binding pose metadynamics

绑定姿势元动力学
  • 文章类型: Journal Article
    JNK3,MAPK家族的成员,在介导细胞对应激信号的反应中起着关键作用,它的激活与无数的炎症有关。虽然JNK3有望成为亨廷顿等神经退行性疾病的治疗靶点,帕金森,和老年痴呆症,有效的JNK3抑制剂市场上仍然存在空白。尽管一些泛JNK抑制剂达到临床试验,没有JNK靶向疗法获得市场批准。为了弥合这个差距,我们的研究引入了一种复杂的虚拟筛查方法.我们从基于能量的筛查开始,随后整合了各种评分技术。这些包括滑翔对接分数,MM/GBSA,以及DeepDock和高级图神经网络等人工评分机制。该虚拟筛选工作流程旨在评估和鉴定具有高结合亲和力的潜在小分子抑制剂。我们已经实施了虚拟筛选工作流程来识别潜在的候选分子。这个过程导致了十个分子的选择。随后,这十种分子已经经历了生物活性评估,以评估它们的潜在功效。令人印象深刻的是,分子化合物6被认为是最有前途的,表现出有效的激酶抑制活性,IC50为130.1nM,巨噬细胞内TNF-α释放显着减少。这表明化合物6可能作为治疗神经炎症和神经变性疾病的有效抑制剂。进一步的药物修饰以优化化合物6的前景为该领域的未来研究和开发提供了有希望的途径。利用结合姿态元动力学和分子动力学模拟,我们深入研究了化合物6与JNK3的明确结合模式。这些见解为精制药物开发战略铺平了道路。总的来说,我们的结果强调了混合虚拟筛选工作流程在鉴定稳健的JNK3抑制剂中的功效,有望对神经炎症和神经退行性疾病进行创新治疗。
    JNK3, a member of the MAPK family, plays a pivotal role in mediating cellular responses to stress signals, with its activation implicated in a myriad of inflammatory conditions. While JNK3 holds promise as a therapeutic target for neurodegenerative disorders such as Huntington\'s, Parkinson\'s, and Alzheimer\'s diseases, there remains a gap in the market for effective JNK3 inhibitors. Despite some pan-JNK inhibitors reaching clinical trials, no JNK-targeted therapies have achieved market approval. To bridge this gap, our study introduces a sophisticated virtual screening approach. We begin with an energy-based screening, subsequently integrating a variety of rescoring techniques. These encompass glide docking scores, MM/GBSA, and artificial scoring mechanisms such as DeepDock and advanced Graph Neural Networks. This virtual screening workflow is designed to evaluate and identify potential small-molecule inhibitors with high binding affinity. We have implemented a virtual screening workflow to identify potential candidate molecules. This process has resulted in the selection of ten molecules. Subsequently, these ten molecules have undergone biological activity evaluation to assess their potential efficacy. Impressively, molecule compound 6 surfaced as the most promising, exhibiting a potent kinase inhibitory activity marked by an IC50 of 130.1 nM and a notable reduction in TNF-α release within macrophages. This suggests that compound 6 could potentially serve as an effective inhibitor for the treatment of neuroinflammation and neurodegenerative diseases. The prospect of further medicinal modifications to optimize compound 6 presents a promising avenue for future research and development in this field. Utilizing binding pose metadynamics coupled with molecular dynamics simulations, we delved into the explicit binding mode of compound 6 to JNK3. Such insights pave the way for refined drug development strategies. Collectively, our results underscore the efficacy of the hybrid virtual screening workflow in the identification of robust JNK3 inhibitors, holding promise for innovative treatments against neuroinflammation and neurodegenerative disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    组蛋白脱乙酰酶6和8(HDAC6/8)的高度结构同源性对实现同种型选择性提出了挑战,并且由于临床应用中的泛抑制而导致不良副作用。此外,双靶点抑制剂的合理设计,以HDAC6/8为中心,需要对它们的选择性机制有深刻的理解。解决了在开发靶向特定亚型的抑制剂时增强特异性的迫切需要,我们阐明了通过计算机内策略选择性抑制HDAC6/8抑制剂的机制.与Asp101和Tyr306的氢键相互作用是使化合物12b能够选择性抑制HDAC8的关键因素。其有利的空间取向将12b的帽组放置在Tyr306和Tyr100之间,导致整体L形构造。这两个因素显著有助于12b对HDAC8的选择性抑制活性。化合物NN-390的锌结合基团(ZBG)与HDAC6的关键残基His610形成氢键,促进与锌离子的稳定螯合。此外,NN-390的帽群通过范德华力与Phe620和Phe680相互作用,导致整体Y形构象。上述因素是NN-390选择性抑制HDAC6的主要原因。此外,Cap组是否在对位或间位将影响HDAC6或HDAC8的选择性抑制。我们相信这些线索可以为合理设计靶向HDAC6/8的选择性抑制剂提供有价值的见解,并为合理设计基于HDAC6/8的双靶标抑制剂铺平道路。由RamaswamyH.Sarma沟通。
    The high structural homology of histone deacetylases 6 and 8 (HDAC6/8) poses a challenge in achieving isoform selectivity and has resulted in adverse side effects due to pan-inhibition in clinical applications. Additionally, the rational design of dual-target inhibitors, centered on HDAC6/8, demands a profound understanding of their selectivity mechanisms. Addressing the urgent need for enhanced specificity in the development of inhibitors targeting specific isoforms, we elucidate the mechanism underpinning the selective inhibition of HDAC6/8 inhibitors through in-silico strategies. The hydrogen bonding interaction with Asp101 and Tyr306 is a key factor that enables compound 12b to selectively inhibit HDAC8. Its favorable spatial orientation places the Cap group of 12b between Tyr306 and Tyr100, resulting in an overall L-shaped conformation. These two factors significantly contribute to the selective inhibitory activity of 12b against HDAC8. The zinc binding group (ZBG) of compound NN-390 forms a hydrogen bond with His610, a key residue of HDAC6, facilitating stable chelation with zinc ions. In addition, the Cap group of NN-390 interacts with Phe620 and Phe680 via van der Waals forces, leading to an overall Y-shaped conformation. The aforementioned factors are the main reasons for the selective inhibition of HDAC6 by NN-390. Furthermore, whether the Cap group is in the para or meta-position will influence the selective inhibition of either HDAC6 or HDAC8. We believe these clues can offer valuable insights for the rational design of selective inhibitors targeting HDAC6/8 and pave the way for rational design of dual-target HDAC6/8-based inhibitors.Communicated by Ramaswamy H. Sarma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    PD-1/PD-L1相互作用是小分子抑制剂在癌症免疫治疗中的一个有希望的靶标,但是瞄准这个界面一直很有挑战性。虽然已经努力鉴定靶向正构位点的化合物,尚无报道探讨小分子靶向PD-1变构区的潜力.因此,我们的研究旨在建立一条管道来鉴定能有效结合PD-1正构或变构口袋的小分子.我们根据对PD-1界面结构的广泛分析,将其分为两个热点区域(P区和N区),动态,和充满活力的属性。这些区域对应于正构和变构PPI位点,分别,单克隆抗体靶向。我们使用了一个引导的虚拟筛选工作流程来识别700万个化合物库中的命中,然后根据结构相似性进行聚类,并通过相互作用指纹图谱进行评估。对选择性和多样化的化学代表进行MD模拟和结合能学计算,以过滤出假阳性并鉴定实际的结合物。结合姿势元动力学计算证实了口袋中最终命中的稳定性。本研究强调需要一个集成的管道,使用分子动力学模拟和结合能来识别动态PD-1/PD-L1界面的潜在粘合剂,由于缺乏小分子共晶。从靶向变构区和正构区的大量分子中仅发现了少数潜在的粘合剂。我们的结果表明,变构位点比正构位点具有更大的抑制剂设计潜力。确定的“计算命中”具有作为体外评估的起点的潜力,然后进行命中到领先优化。总的来说,这项研究是为探索和丰富PPI界面的变构和正构位点建立计算管道的努力,“一个艰难但不可缺少的坚果”。
    The PD-1/PD-L1 interaction is a promising target for small molecule inhibitors in cancer immunotherapy, but targeting this interface has been challenging. While efforts have been made to identify compounds that target the orthosteric sites, no reports have explored the potential of small molecules to target the allosteric region of PD-1. Therefore, our study aims to establish a pipeline to identify small molecules that can effectively bind to either the orthosteric or allosteric pockets of PD-1. We categorized the PD-1 interface into two hot-spot zones (P-and N-zones) based on extensive analysis of its structural, dynamical, and energetic properties. These zones correspond to the orthosteric and allosteric PPI sites, respectively, targeted by monoclonal antibodies. We used a guided virtual screening workflow to identify hits from ∼7 million compounds library, which were then clustered based on structural similarity and assessed by interaction fingerprinting. The selective and diverse chemical representatives were subjected to MD simulations and binding energetics calculations to filter out false positives and identify actual binders. Binding poses metadynamics calculations confirmed the stability of the final hits in the pocket. This study emphasizes the need for an integrated pipeline that uses molecular dynamics simulations and binding energetics to identify potential binders for the dynamic PD-1/PD-L1 interface, due to the lack of small molecule co-crystals. Only a few potential binders were discovered from a large pool of molecules targeting both the allosteric and orthosteric zones. Our results suggest that the allosteric site has more potential than the orthosteric site for inhibitor design. The identified \"computational hits\" hold potential as starting points for in vitro evaluations followed by hit-to-lead optimization. Overall, this study represents an effort to establish a computational pipeline for exploring and enriching both the allosteric and orthosteric sites of PPI interfaces, \"a tough but indispensable nut to crack\".
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    虚拟筛选(VS)是评估用于铅鉴定的化学库的常规方法。因此,为VS选择合适的蛋白质结构是在对接过程中鉴定真正活性物的必要先决条件。但是同一蛋白质的几个晶体结构的存在使得难以合理地选择一个或几个结构进行筛选。因此,已经开发了一种计算优先方案,用于入围晶体结构,以更好的效率识别真正的活性分子。由于小分子抑制剂的鉴定是T790M/L858R(TMLR)EGFR突变体的重要临床要求,它已被选为案例研究。该方法涉及21个共晶配体与相同蛋白质的所有结构的交叉对接,以选择对接具有较低RMSD的非天然配体的结构。然后将交叉对接性能与配体相似性和结合位点构象相似性相关联。最终,结构通过整合交叉对接性能入围,以及配体和结合位点的相似性。此后,结合位元动力学(BPMD)用于鉴定在其各自的结合袋中具有稳定的共晶配体的结构。最后,不同的富集指标,如BEDROC,RIE,AUAC,和EF1%进行了评估,从而鉴定了五个TMLR结构(5HCX,5CAN,5CAP,5CAS和5CAO)。这些结构对接了许多具有低RMSD的非天然配体,含有结构上不同的配体,具有构象不同的结合位点,具有稳定的共晶配体,并且还可以早期鉴定真正的活性物质。本方法可用于任何其他重要治疗性激酶的短列表蛋白质靶标。本文受版权保护。保留所有权利。
    Virtual screening (VS) is a routine method to evaluate chemical libraries for lead identification. Therefore, the selection of appropriate protein structures for VS is an essential prerequisite to identify true actives during docking. But the presence of several crystal structures of the same protein makes it difficult to select one or few structures rationally for screening. Therefore, a computational prioritization protocol has been developed for shortlisting crystal structures that identify true active molecules with better efficiency. As identification of small-molecule inhibitors is an important clinical requirement for the T790M/L858R (TMLR) EGFR mutant, it has been selected as a case study. The approach involves cross-docking of 21 co-crystal ligands with all the structures of the same protein to select structures that dock non-native ligands with lower RMSD. The cross docking performance was then correlated with ligand similarity and binding-site conformational similarity. Eventually, structures were shortlisted by integrating cross-docking performance, and ligand and binding-site similarity. Thereafter, binding pose metadynamics was employed to identify structures having stable co-crystal ligands in their respective binding pockets. Finally, different enrichment metrics like BEDROC, RIE, AUAC, and EF1% were evaluated leading to the identification of five TMLR structures (5HCX, 5CAN, 5CAP, 5CAS, and 5CAO). These structures docked a number of non-native ligands with low RMSD, contain structurally dissimilar ligands, have conformationally dissimilar binding sites, harbor stable co-crystal ligands, and also identify true actives early. The present approach can be implemented for shortlisting protein targets of any other important therapeutic kinases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    铜绿假单胞菌是一种机会性病原体,易于产生耐药性,并且是烧伤患者和患有囊性纤维化或在重症监护病房住院的患者感染的主要原因。该细菌的毒力因子之一是降解宿主组织的细胞外基质并促进侵袭的脂肪酶。溴己新是一种粘液溶解药物,最近被报道作为脂肪酶的竞争性抑制剂,IC50值为49µM。在本研究中,尝试从ChEMBL生物活性化合物数据库中确定更强的抑制剂,与参考化合物溴己新相比。在对接和MD模拟之后,选择四种命中化合物(N1-N4),其显示有希望的结合模式和低RMSD值,指示稳定的蛋白质-配体复合物。从随后的约束姿态元动力学(BPMD)模拟中,其中两种(N2和N4)比溴己新更有效,显示与酶催化位点残基的稳定相互作用。对所有四种化合物进行生物学研究。其中,发现两种相同的化合物是最有效的粘合剂,IC50值为22.1和27.5µM,分别;即大约是参考溴己新的两倍。一起来看,我们的结果表明,这些命中可以是有希望的新的候选物,用作药物的开发针对铜绿假单胞菌脂肪酶酶。由RamaswamyH.Sarma沟通。
    Pseudomonas aeruginosa is an opportunistic pathogen prone to developing drug-resistance and is a major cause of infection for burn patients and patients suffering from cystic fibrosis or are hospitalized in intensive care units. One of the virulence factors of this bacterium is the lipase enzyme that degrades the extracellular matrix of the host tissue and promotes invasion. Bromhexine is a mucolytic drug and has recently been reported to function as a competitive inhibitor of lipase with an IC50 value of 49 µM. In the present study, an attempt was made to identify stronger inhibitors from the ChEMBL database of bioactive compounds, as compared to the reference compound Bromhexine. Following docking and MD simulations, four hit compounds (N1-N4) were selected that showed promising binding modes and low RMSD values indicative of stable protein-ligand complexes. From subsequent binding pose metadynamics (BPMD) simulations, two of these (N2 and N4) stood out as more potent than Bromhexine, displaying stable interactions with residues in the catalytic site of the enzyme. Biological investigations were performed for all four compounds. Among them, the same two hit compounds were found to be the most effective binders with IC50 values of 22.1 and 27.5 µM, respectively; i.e. roughly twice as efficient as the reference Bromhexine. Taken together, our results show that these hits can be promising new candidates to use as leads for the development of drugs targeting the P. aeruginosa lipase enzyme.Communicated by Ramaswamy H. Sarma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    细胞周期蛋白依赖性激酶1(CDK1)已被确定为寻找新的抗肿瘤药物的潜在靶标。然而,目前尚无临床有效的CDK1抑制剂可用于癌症治疗.因此,这项研究旨在提供潜在的CDK1抑制剂,使用从头产生药物,分子对接,和分子动力学(MD)模拟研究。我们首先利用BREED算法(从头药物生成方法)来产生靶向CDK1的新的小分子文库。初步获得具有良好理化性质和优良成药性的新型潜在CDK1抑制剂,我们对我们生成的文库进行了基于虚拟规则的合理药物筛选,发现了10个初始命中.然后,研究了这十个初始命中和CDK1复合物在其全原子MD模拟(共18μs)和结合姿态元动力学模拟过程中的分子相互作用和动态稳定性,最终命中了五次。此外,另一个具有不同力场的MD模拟(总共2.1μs)证明了五次命中对CDK1的结合能力。发现这五次点击,CBMA001(ΔG=-29.88kcal/mol),CBMA002(ΔG=-34.89kcal/mol),CBMA004(ΔG=-32.47kcal/mol),CBMA007(ΔG=-31.16kcal/mol),和CBMA008(ΔG=-34.78kcal/mol)比阳性化合物Flavopiridol(FLP,ΔG=-25.38kcal/mol)。最后,CBMA002和CBMA004在计算机上被鉴定为优异的选择性CDK1抑制剂。一起,本研究为合理的药物设计和两种有前景的选择性CDK1抑制剂提供了工作流程,值得进一步研究.
    Cyclin-dependent kinases 1 (CDK1) has been identified as a potential target for the search for new antitumor drugs. However, no clinically effective CDK1 inhibitors are now available for cancer treatment. Therefore, this study aimed to offer potential CDK1 inhibitors using de novo drug generation, molecular docking, and molecular dynamics (MD) simulation studies. We first utilized the BREED algorithm (a de novo drug generation approach) to produce a novel library of small molecules targeting CDK1. To initially obtain novel potential CDK1 inhibitors with favorable physicochemical properties and excellent druggability, we performed a virtual rule-based rational drug screening on our generated library and found ten initial hits. Then, the molecular interactions and dynamic stability of these ten initial hits and CDK1 complexes during their all-atom MD simulations (total 18 μs) and binding pose metadynamics simulations were investigated, resulting in five final hits. Furthermore, another MD simulation (total 2.1 μs) with different force fields demonstrated the binding ability of the five hits to CDK1. It was found that these five hits, CBMA001 (ΔG = -29.88 kcal/mol), CBMA002 (ΔG = -34.89 kcal/mol), CBMA004 (ΔG = -32.47 kcal/mol), CBMA007 (ΔG = -31.16 kcal/mol), and CBMA008 (ΔG = -34.78 kcal/mol) possessed much greater binding affinity to CDK1 than positive compound Flavopiridol (FLP, ΔG = -25.38 kcal/mol). Finally, CBMA002 and CBMA004 were identified as excellent selective CDK1 inhibitors in silico. Together, this study provides a workflow for rational drug design and two promising selective CDK1 inhibitors that deserve further investigation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    SARS-CoV-2Mpro(Mpro)是冠状病毒病毒复制中的关键半胱氨酸蛋白酶。茶多酚是有效的Mpro抑制剂。因此,我们的目标是从正热达白(ZHDB)白茶甲醇-水(MW)提取物中分离和合成更多可能抑制COVID-19的新型茶多酚。通过分子网络,共鉴定出33个化合物,并将其分为5个簇。Further,天然产物分子网络(MN)分析表明,MN1具有新的苯丙素取代酯-儿茶素(PSEC),MN5具有重要的碱性复合型羟基肉桂酰儿茶素(HCCs)。因此,分离出一个新的PSEC(1,PSEC636),可以在14个绿茶样品中进一步检测到。合成了一系列HCC(2-6),包括三个新的乙酰化HCC(3-5)。然后我们使用表面等离子体共振(SPR)分析了12种儿茶素与Mpro相互作用的平衡解离常数(KD)。PSEC636(1)的KD值,EGC-C(2),和EC-CDA(3)分别为2.25、2.81和2.44μM,分别。此外,化合物1、2和3显示出潜在的Mpro抑制作用,IC50为5.95±0.17、9.09±0.22和23.10±0.69μM,分别。Further,我们使用诱导配合对接(IFD),约束姿态元动力学(BPMD),和分子动力学(MD)探索Mpro-1的稳定结合状态,表明1可以与氨基酸残基THR26,HIS41,CYS44,TYR54,GLU166和ASP187紧密结合。计算机建模研究表明,酯,乙酰基,和邻苯三酚基团可以提高抑制活性。我们的研究表明,这些儿茶素是有效的Mpro抑制剂,并可能被开发为对抗COVID-19的治疗剂。
    SARS-CoV-2 Mpro (Mpro) is the critical cysteine protease in coronavirus viral replication. Tea polyphenols are effective Mpro inhibitors. Therefore, we aim to isolate and synthesize more novel tea polyphenols from Zhenghedabai (ZHDB) white tea methanol-water (MW) extracts that might inhibit COVID-19. Through molecular networking, 33 compounds were identified and divided into 5 clusters. Further, natural products molecular network (MN) analysis showed that MN1 has new phenylpropanoid-substituted ester-catechin (PSEC), and MN5 has the important basic compound type hydroxycinnamoylcatechins (HCCs). Thus, a new PSEC (1, PSEC636) was isolated, which can be further detected in 14 green tea samples. A series of HCCs were synthesized (2-6), including three new acetylated HCCs (3-5). Then we used surface plasmon resonance (SPR) to analyze the equilibrium dissociation constants (KD) for the interaction of 12 catechins and Mpro. The KD values of PSEC636 (1), EGC-C (2), and EC-CDA (3) were 2.25, 2.81, and 2.44 μM, respectively. Moreover, compounds 1, 2, and 3 showed the potential Mpro inhibition with IC50 5.95 ± 0.17, 9.09 ± 0.22, and 23.10 ± 0.69 μM, respectively. Further, we used induced fit docking (IFD), binding pose metadynamics (BPMD), and molecular dynamics (MD) to explore the stable binding pose of Mpro-1, showing that 1 could tightly bond with the amino acid residues THR26, HIS41, CYS44, TYR54, GLU166, and ASP187. The computer modeling studies reveal that the ester, acetyl, and pyrogallol groups could improve inhibitory activity. Our research suggests that these catechins are effective Mpro inhibitors, and might be developed as therapeutics against COVID-19.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    癌症仍然是一个严重的健康问题,是全世界死亡的主要原因之一。发现人碳酸酐酶IX(hCAIX)在许多癌症类型中过表达,并且对其细胞溶质脱靶同种型具有选择性抑制作用,人类碳酸酐酶II(hCAII),代表了开发新型抗癌化合物的潜在研究领域。这项工作涉及使用各种计算机工具来鉴定基于天然产物的分子,这些分子可以选择性地抑制hCAIX而不是hCAII。对从ZINC15数据库进口的近225,000种天然产物进行MM-GBSA辅助的基于结构的针对hCAIX的虚拟筛选。通过考虑乙酰唑胺来检查获得的命中的效力,hCAIX的结合抑制剂,作为参考分子,和121个分子被鉴定为有效的hCAIX抑制剂。在确保它们的效力之后,交叉对接,其次是MM-GBSA计算hCAII的点击率,被执行,并通过将HCAIX选择性化合物SLC-0111作为参考分子来评估其选择性,和50个天然产物被鉴定为有效的和选择性的hCAIX抑制剂。喹啉支架的分子显示出最高的选择性,它们的选择性归因于锌结合基团(ZBG)与活性位点Zn(II)离子的强静电相互作用。此外,通过进行分子动力学(MD)模拟,确保了顶部hCAIX选择性命中的结合模式的稳定性,这清楚地证明了其中一个入围分子是真正的选择性,因为它不与hCAII的活性位点Zn(II)离子相互作用,但在hCAIX中与该离子强烈相互作用。结合位元动力学研究表明,配体从对接研究预测的结合位点移至更稳定的结合位点,并在该结合位点显示出与蛋白质和Zn(II)的更强相互作用。配体不可能具有生物利用度的问题。因此,该配体可用于生物测定测试,随后用作多种癌症类型的可行治疗方法。由RamaswamyH.Sarma沟通。
    Cancer remains a serious health concern representing one of the leading causes of deaths worldwide. The enzyme human carbonic anhydrase IX (hCA IX) is found to be over-expressed in many cancer types and its selective inhibition over its cytosolic off-target isoform, human carbonic anhydrase II (hCA II), represents a potential area of research in the development of novel anticancer compounds. This work is concerned with the use of various in silico tools for the identification of natural product based molecules that can selectively inhibit hCA IX over hCA II. MM-GBSA assisted structure-based virtual screening against hCA IX was performed for nearly 225,000 natural products imported from the ZINC15 database. The obtained hits were checked for their potency by considering acetazolamide, the bound inhibitor of hCA IX, as the reference molecule, and 121 molecules were identified as potent hCA IX inhibitors. After ensuring their potency, cross-docking, followed by MM-GBSA calculations of the hits with hCA II, was performed, and their selectivity was assessed by considering the hCA IX selective compound SLC-0111 as the reference molecule, and 50 natural products were identified as potent as well as selective hCA IX inhibitors. Molecules with the quinoline scaffold showed the highest selectivity, and their selectivity was attributed to the strong electrostatic interactions of the zinc binding group (ZBG) with the active site Zn(II) ion. Furthermore, the stability of the binding modes of the top hCA IX selective hits was ensured by performing molecular dynamics (MD) simulations, which clearly proved that one of the short-listed molecules is truly selective, as it does not interact with the active site Zn(II) ion of hCA II, but interacts strongly with this ion in hCA IX. Bonding pose metadynamics studies revealed that the ligand moves to a more stable binding site from the one predicted by the docking studies and shows stronger interaction with the protein and Zn(II) at this binding site. The ligand is not likely to have issues with bioavailability. As a result, this ligand can be taken for bioassay testing and subsequently used as a feasible therapeutic treatment for a variety of cancer types. Communicated by Ramaswamy H. Sarma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Background: Indicaxanthin, a betaxanthin belonging to the betalain class of compounds, has been recently demonstrated to exert significant antiproliferative effects inducing apoptosis of human melanoma cells through the inhibition of NF-κB as the predominant pathway. Specifically, Indicaxanthin inhibited IκBα degradation in A375 cells. In resting cells, NF-κB is arrested in the cytoplasm by binding to its inhibitor protein IκBα. Upon stimulation, IκBα is phosphorylated by the IKK complex, and degraded by the proteasome, liberating free NF-κB into the nucleus to initiate target gene transcription. Inhibition of the IKK complex leads to the arrest of the NF-κB pathway. Methods: To acquire details at the molecular level of Indicaxanthin\'s inhibitory activity against hIKKβ, molecular modeling and simulation techniques including induced-fit docking (IFD), binding pose metadynamics (BPMD), molecular dynamics simulations, and MM-GBSA (molecular mechanics-generalized Born surface area continuum solvation) have been performed. Results: The computational calculations performed on the active and inactive form, and the allosteric binding site of hIKKβ, revealed that Indicaxanthin inhibits prevalently the active form of the hIKKβ. MM-GBSA computations provide further evidence of Indicaxanthin\'s stability inside the active binding pocket with a binding free energy of -22.2 ± 4.3 kcal/mol with respect to the inactive binding pocket with a binding free energy of -20.7 ± 4.7 kcal/mol. BPMD and MD simulation revealed that Indicaxanthin is likely not an allosteric inhibitor of hIKKβ. Conclusion: As a whole, these in silico pieces of evidence show that Indicaxanthin can inhibit the active form of the hIKKβ adding novel mechanistic insights on its recently discovered ability to impair NF-κB signaling in melanoma A375 cells. Moreover, our results suggest the phytochemical as a new lead compound for novel, more potent IKKβ inhibitors to be employed in the treatment of cancer and inflammation-related conditions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号