网络药理学是以系统生物学等交叉学科为基础,利用网络分析方法对药物进行多靶点研究设计的新兴学科。网络药理学的出现揭示符合传统中药整体性特征的全新研究范式。目前,网络药理学广泛运用于药学相关的各类研究中,为提升药学相关研究的现代化和科技化水平做出了重要贡献。该文对其基本研究方法和运用以及在中药作用机制、有效成分预测、新药发现、质量控制等方面的应用进行综述。
网络药理学可以从多药物、多成分、多靶点的角度出发构建疾病-靶点-药物的网络关系模型,网络模型的构建有利于从整体水平观察有效成分对疾病靶点网络的作用,以作用强弱为判断标准筛选出对疾病网络起到关键作用的药效成分及相关靶点,针对关键靶点展开实验验证研究可以在提高实验准确性的基础上提升实验效率[3,4]。网络药理学多靶点的研究特点可以为揭示有效成分之间的相互作用关系提供方法学基础,基于网络药理学具有体现药物之间的关系以及疾病靶点间相互作用的特征,利用网络药理学将单一靶点整合为靶点网络,可以深入观察药物对疾病靶点网络的影响[5]。我国学者[6]早期利用网络药理学方法,提出 “疾病网络-生物网络-药物网络”的新概念。该网络的构建将传统“单基因-单疾病”模式转化为“多基因多疾病”的全新观点,以生物靶点为桥梁构建药物与病症之间的关系网。以诸多单味中药中所含的化学成分已分离、鉴定明确,有效成分在体内的生物过程具有初步的研究结果为基础,提出的“药物网络-疾病网络”模型,将中药对应的寒热证候细化为生物体内蛋白质之间的相互作用关系[7],提供中药针对寒热证候的具体药效作用机制。利用网络药理学方法可以构建中药靶点网络模型,将传统单一靶点研究模式转化为多靶点整体研究模式,系统观察药物对靶点网的干预以及影响,同时使中药药效理论抽象化为相关靶点间的作用,深入了解蛋白质之间的相互作用关系,为中药的系统性研究提供详细的理论依据。
网络分析方法是以网络模型为基础建立的一种网络药理学方法,可以对特定的疾病网络进行相应网络分析,寻找网络中所有可能受到药物干预的靶点群或关键靶点,以便深入探索该类靶点对整体网络的影响。同时可对筛选所得的靶点进行网络功能分析,阐明药物之间以及有效成分与疾病靶点间的相互作用机制[8,9]。LI等[10]通过网络分析方法,对特定靶点之间的相互关系进行研究,阐明药物多成分之间相互作用的潜在机制,揭示中药多成分多靶点作用的独特之处及其重要性。网络分析方法可以针对特定的药物及疾病网络选取适当的分析方法,从不同角度对靶点的重要性进行评价,为关键靶点的选取提供可靠性依据。通过对重要靶点的筛选及评价可以较好地阐释药物分子水平的相互作用机制,为通过调控少数靶点影响整体网络的进一步研究提供依据[11,12,13]。
随着网络药理学相关技术的逐步成熟,网络药理学方法广泛运用于各类研究中。WU等[14]以网络药理学方法为基础,通过网络分析对G蛋白耦联受体相关药物展开系列研究。实验结果显示,两种通过网络药理学方法预测所得的G蛋白耦联受体药物对前列腺素具有很高的亲和力。研究结果为G蛋白耦联受体相关药物的研究提供有效的网络研究基础。ENGIN等[15]利用网络药理学方法构建 “蛋白质相互作用网络”模型,并进一步对模型进行网络分析后发现对蛋白质网络进行整体干预比干预单一靶点更能揭示生物系统性的变化。已有研究表明[16],药物干预整体网络是通过破坏结构相似蛋白质之间的相互联系,达到阻止通路正常运行或整体破坏网络的目的。
网络药理学运用于现代研究的思路是从整体水平出发,以网络模型为基础,利用适当的分析手段系统地刻画干预措施对局部或整体网络的作用与影响,从而进一步推测特殊靶点对于整体网络的重要作用。将单一靶点研究进一步扩大范围,把研究所需靶点构建为网络作为观测对象,是网络药理学发展的全新研究模式的中心思想。
中药的药理作用较为复杂,且中药具有多成分、多靶点的特征,是中药区别于西药的独特之处,而中药含有多种成分的复杂性使得难以对中药展开全面、系统的研究。网络药理学是以整体网络为研究对象,观察药物对网络的干预与影响,将整体网络的研究与中药的整体性特征结合,可以从多途径、多成分、多靶点的角度阐明中药研究的基础理论,为中药现代化研究提供全新的思路。网络药理学在中药研究领域中起到重要的作用,将网络药理学的研究范式融入到中药相关科学研究中能够提升中药研究的现代化水平。
利用网络药理学方法可以构建中药、疾病靶点网络模型,并通过靶点之间的相互关系将中药与疾病靶点有效地衔接起来,进而对中药有效成分的药效作用机制进行系统化探索,揭示中药的整体药效特征。ZHANG等[17]利用网络药理学方法构建蛋白质相互作用网,富集分析确定研究所需具体信号通路后进行实验验证,表明川芎嗪减轻氧化损伤的作用机制可能是通过川芎嗪抑制磷酸二酯酶的作用实现的。LV等[18]构建药物蛋白质相互关系作用网后从相关网络区域中选取关键靶点,对五味子素抗脑血管疾病的分子靶点及信号通路进行预测分析及验证,表明通过网络药理学方法可以挖掘出对疾病起到重要作用的基因靶点,发现五味子针对脑血管相关疾病的特定信号通路。HUANG等[19]对玄归滴丸的相关靶点进行了探索研究,通过OB、DL等指标筛选候选化合物,找出化合物对应靶点及信号通路后构建网络模型进行分析,阐明玄归滴丸中具有镇痛作用的相应化合物及其对应的分子靶点,为解释复方镇痛疗效的作用机制提供重要依据。
目前,以单一靶点为目标探讨癌症作用机制的研究模式尚未取得突破,而中药的整体性作用特征有望成为研究此类疾病新的突破点。利用网络药理学可从多靶点的角度着手探索抗癌作用的相关机制,将中药研究与癌症研究相结合,发挥中药在防治癌症方面起到的重要作用。POORNIMA等[20]对癌细胞生物分子网络以及肿瘤干细胞的生物代谢过程进行深入探讨后,发现大多数肿瘤干细胞的信号传导以及转录过程都具有亲和力低、时间短以及信号微弱等特点,基于上述发现,判断肿瘤干细胞之间是以复杂的作用机制的形式存在并发挥作用的。周志敏等[21]通过构建子宫内膜癌疾病靶点网络,结合网络药理学和分子对接方法,对补中益气汤的作用机制进行研究,表明部分化合物对应靶点与子宫内膜癌靶点有显著关联性。天然药物具有复杂的机制网络,并非通过单一途径起到治疗作用,网络药理学可以对癌症靶点网络进行预测,能够从多靶点的途径探索抗癌作用机制,将有效成分与靶点研究结合,挖掘中药在防治癌症方面具有的潜在优势。
传统的药物研究模式多以单靶点为目标展开相关探索,然而中药具有的多成分特征使得利用单一靶点研究模式难以系统性地阐明中药中包含的有效成分。采用网络药理学方法构建符合中药特点的网络模型,可以为中药的现代化研究提供全新的概念和方法。TIAN等[22]通过可视化软件对蒙药Sendeng-4所含的化学成分及对应靶点进行了相关研究,分析靶点所涉及的关键信号通路发现不同成分对不同的靶点产生的影响具有差异性,并从中筛选得到3个较为关键的作用靶点。TAO等[23]对抗心血管疾病药材的潜在靶点进行探索,表明58种关键活性成分所对应的靶点数量各不相同,从分子水平揭示传统复方“君臣佐使”的配伍作用原理。采用网络药理学方法以药材以及疾病靶点为中心构建网络模型,选择合适的网络分析方法对网络模型中大量的靶点数据进行分析和筛选,可以对化合物的有效性进行初步评价,在最大可能发现药物所含潜在药效成分的同时,为后期的实验验证提供较为可靠的依据,从而提高实验的成功率。
网络药理学具有针对不同功能的靶点评价指标,根据研究目标选取合适的网络分析方法对大量中药进行分析预测,可以从数据库中筛选出与疾病网络具有密切联系的药材种类,为后期的实验研究提供重要依据和基础。KE等[24]采用分子对接与网络分析的方法对治疗神经退行性疾病的相关药物进行新药预测研究,观察不同药物对疾病网络的干预与影响,表明有504种草药具有治疗神经退行性疾病的潜力。目前,新药所面临的问题是有效性低及毒副作用大等缺陷[25],因此新药在临床阶段的发展受到了限制,导致药物研究和开发生产力下降,为了提高药物生产的有效性以及消除潜在的毒副作用, HOPKINS等[26]提出以网络药理学为主要手段建立更为有效的新药研究模式,将网络药理学方法融入到对新药的临床应用及开发研究中,可以为新药的发现及优化提供现代化的思考方法。网络药理学以其整体网络模型为基础,多种预测方法为分析手段,对新药的发现及潜在药物的挖掘提供合理的实验依据以及全新的研究思路,提升了新药研究的准确性及可靠性。
药材的质量控制及毒理学研究能够以网络药理学方法为基础建立系统化的研究体系,得到更为精确、系统的检测结果,为中药质量控制及毒性研究提供新思路、新方法。研究显示中药对治疗或预防轻微疾病以及严重慢性病的使用量正在持续增加,但中药不具备确切及完善的系统对其质量问题进行保障,所以推出新的传统中药质量评价制度是确保上市产品符合质量和安全标准的关键[27]。PELKONEN等[28]通过基因组工具以及网络药理学方法对传统药物质量检测方式进行革新研究,新的检测方式强调中药之间相互作用的特点,突出中药特有的系统化、整体性特征。LI等[29]综合文献数据,对天然药物的相互关系、化学成分以及药理、毒理学机制等进行了研究整合,结合与中药相关的各类技术基础,为中药的深度机制研究创造了新的机会。
网络药理学在国内研究已达领先水平,但发展时间不足十年,面临着诸多挑战。网络药理学是一种新兴的研究方式,网络药理学的思维模式可以体现传统中药独有的整体性优势,与中药相关的各类数据库在逐渐形成较为完整的体系,但由于技术发展相对不够成熟,存在数据量涵盖范围不够广、数据精准度不够高、构建网络所获数据仅局限于目前研究所得数据等问题,建立一个较为完整的中药相关数据平台,对药物相关数据进行实验验证优化提高实验数据的可靠性,构建完善的蛋白质相互作用关系网和更为全面的信号通路数据库以蛋白质及信号通路间的相互关系探索全新的有效靶点是目前面临的新挑战。
中药配伍原则“君臣佐使”是中药方剂的精髓,分别起到治疗主症、辅助君药、治疗兼症以及引导药物的作用,传统的组方原则所针对的并非单一的疾病网络,网络药理学与中药研究的结合,有望从观察药物对疾病靶点网络影响的角度间接揭示复方独有的配伍特征,对中药方剂的现代化研究起到重要作用。运用网络药理基于疾病-靶点-药物的研究方法可以探究我国独具特色的民族药潜在的药用价值,同时为各类民族药的整理、发掘、研究工作提供全新的研究模式。随着基因、蛋白质数据的不断积累以及多种学科知识的融合,网络药理学将成为中药研究的新范式,这种从多靶点着手探讨药物机制研究的理念是探索众多疾病思维模式的新突破,虽然网络药理学运用于中医药的发展处于起步阶段,但这一模式的重大进步将为中药研究方式的概念转变以及中医药现代化发展做出重要贡献。
[1] |
|
[2] |
网络药理学是指将药物作用网络与生物网络整合在一起,分析药物在此网络中与特定节点或模块的相互作用关系,从而理解药物和机体相互作用的科学。网络药理学突破传统的“一个药物一个靶标,一种疾病”理念,代表了现代生物医药研究的哲学理念与研究模式的转变。以系统生物学和网络生物学基本理论为基础的网络药理学具有整体性、系统性的特点,注重网络平衡(或鲁棒性)和网络扰动,强调理解某个单一生物分子(如基因、mRNA或蛋白等)在生物体系中的生物学地位和动力学过程要比理解其具体生物功能更为重要,揭示药物作用的生物学和动力学谱要比揭示其作用的单个靶标或几个“碎片化”靶标更重要,对认识药物和发现药物的理念产生了深远影响。
[本文引用:1]
|
[3] |
|
[4] |
The purpose of this study was to investigate the therapeutic mechanism(s) ofClematis chinensisOsbeck/Notopterygium incisumK.C. Ting ex H.T (CN). A network pharmacology approach integrating prediction of ingredients, target exploration, network construction, module partition and pathway analysis was used. This approach successfully helped to identify 12 active ingredients of CN, interacting with 13 key targets (Akt1, STAT3, TNFsf13, TP53, EPHB2, IL-10, IL-6, TNF, MAPK8, IL-8, RELA, ROS1 and STAT4). Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that CN-regulated pathways were mainly classified into signal transduction and immune system. The present work may help to illustrate the mechanism(s) of action of CN, and it may provide a better understanding of antirheumatic effects.
[本文引用:1]
|
[5] |
该文通过网络药理学研究预测复方钩藤降压片潜在的主要活性成分和作用靶点,探讨其多成分-多靶点-多通路的治疗高血压的可能作用机制。采用整合药理学平台(TCMIP)构建复方钩藤降压片成分靶标-疾病靶标网络,通过网络分析方法筛选关键节点,并在此基础上对关键节点进行通路富集分析,探索复方钩藤降压片治疗高血压可能参与的生物过程。靶点网络特征分析显示,所预测复方钩藤降压片中有35个活性成分与前列腺素内源性过氧化物合酶(PTGS1,PTGS2)、ATP合成酶(ATP1A1,ATP5A1,ATP5C1,ATP5B)等29个主要蛋白有很强的相互作用,富集到血压调节、G-蛋白耦合受体激活、心肌细胞肾上腺素能信号转导和血小板激活等15条通路,参与对高血压病理过程不同环节的调控。该研究初步揭示了复方钩藤降压片治疗高血压的潜在活性成分及其可能的作用机制,为进一步的药效物质基础和作用机制实验研究提供了理论依据。
[本文引用:1]
|
[6] |
|
[7] |
Abstract Traditional Chinese medicine uses ZHENG as the key pathological principle to understand the human homeostasis and guide the applications of Chinese herbs. Here, a systems biology approach with the combination of computational analysis and animal experiment is used to investigate this complex issue, ZHENG, in the context of the neuro-endocrine-immune (NEI) system. By using the methods of literature mining, network analysis and topological comparison, it is found that hormones are predominant in the Cold ZHENG network, immune factors are predominant in the Hot ZHENG network, and these two networks are connected by neuro-transmitters. In addition, genes related to Hot ZHENG-related diseases are mainly present in the cytokine-cytokine receptor interaction pathway, whereas genes related to both the Cold-related and Hot-related diseases are linked to the neuroactive ligand-receptor interaction pathway. These computational findings were subsequently verified by experiments on a rat model of collagen-induced arthritis, which indicate that the Cold ZHENG-oriented herbs tend to affect the hub nodes in the Cold ZHENG network, and the Hot ZHENG-oriented herbs tend to affect the hub nodes in the Hot ZHENG network. These investigations demonstrate that the thousand-year-old concept of ZHENG may have a molecular basis with NEI as background.
[本文引用:1]
|
[8] |
Drugs designed to act against individual molecular targets cannot usually combat multigenic diseases such as cancer, or diseases that affect multiple tissues or cell types such as diabetes and immunoinflammatory disorders. Combination drugs that impact multiple targets simultaneously are better at controlling complex disease systems, are less prone to drug resistance and are the standard of care in many important therapeutic areas. The combination drugs currently employed are primarily of rational design, but the increased efficacy they provide justifies in vitro discovery efforts for identifying novel multi-target mechanisms. In this review, we discuss the biological rationale for combination therapeutics, review some existing combination drugs and present a systematic approach to identify interactions between molecular pathways that could be leveraged for therapeutic benefit.
[本文引用:1]
|
[9] |
http://www.nature.com/doifinder/10.1038/nrd1609
[本文引用:1]
|
[10] |
Background Deciphering the metabolome is essential for a better understanding of the cellular metabolism as a system. Typical metabolomics data show a few but significant correlations among metabolite levels when data sampling is repeated across individuals grown under strictly controlled conditions. Although several studies have assessed topologies in metabolomic correlation networks, it remains unclear whether highly connected metabolites in these networks have specific functions in known tissue- and/or genotype-dependent biochemical pathways. Results In our study of metabolite profiles we subjected root tissues to gas chromatography-time-of-flight/mass spectrometry (GC-TOF/MS) and used published information on the aerial parts of 3 Arabidopsis genotypes, Col-0 wild-type, methionine over-accumulation 1 (mto1), and transparent testa4 (tt4) to compare systematically the metabolomic correlations in samples of roots and aerial parts. We then applied graph clustering to the constructed correlation networks to extract densely connected metabolites and evaluated the clusters by biochemical-pathway enrichment analysis. We found that the number of significant correlations varied by tissue and genotype and that the obtained clusters were significantly enriched for metabolites included in biochemical pathways. Conclusions We demonstrate that the graph-clustering approach identifies tissue- and/or genotype-dependent metabolomic clusters related to the biochemical pathway. Metabolomic correlations complement information about changes in mean metabolite levels and may help to elucidate the organization of metabolically functional modules.
[本文引用:1]
|
[11] |
Chemotherapies, HIV infections, and treatments to block organ transplant rejection are creating a population of immunocompromised individuals at serious risk of systemic fungal infections. Since single-agent therapies are susceptible to failure due to either inherent or acquired resistance, alternative therapeutic approaches such as multi-agent therapies are needed. We have developed a bioinformatics-driven approach that efficiently predicts compound synergy for such combinatorial therapies. The approach uses chemogenomic profiles in order to identify compound profiles that have a statistically significant degree of similarity to a fluconazole profile. The compounds identified were then experimentally verified to be synergistic with fluconazole and with each other, in both Saccharomyces cerevisiae and the fungal pathogen Candida albicans. Our method is therefore capable of accurately predicting compound synergy to aid the development of combinatorial antifungal therapies.SynopsisDrugs that act against individual molecular targets are often insufficient to combat fungal infections, multigenic diseases, and multiple cell or tissue type diseases (White et al, 1998; Sams-Dodd, 2005; Onyewu and Heitman, 2007; Zimmermann et al, 2007). Combinatorial therapies that impact multiple targets simultaneously are less prone to the development of drug resistance, and increase therapeutic efficacy (Groll and Walsh, 2002; Zimmermann et al, 2007). One of the major benefits of combinatorial therapies is the potential for synergistic effects: that is, the overall therapeutic benefit of the drug combination is greater than the sum of the effects of the drugs individually. These advantages have driven drug discovery efforts towards the search for combinatorial therapies (Borisy et al, 2003; Fitzgerald et al, 2006; Onyewu and Heitman, 2007; Zimmermann et al, 2007).Large-scale searches have demonstrated that high-throughput screens of thousands of compounds can be straightforward (Zhang et al, 2007), but it is unlikely that experimental techniques will be sufficient to survey the complete combinatorial chemical space in a cost-effective and timely manner. Nelander et al (2008) attempted to use data from perturbation screens and prior knowledge regarding the targets of compounds to model the effects of these compounds when they are used alone or in combination. This approach is currently limited to compounds with known targets, but such an approach could potentially be extended to predict synergistic compound pairs. However, there remains a clear need for an approach that reduces the vast combinatorial chemical space to a set of combinations that is sufficiently small for experimental testing yet enriched with synergistic combinations.We introduce here a combined experimental and bioinformatics approach to identify synergistic compound pairs for antifungal combinatorial therapies (Figure 1). Each compound is represented in silico by its chemogenomic profile, which we define as the set of genes corresponding to the single gene deletions in Saccharomyces cerevisiae that confer hypersensitivity to the compound. Therefore, we collected from the literature 1300 chemogenomic profiles generated with a broad range of compounds.We then evaluated a measure of chemogenomic profile similarity for its ability to predict antifungal synergy, where a compound pair is predicted to be synergistic if the measured similarity between the corresponding profiles is sufficiently large. A gold standard set of positive and negative examples of antifungal synergy was assembled for this purpose. The similarity measure quantifies the significance of the overlap between two hypersensitive gene sets and the enrichment of its predictions with true synergies (i.e. positive examples in the gold standard set) is significant relative to the expected baseline levels (P=0.0236). These results suggest that chemogenomic profile similarity predicts antifungal synergy.Fluconazole is a fungistatic drug, and it is thus possible for fungal cells to recover from treatment due to acquired drug resistance (Cowen et al, 2002). However, the drug has favourable pharmacokinetic and toxicological properties (Grant and Clissold, 1990), and would thus be an ideal constituent compound of a combinatorial antifungal therapy. Therefore, we applied our method of predicting antifungal synergy by first generating a de novo chemogenomic profile for fluconazole, which we call the FCZ-Fungicidal profile (Figure 1B). The FCZ-Fungicidal profile specifies the set of genes corresponding to deletions that are lethal in the presence of the drug.In the next step of our method, we found that eight compounds have profiles that are sufficiently similar to the FCZ-Fungicidal profile and are thus predicted to be synergistic with fluconazole. We noticed that many of these compounds are also predicted to be synergistic with each other. In the final step of our method, we thus experimentally tested predicted synergistic combinations involving fluconazole and pairings of the predicted fluconazole partners for antifungal synergy.The combinations were tested in both S. cerevisiae and the fungal pathogen Candida albicans using dose-matrix response assays that measure the growth arrest and monitor the death of treated cells. We showed that eight synergistic combinations identified in S. cerevisiae are also synergistic in C. albicans, and we identified three and two additional synergies only in S. cerevisiae and only in C. albicans, respectively. We also tested the novel synergistic combination of fluconazole (FDA-approved) and wortmannin (analogues are in clinical trials; Noble et al, 2004) in two fluconazole-resistant clinical isolates of C. albicans (Morschhauser et al, 2007; Dunkel et al, 2008). The compounds act synergistically to kill the cells of both isolates (Figure 5), suggesting potential clinical relevance. Taken together, the validation success rate for the predictor of antifungal synergy (69%) implies that our method identifies true synergies at a rate that is 鈭20-fold better than the estimated rate for testing randomly selected compound pairs (Borisy et al, 2003).In summary, we have shown that our method is capable of rapidly and accurately predicting compound pairs that exhibit antifungal synergy. Therefore, our method may enable efficient development of combinatorial therapies to attack the persisting problem of drug-resistant C. albicans strains in the clinic.
[本文引用:1]
|
[12] |
Given the complex nature of biological systems, pathways often need to function in a coordinated fashion in order to produce appropriate physiological responses to both internal and external stimuli. Therefore, understanding the interaction and crosstalk between pathways is important for understanding the function of both cells and more complex systems.We have developed a computational approach to detect crosstalk among pathways based on protein interactions between the pathway components. We built a global mammalian pathway crosstalk network that includes 580 pathways (covering 4753 genes) with 1815 edges between pathways. This crosstalk network follows a power-law distribution: P(k) approximately k(-)(gamma), gamma = 1.45, where P(k) is the number of pathways with k neighbors, thus pathway interactions may exhibit the same scale-free phenomenon that has been documented for protein interaction networks. We further used this network to understand colorectal cancer progression to metastasis based on transcriptomic data.yong.2.li@gsk.comSupplementary data are available at Bioinformatics online.
[本文引用:1]
|
[13] |
Abstract Kinases and proteases are responsible for two fundamental regulatory mechanisms--phosphorylation and proteolysis--that orchestrate the rhythms of life and death in all organisms. Recent studies have highlighted the elaborate interplay between both post-translational regulatory systems. Many intracellular or pericellular proteases are regulated by phosphorylation, whereas multiple kinases are activated or inactivated by proteolytic cleavage. The functional consequences of this regulatory crosstalk are especially relevant in the different stages of cancer progression. What are the clinical implications derived from the fertile dialogue between kinases and proteases in cancer?
[本文引用:1]
|
[14] |
Abstract G protein-coupled receptors (GPCRs) are the largest super family with more than 800 membrane receptors. Currently, over 30% of the approved drugs target human GPCRs. However, only approximately 30 human GPCRs have been resolved three-dimensional crystal structures, which limits traditional structure-based drug discovery. Recent advances in network-based systems pharmacology approaches have demonstrated powerful strategies for identifying new targets of GPCR ligands. In this study, we proposed a network-based systems pharmacology framework for comprehensive identification of new drug-target interactions on GPCRs. Specifically, we reconstructed both global and local drug-target interaction networks for human GPCRs. Network analysis on the known drug-target networks showed rational strategies for designing new GPCR ligands and evaluating side effects of the marketed GPCR drugs. We further built global and local network-based models for predicting new targets of the known GPCR ligands. The area under the receiver operating characteristic curve of more than 0.96 was obtained for the best network-based models in cross validation. In case studies, we identified that several network-predicted GPCR off-targets (e.g. ADRA2A, ADRA2C and CHRM2) were associated with cardiovascular complications (e.g. bradycardia and palpitations) of the marketed GPCR drugs via an integrative analysis of drug-target and off-target-adverse drug event networks. Importantly, we experimentally validated that two newly predicted compounds, AM966 and Ki16425, showed high binding affinities on prostaglandin E2 receptor EP4 subtype with IC 50 =2.67 M and 6.34 M, respectively. In summary, this study offers powerful network-based tools for identifying polypharmacology of GPCR ligands in drug discovery and development. Copyright 2017 Elsevier Ltd. All rights reserved.
[本文引用:1]
|
[15] |
Networks are increasingly used to study the impact of drugs at the systems level. From the algorithmic standpoint, a drug can "attack" nodes or edges of a protein-protein interaction network. In this work, we propose a new network strategy, "The Interface Attack", based on protein-protein interfaces. Similar interface architectures can occur between unrelated proteins. Consequently, in principle, a drug that binds to one has a certain probability of binding to others. The interface attack strategy simultaneously removes from the network all interactions that consist of similar interface motifs. This strategy is inspired by network pharmacology and allows inferring potential off-targets. We introduce a network model that we call "Protein Interface and Interaction Network (P2IN)", which is the integration of protein-protein interface structures and protein interaction networks. This interface-based network organization clarifies which protein pairs have structurally similar interfaces and which proteins may compete to bind the same surface region. We built the P2IN with the p53 signaling network and performed network robustness analysis. We show that (1) "hitting" frequent interfaces (a set of edges distributed around the network) might be as destructive as eleminating high degree proteins (hub nodes), (2) frequent interfaces are not always topologically critical elements in the network, and (3) interface attack may reveal functional changes in the system better than the attack of single proteins. In the off-target detection case study, we found that drugs blocking the interface between CDK6 and CDKN2D may also affect the interaction between CDK4 and CDKN2D.
[本文引用:1]
|
[16] |
Abstract: Systems as diverse as genetic networks or the world wide web are best described as networks with complex topology. A common property of many large networks is that the vertex connectivities follow a scale-free power-law distribution. This feature is found to be a consequence of the two generic mechanisms that networks expand continuously by the addition of new vertices, and new vertices attach preferentially to already well connected sites. A model based on these two ingredients reproduces the observed stationary scale-free distributions, indicating that the development of large networks is governed by robust self-organizing phenomena that go beyond the particulars of the individual systems.
[本文引用:1]
|
[17] |
[Display omitted]
[本文引用:1]
|
[18] |
To illuminate the molecular targets for schisandrin against cerebrovascular disease based on the combined methods of network pharmacology prediction and experimental verification. A protein database was established through constructing the drug-protein network from literature mining data. The protein-protein network was built through an in-depth exploration of the relationships between the proteins. The computational platform was implemented to predict and extract the sensitive sub-network with significant P-values from the protein-protein network. Then the key targets and pathways were identified from the sensitive sub-network. The most related targets and pathways were also confirmed in hydrogen peroxide (H2O2)-induced PC12 cells by Western blotting. Twelve differentially expressed proteins (gene names: NFKB1, RELA, TNFSF10, MAPK1, CHUK, CASP8, PIGS2, MAPK14, CREB1, IFNG, APP, and BCL2) were confirmed as the central nodes of the interaction network (45 nodes, 93 edges). The NF-B signaling pathway was suggested as the most related pathway of schisandrin for cerebrovascular disease. Furthermore, schisandrin was found to suppress the expression and phosphorylation of IKK, as well as p50 and p65 induced by H2O2 in PC12 cells by Western blotting. The computational platform that integrates literature mining data, protein-protein interactions, sensitive sub-network, and pathway results in identification of the NF-B signaling pathway as the key targets and pathways for schisandrin.
[本文引用:1]
|
[19] |
This study aimed to evaluate the clinical analgesic efficacy and identify the molecular targets of XGDP for treating primary dysmenorrhea (PD) by a network pharmacology approach. Analysis of pain disappearance rate of XGDP in PD treatment was conducted based on data from phase II and III randomized, double-blind, double-simulation, and positive parallel controlled clinical trials. The bioactive compounds were obtained by the absorption, distribution, metabolism, and excretion processes with oral bioavailability (OB) and drug-likeness (DL) evaluation. Subsequently, target prediction, pathway identification, and network construction were employed to clarify the mechanisms of the analgesic effect of XGDP on PD. The pain disappearance rates in phase II and III clinical trials of XGDP in PD treatment were 62.5% and 55.8%, respectively, yielding a significant difference (P<0.05) when compared with the control group using Tongjingbao granules (TJBG). Among 331 compounds, 53 compounds in XGDP were identified as the active compounds related to PD through OB, DL, and target prediction. The active compounds and molecular targets of XGDP were identified, and our study showed that XGDP may exert its therapeutic effects on PD through the regulation of the targets related to anti-inflammation analgesia and central analgesia and relieving smooth muscle contraction.
[本文引用:1]
|
[20] |
Despite massive investments in drug research and development, the significant decline in the number of new drugs approved or translated to clinical use raises the question, whether single targeted drug discovery is the right approach. To combat complex systemic diseases that harbour robust biological networks such as cancer, single target intervention is proved to be ineffective. In such cases, network pharmacology approaches are highly useful, because they differ from conventional drug discovery by addressing the ability of drugs to target numerous proteins or networks involved in a disease. Pleiotropic natural products are one of the promising strategies due to their multi-targeting and due to lower side effects. In this review, we discuss the application of network pharmacology for cancer drug discovery. We provide an overview of the current state of knowledge on network pharmacology, focus on different technical approaches and implications for cancer therapy (e.g.polypharmacology and synthetic lethality), and illustrate the therapeutic potential with selected examples green tea polyphenolics,Eleutherococcus senticosus, Rhodiola rosea,andSchisandra chinensis). Finally, we present future perspectives on their plausible applications for diagnosis and therapy of cancer.
[本文引用:1]
|
[21] |
目的:通过网络药理学和计算机辅助药物设计,找出补中益气汤的潜能成分,并探讨其成为治疗子宫内膜癌的药物的可能性.方法:从DisGeNET和BisoGenet应用程序中收集与子宫内膜癌相关的基因和蛋白质以及从中医药数据库中收集复方中药补中益气汤的成分,运用Cytoscape建立网络关系图,并用CytoNCA筛选出能影响子宫内膜癌的蛋白质.再运用Surflex-Dock对补中益气汤的10种中草药的534个成分与子宫内膜癌相关的蛋白质进行柔性分子对接及分析其在活性位点的结合模式,并使用FAF-Drugs3和OSIRIS进行分子结构的筛选以预测其吸收、分布、代谢、排泄和毒性(ADMET).结果:补中益气汤的六氢西红柿红素、八氢西红柿红素、新橙皮苷和橙皮苷这4个化合物与子宫内膜癌相关的蛋白质有好的结合亲和力与良好的药物特性.结论:从研究和文献中发现这4个化合物与癌症相关,有望进一步研究成为治疗子宫内膜癌的药物.
[本文引用:1]
|
[22] |
We collected the data on the Sendeng-4 chemical composition corresponding targets through the literature and from DrugBank, SuperTarget, TTD (Therapeutic Targets Database) and other databases and the relevant signaling pathways from the KEGG (Kyoto Encyclopedia of Genes and Genomes) database and established models of the chemical composition-target network and chemical composition-target-disease network using Cytoscape software, the analysis indicated that the chemical composition had at least nine different types of targets that acted together to exert effects on the diseases, suggesting a ulti-component, multi-target feature of the traditional Mongolian medicine. We also employed the rat model of rheumatoid arthritis induced by Collgen Type II to validate the key targets of the chemical components of Sendeng-4, and three of the key targets were validated through laboratory experiments, further confirming the anti-inflammatory effects of Sendeng-4. In all, this study predicted the active ingredients and targets of Sendeng-4, and explored its mechanism of action, which provided new strategies and methods for further research and development of Sendeng-4 and other traditional Mongolian medicines as well.
[本文引用:1]
|
[23] |
中国科学院机构知识库(CAS IR GRID)以发展机构知识能力和知识管理能力为目标,快速实现对本机构知识资产的收集、长期保存、合理传播利用,积极建设对知识内容进行捕获、转化、传播、利用和审计的能力,逐步建设包括知识内容分析、关系分析和能力审计在内的知识服务能力,开展综合知识管理。
[本文引用:1]
|
[24] |
Neurodegenerative diseases, referring to as the progressive loss of structure and function of neurons, constitute one of the major challenges of modern medicine. Traditional Chinese herbs have been used as a major preventive and therapeutic strategy against disease for thousands years. The numerous species of medicinal herbs and Traditional Chinese Medicine (TCM) compound formulas in nervous system disease therapy make it a large chemical resource library for drug discovery. In this work, we collected 7362 kinds of herbs and 58,147 Traditional Chinese medicinal compounds (Tcmcs). The predicted active compounds in herbs have good oral bioavailability and central nervous system (CNS) permeability. The molecular docking and network analysis were employed to analyze the effects of herbs on neurodegenerative diseases. In order to evaluate the predicted efficacy of herbs, automated text mining was utilized to exhaustively search in PubMed by some related keywords. After that, receiver operator characteristic (ROC) curves was used to estimate the accuracy of predictions. Our study suggested that most herbs were distributed in family of Asteraceae, Fabaceae, Lamiaceae and Apocynaceae. The predictive model yielded good sensitivity and specificity with the AUC values above 0.800. At last, 504 kinds of herbs were obtained by using the optimal cutoff values in ROC curves. These 504 herbs would be the most potential herb resources for neurodegenerative diseases treatment. This study would give us an opportunity to use these herbs as a chemical resource library for drug discovery of anti-neurodegenerative disease.
[本文引用:1]
|
[25] |
DOI:10.1038/nrd1470
[本文引用:1]
|
[26] |
The dominant paradigm in drug discovery is the concept of designing maximally selective ligands to act on individual drug targets. However, many effective drugs act via modulation of multiple proteins rather than single targets. Advances in systems biology are revealing a phenotypic robustness and a network structure that strongly suggests that exquisitely selective compounds, compared with multitarget drugs, may exhibit lower than desired clinical efficacy. This new appreciation of the role of polypharmacology has significant implications for tackling the two major sources of attrition in drug development09”efficacy and toxicity. Integrating network biology and polypharmacology holds the promise of expanding the current opportunity space for druggable targets. However, the rational design of polypharmacology faces considerable challenges in the need for new methods to validate target combinations and optimize multiple structure-activity relationships while maintaining drug-like properties. Advances in these areas are creating the foundation of the next paradigm in drug discovery: network pharmacology.
[本文引用:1]
|
[27] |
Abstract Top of page Abstract Introduction Efficacy Safety The future for complementary medicines References This is the second of two papers which review issues concerning complementary medicines. The first reviewed the extent of use of complementary medicines, and issues related to the regulation and pharmaceutical quality of these products; the second considers evidence for the efficacy of several well-known complementary medicines, and discusses complementary-medicines pharmacovigilance. The term complementary medicines describes a range of pharmaceutical-type preparations, including herbal medicines, homoeopathic remedies, essential oils and dietary supplements, which mainly sit outside conventional medicine. The use of complementary medicines is a popular healthcare approach in the UK, and there are signs that the use of such products is continuing to increase. Patients and the public use complementary medicines for health maintenance, for the treatment or prevention of minor ailments, and also for serious, chronic illnesses. There is a growing body of evidence from randomized controlled trials and systematic reviews to support the efficacy of certain herbal extracts and dietary supplements in particular conditions. However, many other preparations remain untested. Strictly speaking, evidence of efficacy (and safety) for herbal medicines should be considered to be extract specific. Pharmacovigilance for complementary medicines is in its infancy. Data are lacking in several areas relevant to safety. Standard pharmacovigilance tools have additional limitations when applied to investigating safety concerns with complementary medicines.
[本文引用:1]
|
[28] |
Research on herbal medicinal products is increasingly published in estern scientific journals dedicated primarily to conventional medicines. Publications are concerned mainly not only on the issues of safety and interactions, but also on efficacy. In reviews, a recurring complaint has been a lack of quality studies. In this opinion article, we present the case of Chinese herbal medicines as an example, as they have been extensively used in the global market and increasingly studied worldwide. We analyze the potential reasons for problems and propose some ways forward. As in the case of any drug, clinical trials for safety, efficacy, and/or effectiveness are the ultimate demonstration of therapeutic usefulness of herbal products. These will only make scientific sense when the tested herbal products are authentic, standardized, and quality controlled, if good practice guidelines of evidence-based medicine are followed, and if relevant controls and outcome measures are scientifically defined. Herbal products are complex mixtures, and for such complexity, an obvious approach for mechanistic studies is network pharmacology based on omic tools and approaches, which has already begun to revolutionize the study of conventional drugs, emphasizing networks, interactions, and polypharmacological features behind the action of many drugs.
[本文引用:1]
|
[29] |
Multi-target therapeutics is a promising paradigm for drug discovery which is expected to produce greater levels of efficacy with fewer adverse effects and toxicity than monotherapies. Medical herbs featuring multi-components and multi-targets may serve as valuable resources for network-based multi-target drug discovery.In this study, we report an integrated systems pharmacology platform for drug discovery and combination, with a typical example applied to herbal medicines in the treatment of cardiovascular diseases.First, a disease-specific drug-target network was constructed and examined at systems level to capture the key disease-relevant biology for discovery of multi-targeted agents. Second, considering an integration of disease complexity and multilevel connectivity, a comprehensive database of literature-reported associations, chemicals and pharmacology for herbal medicines was designed. Third, a large-scale systematic analysis combining pharmacokinetics, chemogenomics, pharmacology and systems biology data through computational methods was performed and validated experimentally, which results in a superior output of information for systematic drug design strategies for complex diseases.This strategy integrating different types of technologies is expected to help create new opportunities for drug discovery and combination.
[本文引用:1]
|