Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Acta Pharmacol Sin ; 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38902503

RESUMO

Identification of compounds to modulate NADPH metabolism is crucial for understanding complex diseases and developing effective therapies. However, the complex nature of NADPH metabolism poses challenges in achieving this goal. In this study, we proposed a novel strategy named NADPHnet to predict key proteins and drug-target interactions related to NADPH metabolism via network-based methods. Different from traditional approaches only focusing on one single protein, NADPHnet could screen compounds to modulate NADPH metabolism from a comprehensive view. Specifically, NADPHnet identified key proteins involved in regulation of NADPH metabolism using network-based methods, and characterized the impact of natural products on NADPH metabolism using a combined score, NADPH-Score. NADPHnet demonstrated a broader applicability domain and improved accuracy in the external validation set. This approach was further employed along with molecular docking to identify 27 compounds from a natural product library, 6 of which exhibited concentration-dependent changes of cellular NADPH level within 100 µM, with Oxyberberine showing promising effects even at 10 µM. Mechanistic and pathological analyses of Oxyberberine suggest potential novel mechanisms to affect diabetes and cancer. Overall, NADPHnet offers a promising method for prediction of NADPH metabolism modulation and advances drug discovery for complex diseases.

2.
Comput Biol Med ; 168: 107831, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38081118

RESUMO

Identification of adverse drug events (ADEs) is crucial to reduce human health risks and accelerate drug safety assessment. ADEs are mainly caused by unintended interactions with primary or additional targets (off-targets). In this study, we proposed a novel interpretable method named mtADENet, which integrates multiple types of network-based inference approaches for ADE prediction. Different from phenotype-based methods, mtADENet introduced computational target profiles predicted by network-based methods to bridge the gap between chemical structures and ADEs, and hence can not only predict ADEs for drugs and novel compounds within or outside the drug-ADE association network, but also provide insights for the elucidation of molecular mechanisms of the ADEs caused by drugs. We constructed a series of network-based prediction models for 23 ADE categories. These models achieved high AUC values ranging from 0.865 to 0.942 in 10-fold cross validation. The best model further showed high performance on four external validation sets, which outperformed two previous network-based methods. To show the practical value of mtADENet, we performed case studies on developmental neurotoxicity and cardio-oncology, and over 50 % of predicted ADEs and targets for drugs and novel compounds were validated by literature. Moreover, mtADENet is freely available at our web server named NetInfer (http://lmmd.ecust.edu.cn/netinfer/). In summary, mtADENet would be a powerful tool for ADE prediction and drug safety assessment in drug discovery and development.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Descoberta de Drogas
3.
J Alzheimers Dis ; 2023 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-36744334

RESUMO

BACKGROUND: The oxidative stress hypothesis is challenging the dominant position of amyloid-ß (Aß) in the field of understanding the mechanisms of Alzheimer's disease (AD), a complicated and untreatable neurodegenerative disease. OBJECTIVE: The goal of the present study was to uncover the oxidative stress mechanisms causing AD, as well as the potential therapeutic targets and neuroprotective drugs against oxidative stress mechanisms. METHODS: In this study, a systematic workflow combining pharmacological experiments and computational prediction were proposed. 222 drugs and natural products were collected first and then tested on SH-SY5Y cells to obtain phenotypic screening data on neuroprotection. The preliminary screening data were integrated with drug-target interactions (DTIs) and multi-scale biomedical data, which were analyzed with statistical tests and gene set enrichment analysis. A polypharmacology network was further constructed for investigation. RESULTS: 340 DTIs were matched in multiple databases, and 222 cell viability ratios were calculated for experimental compounds. We identified significant potential therapeutic targets based on oxidative stress mechanisms for AD, including NR3C1, SHBG, ESR1, PGR, and AVPR1A, which might be closely related to neuroprotective effects and pathogenesis. 50% of the top 14 enriched pathways were found to correlate with AD, such as arachidonic acid metabolism and neuroactive ligand-receptor interaction. Several approved drugs in this research were also found to exert neuroprotective effects against oxidative stress mechanisms, including beclometasone, methylprednisolone, and conivaptan. CONCLUSION: Our results indicated that NR3C1, SHBG, ESR1, PGR, and AVPR1A were promising therapeutic targets and several drugs may be repurposed from the perspective of oxidative stress and AD.

4.
Brief Bioinform ; 23(5)2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35998896

RESUMO

Nuclear receptors (NRs) are ligand-activated transcription factors, which constitute one of the most important targets for drug discovery. Current computational strategies mainly focus on a single target, and the transfer of learned knowledge among NRs was not considered yet. Herein we proposed a novel computational framework named NR-Profiler for prediction of potential NR modulators with high affinity and specificity. First, we built a comprehensive NR data set including 42 684 interactions to connect 42 NRs and 31 033 compounds. Then, we used multi-task deep neural network and multi-task graph convolutional neural network architectures to construct multi-task multi-classification models. To improve the predictive capability and robustness, we built a consensus model with an area under the receiver operating characteristic curve (AUC) = 0.883. Compared with conventional machine learning and structure-based approaches, the consensus model showed better performance in external validation. Using this consensus model, we demonstrated the practical value of NR-Profiler in virtual screening for NRs. In addition, we designed a selectivity score to quantitatively measure the specificity of NR modulators. Finally, we developed a freely available standalone software for users to make profiling predictions for their compounds of interest. In summary, our NR-Profiler provides a useful tool for NR-profiling prediction and is expected to facilitate NR-based drug discovery.


Assuntos
Aprendizado Profundo , Receptores Artificiais , Receptores dos Hormônios Gastrointestinais , Receptores de Imunoglobulina Polimérica , Receptor do Fator Ativador de Células B , Proteína Semelhante a Receptor de Calcitonina , Receptor gp130 de Citocina , Antagonistas dos Receptores H2 da Histamina , Ligantes , Antagonistas dos Receptores de Neurocinina-1 , Proteínas Proto-Oncogênicas c-met , Receptor de Glutamato Metabotrópico 5 , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores , Receptores de Hidrocarboneto Arílico , Receptores de Calcitriol , Receptores Citoplasmáticos e Nucleares , Receptores Muscarínicos
5.
J Chem Inf Model ; 62(3): 486-497, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35041411

RESUMO

Estrogen-related receptor α (ERRα), a member of nuclear receptors (NRs), plays a role in the regulation of cellular energy metabolism and is reported to be a novel potential target for type 2 diabetes therapy. To date, only a few agonists of ERRα have been identified to improve insulin sensitivity and decrease blood glucose levels. Herein, the discovery of novel potent agonists of ERRα determined using a combined virtual screening approach is described. Molecular dynamics (MD) simulations were used to obtain structural ensembles that can consider receptor flexibility. Then, an efficient virtual screening strategy with a combination of similarity search and ensemble docking was performed against the Enamine, SPECS, and Drugbank databases to identify potent ERRα agonists. Finally, a total of 66 compounds were purchased for experimental testing. Biological investigation of promising candidates identified seven compounds that have activity against ERRα with EC50 values ranging from 1.11 to 21.70 µM, with novel scaffolds different from known ERRα agonists until now. Additionally, the molecule GX66 showed micromolar inverse activity against ERRα with an IC50 of 0.82 µM. The predicted binding modes showed that these compounds were anchored in ERRα-LBP via interactions with several residues of ERRα. Overall, this study not only identified the novel potent ERRα agonists or an inverse agonist that would be the promising starting point for further exploration but also demonstrated a successful molecular dynamics-guided approach applicable in virtual screening for ERRα agonists.


Assuntos
Diabetes Mellitus Tipo 2 , Humanos , Simulação de Dinâmica Molecular , Receptores de Estrogênio/metabolismo , Receptor ERRalfa Relacionado ao Estrogênio
6.
J Chem Inf Model ; 61(5): 2486-2498, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33955748

RESUMO

NAD(P)H:quinone oxidoreductase 1 (NQO1) has been shown to be a potential therapeutic target for various human diseases, such as cancer and neurodegenerative disorders. Recent advances in computational methods, especially network-based methods, have made it possible to identify novel compounds for a target with high efficiency and low cost. In this study, we designed a workflow combining network-based methods and identification of privileged substructures to discover new compounds targeting NQO1 from a natural product library. According to the prediction results, we purchased 56 compounds for experimental validation. Without the assistance of privileged substructures, 31 compounds (31/56 = 55.4%) showed IC50 < 100 µM, and 11 compounds (11/56 = 19.6%) showed IC50 < 10 µM. With the assistance of privileged substructures, the two success rates were increased to 61.8 and 26.5%, respectively. Seven natural products further showed inhibitory activity on NQO1 at the cellular level, which may serve as lead compounds for further development. Moreover, network analysis revealed that osthole may exert anticancer effects against multiple cancer types by inhibiting not only carbonic anhydrases IX and XII but also NQO1. Our workflow and computational methods can be easily applied in other targets and become useful tools in drug discovery and development.


Assuntos
Produtos Biológicos , Neoplasias , Produtos Biológicos/farmacologia , Descoberta de Drogas , Humanos , NAD(P)H Desidrogenase (Quinona) , Neoplasias/tratamento farmacológico , Quinonas
7.
J Chem Inf Model ; 61(5): 2475-2485, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33900090

RESUMO

Identification of drug-pathway associations plays an important role in pathway-based drug repurposing. However, it is time-consuming and costly to uncover new drug-pathway associations experimentally. The drug-induced transcriptomics data provide a global view of cellular pathways and tell how these pathways change under different treatments. These data enable computational approaches for large-scale prediction of drug-pathway associations. Here we introduced DPNetinfer, a novel computational method to predict potential drug-pathway associations based on substructure-drug-pathway networks via network-based approaches. The results demonstrated that DPNetinfer performed well in a pan-cancer network with an AUC (area under curve) = 0.9358. Meanwhile, DPNetinfer was shown to have a good capability of generalization on two external validation sets (AUC = 0.8519 and 0.7494, respectively). As a case study, DPNetinfer was used in pathway-based drug repurposing for cancer therapy. Unexpected anticancer activities of some nononcology drugs were then identified on the PI3K-Akt pathway. Considering tumor heterogeneity, seven primary site-based models were constructed by DPNetinfer in different drug-pathway networks. In a word, DPNetinfer provides a powerful tool for large-scale prediction of drug-pathway associations in pathway-based drug repurposing. A web tool for DPNetinfer is freely available at http://lmmd.ecust.edu.cn/netinfer/.


Assuntos
Neoplasias , Preparações Farmacêuticas , Biologia Computacional , Reposicionamento de Medicamentos , Humanos , Fosfatidilinositol 3-Quinases
8.
BMC Complement Med Ther ; 20(1): 322, 2020 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-33109189

RESUMO

BACKGROUND: Arnebia euchroma (A. euchroma) is a traditional Chinese medicine (TCM) used for the treatment of blood diseases including leukemia. In recent years, many studies have been conducted on the anti-tumor effect of shikonin and its derivatives, the major active components of A. euchroma. However, the underlying mechanism of action (MoA) for all the components of A. euchroma on leukemia has not been explored systematically. METHODS: In this study, we analyzed the MoA of A. euchroma on leukemia via network pharmacology approach. Firstly, the chemical components and their concentrations in A. euchroma as well as leukemia-related targets were collected. Next, we predicted compound-target interactions (CTIs) with our balanced substructure-drug-target network-based inference (bSDTNBI) method. The known and predicted targets of A. euchroma and leukemia-related targets were merged together to construct A. euchroma-leukemia protein-protein interactions (PPIs) network. Then, weighted compound-target bipartite network was constructed according to combination of eight central attributes with concentration information through Cytoscape. Additionally, molecular docking simulation was performed to calculate whether the components and predicted targets have interactions or not. RESULTS: A total of 65 components of A. euchroma were obtained and 27 of them with concentration information, which were involved in 157 targets and 779 compound-target interactions (CTIs). Following the calculation of eight central attributes of targets in A. euchroma-leukemia PPI network, 37 targets with all central attributes greater than the median values were selected to construct the weighted compound-target bipartite network and do the KEGG pathway analysis. We found that A. euchroma candidate targets were significantly associated with several apoptosis and inflammation-related biological pathways, such as MAPK signaling, PI3K-Akt signaling, IL-17 signaling, and T cell receptor signaling pathways. Moreover, molecular docking simulation demonstrated that there were eight pairs of predicted CTIs had the strong binding free energy. CONCLUSIONS: This study deciphered that the efficacy of A. euchroma in the treatment of leukemia might be attributed to 10 targets and 14 components, which were associated with inhibiting leukemia cell survival and inducing apoptosis, relieving inflammatory environment and inhibiting angiogenesis.


Assuntos
Boraginaceae/química , Leucemia/tratamento farmacológico , Medicina Tradicional Chinesa , Simulação de Acoplamento Molecular , Mapas de Interação de Proteínas , Humanos , Estrutura Molecular
9.
J Med Chem ; 63(3): 1051-1067, 2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-31910018

RESUMO

Our previous study had identified ciclopirox (CPX) as a promising lead compound for treatment of ischemic stroke. To find better neuroprotective agents, a series of N-hydroxypyridone derivatives based on CPX were designed, synthesized, and evaluated in this study. Among these derivatives, compound 11 exhibits significant neuroprotection against oxygen glucose deprivation and oxidative stress-induced injuries in neuronal cells. Moreover, compound 11 possesses good blood-brain barrier permeability and superior antioxidant capability. In addition, a complex of compound 11 with olamine-11·Ola possesses good water solubility, negligible hERG inhibition, and superior metabolic stability. The in vivo experiment demonstrates that 11·Ola significantly reduces brain infarction and alleviates neurological deficits in middle cerebral artery occlusion rats. Hence, compound 11·Ola is identified in our research as a prospective prototype in the innovation of stroke treatment.


Assuntos
Ciclopirox/análogos & derivados , Ciclopirox/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Antioxidantes/síntese química , Antioxidantes/uso terapêutico , Antioxidantes/toxicidade , Apoptose/efeitos dos fármacos , Encéfalo/patologia , Linhagem Celular Tumoral , Ciclopirox/toxicidade , Desenho de Fármacos , Humanos , Infarto da Artéria Cerebral Média/patologia , Masculino , Estrutura Molecular , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/toxicidade , Ratos Sprague-Dawley , Relação Estrutura-Atividade
10.
J Biomol Struct Dyn ; 38(13): 3867-3878, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31498028

RESUMO

Estrogen-related receptor alpha (ERRα), a member of nuclear receptors (NRs), participates in energy metabolism. Recent experiments identified that several agonists to increase the activity of ERRα, which have a therapeutic effect in improving insulin sensitivity and lowering blood glucose levels. However, the detailed molecular mechanism about how the ligands affect the structure of ERRα remains elusive. To better understand the conformational change of ERRα complexed with agonists and inverse agonists, unbiased molecular dynamics (MD) simulations were performed on the ligand binding domain of ERRα (ERRα-LBD) bound with different ligands. According to the results, the ERRα-agonist interactions were more stable in the presence of the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). More importantly, we observed that the binding of inverse agonists would decrease the stability of helix 12 (H12) of ERRα. Moreover, we suggested that Phe232 and Phe414 should be key residues in the interaction pathway from ligands to H12, which provided a possible explanation about how ligands impact the structure of ERRα. These results would provide insights into the design of novel and efficient agonists of ERRα to treat metabolic diabetes.Communicated by Ramaswamy H. Sarma.


Assuntos
Simulação de Dinâmica Molecular , Receptores de Estrogênio , Metabolismo Energético , Ligantes , Receptor ERRalfa Relacionado ao Estrogênio
11.
Pharmacol Res ; 129: 400-413, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29133212

RESUMO

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 approved 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 approved 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 IC50=2.67µM and 6.34µM, respectively. In summary, this study offers powerful network-based tools for identifying polypharmacology of GPCR ligands in drug discovery and development.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Simulação por Computador , Descoberta de Drogas , Humanos , Ligantes , Polifarmacologia
12.
J Chem Inf Model ; 57(11): 2657-2671, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-28956927

RESUMO

Natural products with diverse chemical scaffolds have been recognized as an invaluable source of compounds in drug discovery and development. However, systematic identification of drug targets for natural products at the human proteome level via various experimental assays is highly expensive and time-consuming. In this study, we proposed a systems pharmacology infrastructure to predict new drug targets and anticancer indications of natural products. Specifically, we reconstructed a global drug-target network with 7,314 interactions connecting 751 targets and 2,388 natural products and built predictive network models via a balanced substructure-drug-target network-based inference approach. A high area under receiver operating characteristic curve of 0.96 was yielded for predicting new targets of natural products during cross-validation. The newly predicted targets of natural products (e.g., resveratrol, genistein, and kaempferol) with high scores were validated by various literature studies. We further built the statistical network models for identification of new anticancer indications of natural products through integration of both experimentally validated and computationally predicted drug-target interactions of natural products with known cancer proteins. We showed that the significantly predicted anticancer indications of multiple natural products (e.g., naringenin, disulfiram, and metformin) with new mechanism-of-action were validated by various published experimental evidence. In summary, this study offers powerful computational systems pharmacology approaches and tools for the development of novel targeted cancer therapies by exploiting the polypharmacology of natural products.


Assuntos
Produtos Biológicos/metabolismo , Produtos Biológicos/farmacologia , Biologia Computacional/métodos , Simulação por Computador , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Produtos Biológicos/uso terapêutico , Neoplasias/metabolismo
13.
J Med Chem ; 60(5): 1817-1828, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28218840

RESUMO

Acute lung injury (ALI) and idiopathic pulmonary fibrosis (IPF) are both serious public health problems with high incidence and mortality rate in adults, and with few drugs available for the efficient treatment in clinic. In this study, we identified that two known histone deacetylase (HDAC) inhibitors, suberanilohydroxamic acid (SAHA, 1) and its analogue 4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]benzamide (2), are effective inhibitors of Leukotriene A4 hydrolase (LTA4H), a key enzyme in the biosynthesis of leukotriene B4 (LTB4), across a panel of 18 HDAC inhibitors, using enzymatic assay, thermofluor assay, and X-ray crystallographic investigation. Importantly, both 1 and 2 markedly diminish early neutrophilic inflammation in mouse models of ALI and IPF under a clinical safety dose. Detailed mechanisms of down-regulation of proinflammatory cytokines by 1 or 2 were determined in vivo. Collectively, 1 and 2 would provide promising agents with well-known clinical safety for potential treatment in patients with ALI and IPF via pharmacologically inhibiting LAT4H and blocking LTB4 biosynthesis.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Epóxido Hidrolases/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Fibrose Pulmonar Idiopática/prevenção & controle , Leucotrieno B4/antagonistas & inibidores , Neutrófilos/efeitos dos fármacos , Lesão Pulmonar Aguda/patologia , Animais , Feminino , Fibrose Pulmonar Idiopática/patologia , Leucotrieno B4/biossíntese , Camundongos , Camundongos Endogâmicos C57BL
14.
Brief Bioinform ; 18(2): 333-347, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-26944082

RESUMO

Computational prediction of drug-target interactions (DTIs) and drug repositioning provides a low-cost and high-efficiency approach for drug discovery and development. The traditional social network-derived methods based on the naïve DTI topology information cannot predict potential targets for new chemical entities or failed drugs in clinical trials. There are currently millions of commercially available molecules with biologically relevant representations in chemical databases. It is urgent to develop novel computational approaches to predict targets for new chemical entities and failed drugs on a large scale. In this study, we developed a useful tool, namely substructure-drug-target network-based inference (SDTNBI), to prioritize potential targets for old drugs, failed drugs and new chemical entities. SDTNBI incorporates network and chemoinformatics to bridge the gap between new chemical entities and known DTI network. High performance was yielded in 10-fold and leave-one-out cross validations using four benchmark data sets, covering G protein-coupled receptors, kinases, ion channels and nuclear receptors. Furthermore, the highest areas under the receiver operating characteristic curve were 0.797 and 0.863 for two external validation sets, respectively. Finally, we identified thousands of new potential DTIs via implementing SDTNBI on a global network. As a proof-of-principle, we showcased the use of SDTNBI to identify novel anticancer indications for nonsteroidal anti-inflammatory drugs by inhibiting AKR1C3, CA9 or CA12. In summary, SDTNBI is a powerful network-based approach that predicts potential targets for new chemical entities on a large scale and will provide a new tool for DTI prediction and drug repositioning. The program and predicted DTIs are available on request.


Assuntos
Reposicionamento de Medicamentos , Simulação por Computador , Sistemas de Liberação de Medicamentos , Descoberta de Drogas , Curva ROC
15.
Br J Pharmacol ; 173(23): 3372-3385, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27646592

RESUMO

BACKGROUND AND PURPOSE: Deciphering chemical mechanism of action (MoA) enables the development of novel therapeutics (e.g. drug repositioning) and evaluation of drug side effects. Development of novel computational methods for chemical MoA assessment under a systems pharmacology framework would accelerate drug discovery and development with greater efficiency and low cost. EXPERIMENTAL APPROACH: In this study, we proposed an improved network-based inference method, balanced substructure-drug-target network-based inference (bSDTNBI), to predict MoA for old drugs, clinically failed drugs and new chemical entities. Specifically, three parameters were introduced into network-based resource diffusion processes to adjust the initial resource allocation of different node types, the weighted values of different edge types and the influence of hub nodes. The performance of the method was systematically validated by benchmark datasets and bioassays. KEY RESULTS: High performance was yielded for bSDTNBI in both 10-fold and leave-one-out cross validations. A global drug-target network was built to explore MoA of anticancer drugs and repurpose old drugs for 15 cancer types/subtypes. In a case study, 27 predicted candidates among 56 commercially available compounds were experimentally validated to have binding affinities on oestrogen receptor α with IC50 or EC50 values ≤10 µM. Furthermore, two dual ligands with both agonistic and antagonistic activities ≤1 µM would provide potential lead compounds for the development of novel targeted therapy in breast cancer or osteoporosis. CONCLUSION AND IMPLICATIONS: In summary, bSDTNBI would provide a powerful tool for the MoA assessment on both old drugs and novel compounds in drug discovery and development.


Assuntos
Antineoplásicos/farmacologia , Simulação por Computador , Desenho de Fármacos , Neoplasias/tratamento farmacológico , Antineoplásicos/administração & dosagem , Descoberta de Drogas/métodos , Reposicionamento de Medicamentos/métodos , Humanos , Concentração Inibidora 50 , Ligantes , Terapia de Alvo Molecular
16.
Oncotarget ; 7(29): 45584-45596, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27329603

RESUMO

As the recent development of high-throughput technologies in cancer pharmacogenomics, there is an urgent need to develop new computational approaches for comprehensive identification of new pharmacogenomic biomarkers, such as microRNAs (miRNAs). In this study, a network-based framework, namely the SMiR-NBI model, was developed to prioritize miRNAs as potential biomarkers characterizing treatment responses of anticancer drugs on the basis of a heterogeneous network connecting drugs, miRNAs and genes. A high area under the receiver operating characteristic curve of 0.820 ± 0.013 was yielded during 10-fold cross validation. In addition, high performance was further validated in identifying new anticancer mechanism-of-action for natural products and non-steroidal anti-inflammatory drugs. Finally, the newly predicted miRNAs for tamoxifen and metformin were experimentally validated in MCF-7 and MDA-MB-231 breast cancer cell lines via qRT-PCR assays. High success rates of 60% and 65% were yielded for tamoxifen and metformin, respectively. Specifically, 11 oncomiRNAs (e.g. miR-20a-5p, miR-27a-3p, miR-29a-3p, and miR-146a-5p) from the top 20 predicted miRNAs were experimentally verified as new pharmacogenomic biomarkers for metformin in MCF-7 or MDA-MB-231 cell lines. In summary, the SMiR-NBI model would provide a powerful tool to identify potential pharmacogenomic biomarkers characterized by miRNAs in the emerging field of precision cancer medicine, which is available at http://lmmd.ecust.edu.cn/database/smir-nbi/.


Assuntos
Biomarcadores Tumorais/genética , Ensaios de Seleção de Medicamentos Antitumorais/métodos , MicroRNAs/análise , Testes Farmacogenômicos/métodos , Área Sob a Curva , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Medicina de Precisão/métodos , Curva ROC
17.
Mol Inform ; 34(4): 228-35, 2015 04.
Artigo em Inglês | MEDLINE | ID: mdl-27490168

RESUMO

Carcinogenicity is one of the most concerned properties of chemicals to human health, thus it is important to identify chemical carcinogenicity as early as possible. In this study, 829 diverse compounds with rat carcinogenicity were collected from Carcinogenic Potency Database (CPDB). Using six types of fingerprints to represent the molecules, 30 binary and ternary classification models were generated to predict chemical carcinogenicity by five machine learning methods. The models were evaluated by an external validation set containing 87 chemicals from ISSCAN database. The best binary model was developed by MACCS keys and kNN algorithm with predictive accuracy at 83.91 %, while the best ternary model was also generated by MACCS keys and kNN algorithm with overall accuracy at 80.46 %. Furthermore, the best binary and ternary classification models were used to estimate carcinogenicity of tobacco smoke components containing 2251 compounds. 981 ones were predicted as carcinogens by binary classification model, while 110 compounds were predicted as strong carcinogens and 807 ones as weak carcinogens by ternary classification model. The results indicated that our models would be helpful for prediction of chemical carcinogenicity.


Assuntos
Carcinógenos/química , Carcinógenos/classificação , Simulação por Computador , Bases de Dados de Compostos Químicos , Modelos Químicos , Poluição por Fumaça de Tabaco , Humanos
18.
Sci Rep ; 4: 5576, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24992957

RESUMO

MicroRNAs (miRNAs) play important roles in multiple biological processes and have attracted much scientific attention recently. Their expression can be altered by environmental factors (EFs), which are associated with many diseases. Identification of the phenotype-genotype relationships among miRNAs, EFs, and diseases at the network level will help us to better understand toxicology mechanisms and disease etiologies. In this study, we developed a computational systems toxicology framework to predict new associations among EFs, miRNAs and diseases by integrating EF structure similarity and disease phenotypic similarity. Specifically, three comprehensive bipartite networks: EF-miRNA, EF-disease and miRNA-disease associations, were constructed to build predictive models. The areas under the receiver operating characteristic curves using 10-fold cross validation ranged from 0.686 to 0.910. Furthermore, we successfully inferred novel EF-miRNA-disease networks in two case studies for breast cancer and cigarette smoke. Collectively, our methods provide a reliable and useful tool for the study of chemical risk assessment and disease etiology involving miRNAs.


Assuntos
Neoplasias da Mama/genética , Exposição Ambiental , Neoplasias Pulmonares/etiologia , MicroRNAs/fisiologia , Poluição por Fumaça de Tabaco/efeitos adversos , Área Sob a Curva , Neoplasias da Mama/metabolismo , Simulação por Computador , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Interação Gene-Ambiente , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Interferência de RNA , Curva ROC , Fatores de Risco
19.
J Chem Inf Model ; 53(4): 753-62, 2013 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-23527559

RESUMO

Prediction of polypharmacological profiles of drugs enables us to investigate drug side effects and further find their new indications, i.e. drug repositioning, which could reduce the costs while increase the productivity of drug discovery. Here we describe a new computational framework to predict polypharmacological profiles of drugs by the integration of chemical, side effect, and therapeutic space. On the basis of our previous developed drug side effects database, named MetaADEDB, a drug side effect similarity inference (DSESI) method was developed for drug-target interaction (DTI) prediction on a known DTI network connecting 621 approved drugs and 893 target proteins. The area under the receiver operating characteristic curve was 0.882 ± 0.011 averaged from 100 simulated tests of 10-fold cross-validation for the DSESI method, which is comparative with drug structural similarity inference and drug therapeutic similarity inference methods. Seven new predicted candidate target proteins for seven approved drugs were confirmed by published experiments, with the successful hit rate more than 15.9%. Moreover, network visualization of drug-target interactions and off-target side effect associations provide new mechanism-of-action of three approved antipsychotic drugs in a case study. The results indicated that the proposed methods could be helpful for prediction of polypharmacological profiles of drugs.


Assuntos
Antipsicóticos/química , Reposicionamento de Medicamentos/estatística & dados numéricos , Modelos Estatísticos , Medicamentos sob Prescrição/química , Algoritmos , Antipsicóticos/efeitos adversos , Área Sob a Curva , Simulação por Computador , Mineração de Dados , Bases de Dados de Produtos Farmacêuticos , Humanos , Ligantes , Valor Preditivo dos Testes , Medicamentos sob Prescrição/efeitos adversos , Transtornos Psicóticos/tratamento farmacológico , Relação Quantitativa Estrutura-Atividade , Curva ROC
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA