合物芹菜素的稳定性。此外,与芹菜素相比,该原药在 ALP 水平升高的恶性细胞中表现出更强的抗增殖作用。磷酸芹菜素显著抑制了 PC-3 异种移植肿瘤的生长,抑制率为 52.8%。
{"title":"Development of a novel apigenin prodrug programmed for alkaline-phosphatase instructed self-inhibition to combat cancer.","authors":"Dimitrios Diamantis, Antonios D Tsiailanis, Christina Papaemmanouil, Maria-Christina Nika, Zoi Kanaki, Simona Golic Grdadolnik, Andrej Babic, Eleftherios Paraskevas Tzakos, Isabelle Fournier, Michel Salzet, Prem Prakash Kushwaha, Nikolaos S Thomaidis, Theodoros Rampias, Eswar Shankar, Serdar Karakurt, Sanjay Gupta, Andreas G Tzakos","doi":"10.1080/07391102.2023.2247083","DOIUrl":"10.1080/07391102.2023.2247083","url":null,"abstract":"<p><p>Elevated levels of alkaline phosphatase (ALP) in the tumor microenvironment (TME) are a hallmark of cancer progression and thus inhibition of ALP could serve as an effective approach against cancer. Herein, we developed a novel prodrug approach to tackle cancer that bears self-inhibiting alkaline phosphatase-responsiveness properties that can enhance at the same time the solubility of the parent compound. To probe this novel concept, we selected apigenin as the cytotoxic agent since we first unveiled, that it directly interacts and inhibits ALP activity. Consequently, we rationally designed and synthesized, using a self-immolative linker, an ALP responsive apigenin-based phosphate prodrug, phospho-apigenin. Phospho-apigenin markedly increased the stability of the parent compound apigenin. Furthermore, the prodrug exhibited enhanced antiproliferative effect in malignant cells with elevated ALP levels, compared to apigenin. This recorded potency of the developed prodrug was further confirmed <i>in vivo</i> where phospho-apigenin significantly suppressed by 52.8% the growth of PC-3 xenograft tumors.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10466155","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In-silico screening of Acacia pennata and Bridelia retusa reveals pinocembrin-7-O-β-D-glucopyranoside as a promising β-lactamase inhibitor to combat antibiotic resistance. 通过对金合欢(Acacia pennata)和金合欢属(Bridelia retusa)植物进行室内筛选,发现 pinocembrin-7-O-β-D-glucopyranoside 是一种很有前景的β-内酰胺酶抑制剂,可用于对抗抗生素耐药性。
IF 2.7 3区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-10-01 Epub Date: 2023-08-17 DOI: 10.1080/07391102.2023.2248272
Abd Kakhar Umar, Dhritiman Roy, Mohnad Abdalla, Yosra Modafer, Nawal Al-Hoshani, Han Yu, James H Zothantluanga

The β-lactamase of Pseudomonas aeruginosa is known to degrade β-lactam antibiotics such as penicillins, cephalosporins, monobactams, and carbapenems. With the discovery of an extended-spectrum β-lactamase in a clinical isolate of P. aeruginosa, the bacterium has become multi-drug resistant. In this study, we aim to identify new β-lactamase inhibitors by virtually screening a total of 43 phytocompounds from two Indian medicinal plants. In the molecular docking studies, pinocembrin-7-O-β-D-glucopyranoside (P7G) (-9.6 kcal/mol) from Acacia pennata and ellagic acid (EA) (-9.2 kcal/mol) from Bridelia retusa had lower binding energy than moxalactam (-8.4 kcal/mol). P7G and EA formed 5 (Ser62, Asn125, Asn163, Thr209, and Ser230) and 4 (Lys65, Ser123, Asn125, and Glu159) conventional hydrogens bonds with the active site residues. 100 ns MD simulations revealed that moxalactam and P7G (but not EA) were able to form a stable complex. The binding free energy calculations further revealed that P7G (-59.6526 kcal/mol) formed the most stable complex with β-lactamase when compared to moxalactam (-46.5669 kcal/mol) and EA (-28.4505 kcal/mol). The HOMO-LUMO and other DFT parameters support the stability and chemical reactivity of P7G at the active site of β-lactamase. P7G passed all the toxicity tests and bioavailability tests indicating that it possesses drug-likeness. Among the studied compounds, we identified P7G of A. pennata as the most promising phytocompound to combat antibiotic resistance by potentially inhibiting the β-lactamase of P. aeruginosa.Communicated by Ramaswamy H. Sarma.

众所周知,铜绿假单胞菌的β-内酰胺酶能降解β-内酰胺类抗生素,如青霉素类、头孢菌素类、单内酰胺类和碳青霉烯类。随着在铜绿假单胞菌的临床分离株中发现了一种广谱β-内酰胺酶,该细菌变得具有多重耐药性。在这项研究中,我们从两种印度药用植物中虚拟筛选出 43 种植物化合物,旨在找出新的β-内酰胺酶抑制剂。在分子对接研究中,来自相思树的 pinocembrin-7-O-β-D-glucopyranoside (P7G) (-9.6 kcal/mol) 和来自 Bridelia retusa 的 ellagic acid (EA) (-9.2 kcal/mol) 的结合能低于莫沙内酰胺(-8.4 kcal/mol)。P7G 和 EA 与活性位点残基形成了 5 个(Ser62、Asn125、Asn163、Thr209 和 Ser230)和 4 个(Lys65、Ser123、Asn125 和 Glu159)常规氢键。100 ns MD 模拟显示,莫沙内酰胺和 P7G(而非 EA)能够形成稳定的复合物。结合自由能计算进一步表明,与莫沙内酰胺(-46.5669 kcal/mol)和 EA(-28.4505 kcal/mol)相比,P7G(-59.6526 kcal/mol)与β-内酰胺酶形成的复合物最为稳定。HOMO-LUMO 和其他 DFT 参数支持 P7G 在 β-内酰胺酶活性位点的稳定性和化学反应活性。P7G 通过了所有毒性测试和生物利用度测试,表明它具有药物相似性。在所研究的化合物中,我们发现 A. pennata 的 P7G 是最有希望通过潜在抑制铜绿假单胞菌的 β-内酰胺酶来对抗抗生素耐药性的植物化合物。
{"title":"<i>In-silico</i> screening of <i>Acacia pennata</i> and <i>Bridelia retusa</i> reveals pinocembrin-7-O-β-D-glucopyranoside as a promising β-lactamase inhibitor to combat antibiotic resistance.","authors":"Abd Kakhar Umar, Dhritiman Roy, Mohnad Abdalla, Yosra Modafer, Nawal Al-Hoshani, Han Yu, James H Zothantluanga","doi":"10.1080/07391102.2023.2248272","DOIUrl":"10.1080/07391102.2023.2248272","url":null,"abstract":"<p><p>The β-lactamase of <i>Pseudomonas aeruginosa</i> is known to degrade β-lactam antibiotics such as penicillins, cephalosporins, monobactams, and carbapenems. With the discovery of an extended-spectrum β-lactamase in a clinical isolate of <i>P. aeruginosa</i>, the bacterium has become multi-drug resistant. In this study, we aim to identify new β-lactamase inhibitors by virtually screening a total of 43 phytocompounds from two Indian medicinal plants. In the molecular docking studies, pinocembrin-7-<i>O</i>-β-D-glucopyranoside (P7G) (-9.6 kcal/mol) from <i>Acacia pennata</i> and ellagic acid (EA) (-9.2 kcal/mol) from <i>Bridelia retusa</i> had lower binding energy than moxalactam (-8.4 kcal/mol). P7G and EA formed 5 (<i>Ser62, Asn125, Asn163, Thr209,</i> and <i>Ser230</i>) and 4 (<i>Lys65, Ser123, Asn125,</i> and <i>Glu159</i>) conventional hydrogens bonds with the active site residues. 100 ns MD simulations revealed that moxalactam and P7G (but not EA) were able to form a stable complex. The binding free energy calculations further revealed that P7G (-59.6526 kcal/mol) formed the most stable complex with β-lactamase when compared to moxalactam (-46.5669 kcal/mol) and EA (-28.4505 kcal/mol). The HOMO-LUMO and other DFT parameters support the stability and chemical reactivity of P7G at the active site of β-lactamase. P7G passed all the toxicity tests and bioavailability tests indicating that it possesses drug-likeness. Among the studied compounds, we identified P7G of <i>A. pennata</i> as the most promising phytocompound to combat antibiotic resistance by potentially inhibiting the β-lactamase of <i>P. aeruginosa</i>.Communicated by Ramaswamy H. Sarma.</p>","PeriodicalId":15272,"journal":{"name":"Journal of Biomolecular Structure & Dynamics","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10031393","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Biomolecular Structure & Dynamics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1