Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Int J Mol Sci ; 25(6)2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38542239

RESUMO

Animal studies are typically utilized to understand the complex mechanisms associated with toxicant-induced hepatotoxicity. Among the alternative approaches to animal studies, in vitro pooled human hepatocytes have the potential to capture population variability. Here, we examined the effect of the hepatotoxicant thioacetamide on pooled human hepatocytes, divided into five lots, obtained from forty diverse donors. For 24 h, pooled human hepatocytes were exposed to vehicle, 1.33 mM (low dose), and 12 mM (high dose) thioacetamide, followed by RNA-seq analysis. We assessed gene expression variability using heat maps, correlation plots, and statistical variance. We used KEGG pathways and co-expression modules to identify underlying physiological processes/pathways. The co-expression module analysis showed that the majority of the lots exhibited activation for the bile duct proliferation module. Despite lot-to-lot variability, we identified a set of common differentially expressed genes across the lots with similarities in their response to amino acid, lipid, and carbohydrate metabolism. We also examined efflux transporters and found larger lot-to-lot variability in their expression patterns, indicating a potential for alteration in toxicant bioavailability within the cells, which could in turn affect the gene expression patterns between the lots. Overall, our analysis highlights the challenges in using pooled hepatocytes to understand mechanisms of toxicity.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Tioacetamida , Animais , Humanos , Tioacetamida/toxicidade , Fígado/metabolismo , Hepatócitos/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo
2.
Int J Mol Sci ; 24(8)2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-37108594

RESUMO

Acute kidney injury, which is associated with high levels of morbidity and mortality, affects a significant number of individuals, and can be triggered by multiple factors, such as medications, exposure to toxic chemicals or other substances, disease, and trauma. Because the kidney is a critical organ, understanding and identifying early cellular or gene-level changes can provide a foundation for designing medical interventions. In our earlier work, we identified gene modules anchored to histopathology phenotypes associated with toxicant-induced liver and kidney injuries. Here, using in vivo and in vitro experiments, we assessed and validated these kidney injury-associated modules by analyzing gene expression data from the kidneys of male Hartley guinea pigs exposed to mercuric chloride. Using plasma creatinine levels and cell-viability assays as measures of the extent of renal dysfunction under in vivo and in vitro conditions, we performed an initial range-finding study to identify the appropriate doses and exposure times associated with mild and severe kidney injuries. We then monitored changes in kidney gene expression at the selected doses and time points post-toxicant exposure to characterize the mechanisms of kidney injury. Our injury module-based analysis revealed a dose-dependent activation of several phenotypic cellular processes associated with dilatation, necrosis, and fibrogenesis that were common across the experimental platforms and indicative of processes that initiate kidney damage. Furthermore, a comparison of activated injury modules between guinea pigs and rats indicated a strong correlation between the modules, highlighting their potential for cross-species translational studies.


Assuntos
Injúria Renal Aguda , Cloreto de Mercúrio , Ratos , Masculino , Cobaias , Animais , Cloreto de Mercúrio/toxicidade , Rim/metabolismo , Testes de Função Renal , Injúria Renal Aguda/metabolismo , Fígado/metabolismo
3.
Toxicol Appl Pharmacol ; 430: 115713, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34492290

RESUMO

To study the complex processes involved in liver injuries, researchers rely on animal investigations, using chemically or surgically induced liver injuries, to extrapolate findings and infer human health risks. However, this presents obvious challenges in performing a detailed comparison and validation between the highly controlled animal models and development of liver injuries in humans. Furthermore, it is not clear whether there are species-dependent and -independent molecular initiating events or processes that cause liver injury before they eventually lead to end-stage liver disease. Here, we present a side-by-side study of rats and guinea pigs using thioacetamide to examine the similarities between early molecular initiating events during an acute-phase liver injury. We exposed Sprague Dawley rats and Hartley guinea pigs to a single dose of 25 or 100 mg/kg thioacetamide and collected blood plasma for metabolomic analysis and liver tissue for RNA-sequencing. The subsequent toxicogenomic analysis identified consistent liver injury trends in both genomic and metabolomic data within 24 and 33 h after thioacetamide exposure in rats and guinea pigs, respectively. In particular, we found species similarities in the key injury phenotypes of inflammation and fibrogenesis in our gene module analysis for liver injury phenotypes. We identified expression of several common genes (e.g., SPP1, TNSF18, SERPINE1, CLDN4, TIMP1, CD44, and LGALS3), activation of injury-specific KEGG pathways, and alteration of plasma metabolites involved in amino acid and bile acid metabolism as some of the key molecular processes that changed early upon thioacetamide exposure and could play a major role in the initiation of acute liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Perfilação da Expressão Gênica , Fígado/metabolismo , Metaboloma , Metabolômica , Tioacetamida , Transcriptoma , Animais , Biomarcadores/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Modelos Animais de Doenças , Redes Reguladoras de Genes , Cobaias , Fígado/patologia , Masculino , Ratos Sprague-Dawley , Especificidade da Espécie , Fatores de Tempo
4.
Toxicol Pathol ; 46(2): 202-223, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29378501

RESUMO

The past decade has seen an increase in the development and clinical use of biomarkers associated with histological features of liver disease. Here, we conduct a comparative histological and global proteomics analysis to identify coregulated modules of proteins in the progression of hepatic steatosis or fibrosis. We orally administered the reference chemicals bromobenzene (BB) or 4,4'-methylenedianiline (4,4'-MDA) to male Sprague-Dawley rats for either 1 single administration or 5 consecutive daily doses. Livers were preserved for histopathology and global proteomics assessment. Analysis of liver sections confirmed a dose- and time-dependent increase in frequency and severity of histopathological features indicative of lipid accumulation after BB or fibrosis after 4,4'-MDA. BB administration resulted in a dose-dependent increase in the frequency and severity of inflammation and vacuolation. 4,4'-MDA administration resulted in a dose-dependent increase in the frequency and severity of periportal collagen accumulation and inflammation. Pathway analysis identified a time-dependent enrichment of biological processes associated with steatogenic or fibrogenic initiating events, cellular functions, and toxicological states. Differentially expressed protein modules were consistent with the observed histology, placing physiologically linked protein networks into context of the disease process. This study demonstrates the potential for protein modules to provide mechanistic links between initiating events and histopathological outcomes.


Assuntos
Biomarcadores/análise , Fígado Gorduroso/metabolismo , Cirrose Hepática/metabolismo , Proteômica/métodos , Administração Oral , Compostos de Anilina/toxicidade , Animais , Bromobenzenos/toxicidade , Fígado Gorduroso/induzido quimicamente , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Masculino , Ratos , Ratos Sprague-Dawley
5.
J Chem Inf Model ; 57(9): 2194-2202, 2017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28796500

RESUMO

The quantitative structure-activity relationship (QSAR) approach has been used to model a wide range of chemical-induced biological responses. However, it had not been utilized to model chemical-induced genomewide gene expression changes until very recently, owing to the complexity of training and evaluating a very large number of models. To address this issue, we examined the performance of a variable nearest neighbor (v-NN) method that uses information on near neighbors conforming to the principle that similar structures have similar activities. Using a data set of gene expression signatures of 13 150 compounds derived from cell-based measurements in the NIH Library of Integrated Network-based Cellular Signatures program, we were able to make predictions for 62% of the compounds in a 10-fold cross validation test, with a correlation coefficient of 0.61 between the predicted and experimentally derived signatures-a reproducibility rivaling that of high-throughput gene expression measurements. To evaluate the utility of the predicted gene expression signatures, we compared the predicted and experimentally derived signatures in their ability to identify drugs known to cause specific liver, kidney, and heart injuries. Overall, the predicted and experimentally derived signatures had similar receiver operating characteristics, whose areas under the curve ranged from 0.71 to 0.77 and 0.70 to 0.73, respectively, across the three organ injury models. However, detailed analyses of enrichment curves indicate that signatures predicted from multiple near neighbors outperformed those derived from experiments, suggesting that averaging information from near neighbors may help improve the signal from gene expression measurements. Our results demonstrate that the v-NN method can serve as a practical approach for modeling large-scale, genomewide, chemical-induced, gene expression changes.


Assuntos
Biologia Computacional/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Aprendizado de Máquina , Relação Quantitativa Estrutura-Atividade
6.
BMC Genomics ; 17(1): 790, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27724849

RESUMO

BACKGROUND: Acute kidney injury (AKI) caused by drug and toxicant ingestion is a serious clinical condition associated with high mortality rates. We currently lack detailed knowledge of the underlying molecular mechanisms and biological networks associated with AKI. In this study, we carried out gene co-expression analyses using DrugMatrix-a large toxicogenomics database with gene expression data from rats exposed to diverse chemicals-and identified gene modules associated with kidney injury to probe the molecular-level details of this disease. RESULTS: We generated a comprehensive set of gene co-expression modules by using the Iterative Signature Algorithm and found distinct clusters of modules that shared genes and were associated with similar chemical exposure conditions. We identified two module clusters that showed specificity for kidney injury in that they 1) were activated by chemical exposures causing kidney injury, 2) were not activated by other chemical exposures, and 3) contained known AKI-relevant genes such as Havcr1, Clu, and Tff3. We used the genes in these AKI-relevant module clusters to develop a signature of 30 genes that could assess the potential of a chemical to cause kidney injury well before injury actually occurs. We integrated AKI-relevant module cluster genes with protein-protein interaction networks and identified the involvement of immunoproteasomes in AKI. To identify biological networks and processes linked to Havcr1, we determined genes within the modules that frequently co-express with Havcr1, including Cd44, Plk2, Mdm2, Hnmt, Macrod1, and Gtpbp4. We verified this procedure by showing that randomized data did not identify Havcr1 co-expression genes and that excluding up to 10 % of the data caused only minimal degradation of the gene set. Finally, by using an external dataset from a rat kidney ischemic study, we showed that the frequently co-expressed genes of Havcr1 behaved similarly in a model of non-chemically induced kidney injury. CONCLUSIONS: Our study demonstrated that co-expression modules and co-expressed genes contain rich information for generating novel biomarker hypotheses and constructing mechanism-based molecular networks associated with kidney injury.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Mineração de Dados , Perfilação da Expressão Gênica , Toxicogenética , Transcriptoma , Injúria Renal Aguda/metabolismo , Animais , Biomarcadores , Análise por Conglomerados , Biologia Computacional/métodos , Bases de Dados Genéticas , Fenótipo , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Curva ROC , Ratos , Transdução de Sinais , Toxicogenética/métodos
7.
Chem Res Toxicol ; 29(10): 1729-1740, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27603675

RESUMO

The pregnane X receptor (PXR) is a ligand-activated transcription factor that acts as a master regulator of metabolizing enzymes and transporters. To avoid adverse drug-drug interactions and diseases such as steatosis and cancers associated with PXR activation, identifying drugs and chemicals that activate PXR is of crucial importance. In this work, we developed ligand-based predictive computational models for both rat and human PXR activation, which allowed us to identify potentially harmful chemicals and evaluate species-specific effects of a given compound. We utilized a large publicly available data set of nearly 2000 compounds screened in cell-based reporter gene assays to develop Bayesian quantitative structure-activity relationship models using physicochemical properties and structural descriptors. Our analysis showed that PXR activators tend to be hydrophobic and significantly different from nonactivators in terms of their physicochemical properties such as molecular weight, logP, number of rings, and solubility. Our Bayesian models, evaluated by using 5-fold cross-validation, displayed a sensitivity of 75% (76%), specificity of 76% (75%), and accuracy of 89% (89%) for human (rat) PXR activation. We identified structural features shared by rat and human PXR activators as well as those unique to each species. We compared rat in vitro PXR activation data to in vivo data by using DrugMatrix, a large toxicogenomics database with gene expression data obtained from rats after exposure to diverse chemicals. Although in vivo gene expression data pointed to cross-talk between nuclear receptor activators that is captured only by in vivo assays, overall we found broad agreement between in vitro and in vivo PXR activation. Thus, the models developed here serve primarily as efficient initial high-throughput in silico screens of in vitro activity.


Assuntos
Teorema de Bayes , Receptores de Esteroides/metabolismo , Animais , Humanos , Ligantes , Modelos Moleculares , Receptor de Pregnano X , Ratos , Ratos Sprague-Dawley
8.
J Appl Toxicol ; 36(9): 1137-49, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26725466

RESUMO

Organ injuries caused by environmental chemical exposures or use of pharmaceutical drugs pose a serious health risk that may be difficult to assess because of a lack of non-invasive diagnostic tests. Mapping chemical injuries to organ-specific histopathology outcomes via biomarkers will provide a foundation for designing precise and robust diagnostic tests. We identified co-expressed genes (modules) specific to injury endpoints using the Open Toxicogenomics Project-Genomics Assisted Toxicity Evaluation System (TG-GATEs) - a toxicogenomics database containing organ-specific gene expression data matched to dose- and time-dependent chemical exposures and adverse histopathology assessments in Sprague-Dawley rats. We proposed a protocol for selecting gene modules associated with chemical-induced injuries that classify 11 liver and eight kidney histopathology endpoints based on dose-dependent activation of the identified modules. We showed that the activation of the modules for a particular chemical exposure condition, i.e., chemical-time-dose combination, correlated with the severity of histopathological damage in a dose-dependent manner. Furthermore, the modules could distinguish different types of injuries caused by chemical exposures as well as determine whether the injury module activation was specific to the tissue of origin (liver and kidney). The generated modules provide a link between toxic chemical exposures, different molecular initiating events among underlying molecular pathways and resultant organ damage. Published 2016. This article is a U.S. Government work and is in the public domain in the USA. Journal of Applied Toxicology published by John Wiley & Sons, Ltd.


Assuntos
Redes Reguladoras de Genes , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Xenobióticos/toxicidade , Animais , Biomarcadores/metabolismo , Bases de Dados Factuais , Relação Dose-Resposta a Droga , Determinação de Ponto Final , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genômica , Rim/metabolismo , Fígado/metabolismo , Masculino , Modelos Biológicos , Análise de Componente Principal , Ratos , Ratos Sprague-Dawley , Toxicogenética
9.
Bioorg Med Chem ; 22(7): 2149-56, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24631364

RESUMO

Molecular dynamics (MD) simulations and hybrid quantum mechanical/molecular mechanical (QM/MM) calculations have been performed to explore the dynamic behaviors of cytochrome P450 2A6 (CYP2A6) binding with nicotine analogs (that are typical inhibitors) and to calculate their binding free energies in combination with Poisson-Boltzmann surface area (PBSA) calculations. The combined MD simulations and QM/MM-PBSA calculations reveal that the most important structural parameters affecting the CYP2A6-inhibitor binding affinity are two crucial internuclear distances, that is, the distance between the heme iron atom of CYP2A6 and the coordinating atom of the inhibitor, and the hydrogen-bonding distance between the N297 side chain of CYP2A6 and the pyridine nitrogen of the inhibitor. The combined MD simulations and QM/MM-PBSA calculations have led to dynamic CYP2A6-inhibitor binding structures that are consistent with the observed dynamic behaviors and structural features of CYP2A6-inhibitor binding, and led to the binding free energies that are in good agreement with the experimentally-derived binding free energies. The agreement between the calculated binding free energies and the experimentally-derived binding free energies suggests that the combined MD and QM/MM-PBSA approach may be used as a valuable tool to accurately predict the CYP2A6-inhibitor binding affinities in future computational design of new, potent and selective CYP2A6 inhibitors.


Assuntos
Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Simulação de Dinâmica Molecular , Nicotina/farmacologia , Teoria Quântica , Hidrocarboneto de Aril Hidroxilases/metabolismo , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Modelos Moleculares , Estrutura Molecular , Nicotina/análogos & derivados , Nicotina/química , Relação Estrutura-Atividade
10.
Bioorg Med Chem ; 22(3): 1148-55, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24405813

RESUMO

Natural products represent the fourth generation of multidrug resistance (MDR) reversal agents that resensitize MDR cancer cells overexpressing P-glycoprotein (Pgp) to cytotoxic agents. We have developed an effective synthetic route to prepare various Strychnos alkaloids and their derivatives. Molecular modeling of these alkaloids docked to a homology model of Pgp was employed to optimize ligand-protein interactions and design analogues with increased affinity to Pgp. Moreover, the compounds were evaluated for their (1) binding affinity to Pgp by fluorescence quenching, and (2) MDR reversal activity using a panel of in vitro and cell-based assays and compared to verapamil, a known inhibitor of Pgp activity. Compound 7 revealed the highest affinity to Pgp of all Strychnos congeners (Kd=4.4µM), the strongest inhibition of Pgp ATPase activity, and the strongest MDR reversal effect in two Pgp-expressing cell lines. Altogether, our findings suggest the clinical potential of these synthesized compounds as viable Pgp modulators justifies further investigation.


Assuntos
Alcaloides/química , Alcaloides/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Strychnos/química , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/química , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Adenosina Trifosfatases/metabolismo , Alcaloides/síntese química , Antineoplásicos Fitogênicos/síntese química , Antineoplásicos Fitogênicos/química , Linhagem Celular Tumoral/efeitos dos fármacos , Técnicas de Química Sintética , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Compostos Heterocíclicos de 4 ou mais Anéis/síntese química , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Alcaloides Indólicos/síntese química , Alcaloides Indólicos/química , Alcaloides Indólicos/farmacologia , Indóis/síntese química , Indóis/química , Indóis/farmacologia , Simulação de Acoplamento Molecular , Conformação Proteica , Tubocurarina/análogos & derivados , Tubocurarina/síntese química , Tubocurarina/química , Tubocurarina/farmacologia , Verapamil/farmacologia
11.
Front Toxicol ; 6: 1390196, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903859

RESUMO

Toxicants with the potential to bioaccumulate in humans and animals have long been a cause for concern, particularly due to their association with multiple diseases and organ injuries. Per- and polyfluoro alkyl substances (PFAS) and polycyclic aromatic hydrocarbons (PAH) are two such classes of chemicals that bioaccumulate and have been associated with steatosis in the liver. Although PFAS and PAH are classified as chemicals of concern, their molecular mechanisms of toxicity remain to be explored in detail. In this study, we aimed to identify potential mechanisms by which an acute exposure to PFAS and PAH chemicals can induce lipid accumulation and whether the responses depend on chemical class, dose, and sex. To this end, we analyzed mechanisms beginning with the binding of the chemical to a molecular initiating event (MIE) and the consequent transcriptomic alterations. We collated potential MIEs using predictions from our previously developed ToxProfiler tool and from published steatosis adverse outcome pathways. Most of the MIEs are transcription factors, and we collected their target genes by mining the TRRUST database. To analyze the effects of PFAS and PAH on the steatosis mechanisms, we performed a computational MIE-target gene analysis on high-throughput transcriptomic measurements of liver tissue from male and female rats exposed to either a PFAS or PAH. The results showed peroxisome proliferator-activated receptor (PPAR)-α targets to be the most dysregulated, with most of the genes being upregulated. Furthermore, PFAS exposure disrupted several lipid metabolism genes, including upregulation of fatty acid oxidation genes (Acadm, Acox1, Cpt2, Cyp4a1-3) and downregulation of lipid transport genes (Apoa1, Apoa5, Pltp). We also identified multiple genes with sex-specific behavior. Notably, the rate-limiting genes of gluconeogenesis (Pck1) and bile acid synthesis (Cyp7a1) were specifically downregulated in male rats compared to female rats, while the rate-limiting gene of lipid synthesis (Scd) showed a PFAS-specific upregulation. The results suggest that the PPAR signaling pathway plays a major role in PFAS-induced lipid accumulation in rats. Together, these results show that PFAS exposure induces a sex-specific multi-factorial mechanism involving rate-limiting genes of gluconeogenesis and bile acid synthesis that could lead to activation of an adverse outcome pathway for steatosis.

12.
ACS Omega ; 8(24): 21853-21861, 2023 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-37360478

RESUMO

The bile salt export pump (BSEP) is a key transporter involved in the efflux of bile salts from hepatocytes to bile canaliculi. Inhibition of BSEP leads to the accumulation of bile salts within the hepatocytes, leading to possible cholestasis and drug-induced liver injury. Screening for and identification of chemicals that inhibit this transporter aid in understanding the safety liabilities of these chemicals. Moreover, computational approaches to identify BSEP inhibitors provide an alternative to the more resource-intensive, gold standard experimental approaches. Here, we used publicly available data to develop predictive machine learning models for the identification of potential BSEP inhibitors. Specifically, we analyzed the utility of a graph convolutional neural network (GCNN)-based approach in combination with multitask learning to identify BSEP inhibitors. Our analyses showed that the developed GCNN model performed better than the variable-nearest neighbor and Bayesian machine learning approaches, with a cross-validation receiver operating characteristic area under the curve of 0.86. In addition, we compared GCNN-based single-task and multitask models and evaluated their utility in addressing data limitation challenges commonly observed in bioactivity modeling. We found that multitask models performed better than single-task models and can be utilized to identify active molecules for targets with limited data availability. Overall, our developed multitask GCNN-based BSEP model provides a useful tool for prioritizing hits during early drug discovery and in risk assessment of chemicals.

13.
Bioorg Med Chem Lett ; 22(4): 1629-32, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22266037

RESUMO

3-Phosphoinositide-dependent protein kinase-1 (PDK1) has been recognized as a promising anticancer target. Thus, it is interesting to identify new inhibitors of PDK1 for anticancer drug discovery. Through a combined use of virtual screening and wet experimental activity assays, we have identified a new PDK1 inhibitor with IC(50)=~200 nM. The anticancer activities of this compound have been confirmed by the anticancer activity assays using 60 cancer cell lines. The obtained new PDK1 inhibitor and its PDK1-inhibitor binding mode should be valuable in future de novo design of novel, more potent and selective PDK1 inhibitors for future development of anticancer therapeutics.


Assuntos
Antineoplásicos/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Antineoplásicos/química , Domínio Catalítico , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Estrutura Molecular
14.
J Chem Inf Model ; 52(10): 2559-69, 2012 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-23013546

RESUMO

As novel and drug-resistant bacterial strains continue to present an emerging health threat, the development of new antibacterial agents is critical. This includes making improvements to existing antibacterial scaffolds as well as identifying novel ones. The aim of this study is to apply a Bayesian classification QSAR approach to rapidly screen chemical libraries for compounds predicted to have antibacterial activity. Toward this end we assembled a data set of 317 known antibacterial compounds as well as a second data set of diverse, well-validated, non-antibacterial compounds from 215 PubChem Bioassays against various bacterial species. We constructed a Bayesian classification model using structural fingerprints and physicochemical property descriptors and achieved an accuracy of 84% and precision of 86% on an independent test set in identifying antibacterial compounds. To demonstrate the practical applicability of the model in virtual screening, we screened an independent data set of ~200k compounds. The results show that the model can screen top hits of PubChem Bioassay actives with accuracy up to ~76%, representing a 1.5-2-fold enrichment. The top screened hits represented a mixture of both known antibacterial scaffolds as well as novel scaffolds. Our study suggests that a well-validated Bayesian classification QSAR approach could compliment other screening approaches in identifying novel and promising hits. The data sets used in constructing and validating this model have been made publicly available.


Assuntos
Antibacterianos/química , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas/química , Interface Usuário-Computador , Antibacterianos/farmacologia , Teorema de Bayes , Simulação por Computador , Bases de Dados de Compostos Químicos , Desenho de Fármacos , Descoberta de Drogas , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Modelos Químicos , Estrutura Molecular , Curva ROC , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas/farmacologia
15.
J Chem Inf Model ; 52(2): 492-505, 2012 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-22196353

RESUMO

Polypharmacology has emerged as a new theme in drug discovery. In this paper, we studied polypharmacology using a ligand-based target fishing (LBTF) protocol. To implement the protocol, we first generated a chemogenomic database that links individual protein targets with a specified set of drugs or target representatives. Target profiles were then generated for a given query molecule by computing maximal shape/chemistry overlap between the query molecule and the drug sets assigned to each protein target. The overlap was computed using the program ROCS (Rapid Overlay of Chemical Structures). We validated this approach using the Directory of Useful Decoys (DUD). DUD contains 2950 active compounds, each with 36 property-matched decoys, against 40 protein targets. We chose a set of known drugs to represent each DUD target, and we carried out ligand-based virtual screens using data sets of DUD actives seeded into DUD decoys for each target. We computed Receiver Operator Characteristic (ROC) curves and associated area under the curve (AUC) values. For the majority of targets studied, the AUC values were significantly better than for the case of a random selection of compounds. In a second test, the method successfully identified off-targets for drugs such as rimantadine, propranolol, and domperidone that were consistent with those identified by recent experiments. The results from our ROCS-based target fishing approach are promising and have potential application in drug repurposing for single and multiple targets, identifying targets for orphan compounds, and adverse effect prediction.


Assuntos
Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Preparações Farmacêuticas/química , Curva ROC , Área Sob a Curva , Simulação por Computador , Ligantes , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas
16.
Toxicology ; 442: 152530, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32599119

RESUMO

Kidney injury caused by disease, trauma, environmental exposures, or drugs may result in decreased renal function, chronic kidney disease, or acute kidney failure. Diagnosis of kidney injury using serum creatinine levels, a common clinical test, only identifies renal dysfunction after the kidneys have undergone severe damage. Other indicators sensitive to kidney injury, such as the level of urine kidney injury molecule-1 (KIM-1), lack the ability to differentiate between injury phenotypes. To address early detection as well as detailed categorization of kidney-injury phenotypes in preclinical animal or cellular studies, we previously identified eight sets (modules) of co-expressed genes uniquely associated with kidney histopathology. Here, we used mercuric chloride (HgCl2)-a model nephrotoxicant-to chemically induce kidney injuries as monitored by KIM-1 levels in Sprague Dawley rats at two doses (0.25 or 0.50 mg/kg) and two exposure lengths (10 or 34 h). We collected whole transcriptome RNA-seq data derived from five animals at each dose and time point to perform a toxicogenomics analysis. Consistent with documented injury phenotypes for HgCl2 toxicity, our kidney-injury-module approach identified the onset of necrosis and dilation as early as 10 h after a dose of 0.50 mg/kg that produced only mild injury as judged by urinary KIM-1 excretion. The results of these animal studies highlight the potential of the kidney-injury-module approach to provide a sensitive and histopathology-specific readout of renal toxicity.


Assuntos
Nefropatias/induzido quimicamente , Nefropatias/patologia , Cloreto de Mercúrio/toxicidade , Toxicogenética/métodos , Animais , Aspartato Aminotransferases/sangue , Sequência de Bases , Biomarcadores/urina , Peso Corporal/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Moléculas de Adesão Celular/urina , Expressão Gênica/efeitos dos fármacos , Masculino , Necrose , Dobramento de Proteína/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
17.
Front Chem ; 7: 701, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31709231

RESUMO

High throughput screening (HTS) is an important component of lead discovery, with virtual screening playing an increasingly important role. Both methods typically suffer from lack of sensitivity and specificity against their true biological targets. With ever-increasing screening libraries and virtual compound collections, it is now feasible to conduct follow-up experimental testing on only a small fraction of hits. In this context, advances in virtual screening that achieve enrichment of true actives among top-ranked compounds ("early recognition") and, hence, reduce the number of hits to test, are highly desirable. The standard ligand-based virtual screening method for large compound libraries uses a molecular similarity search method that ranks the likelihood of a compound to be active against a drug target by its highest Tanimoto similarity to known active compounds. This approach assumes that the distributions of Tanimoto similarity values to all active compounds are identical (i.e., same mean and standard deviation)-an assumption shown to be invalid (Baldi and Nasr, 2010). Here, we introduce two methods that improve early recognition of actives by exploiting similarity information of all molecules. The first method ranks a compound by its highest z-score instead of its highest Tanimoto similarity, and the second by an aggregated score calculated from its Tanimoto similarity values to all known actives and inactives (or a large number of structurally diverse molecules when information on inactives is unavailable). Our evaluations, which use datasets of over 20 HTS campaigns downloaded from PubChem, indicate that compared to the conventional approach, both methods achieve a ~10% higher Boltzmann-enhanced discrimination of receiver operating characteristic (BEDROC) score-a metric of early recognition. Given the increasing use of virtual screening in early lead discovery, these methods provide straightforward means to enhance early recognition.

18.
Front Genet ; 10: 1007, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31681434

RESUMO

Exposure to chemicals contributes to the development and progression of fatty liver, or steatosis, a process characterized by abnormal accumulation of lipids within liver cells. However, lack of knowledge on how chemicals cause steatosis has prevented any large-scale assessment of the 80,000+ chemicals in current use. To address this gap, we mined a large, publicly available toxicogenomic dataset associated with 18 known steatogenic chemicals to assess responses across assays (in vitro and in vivo) and species (i.e., rats and humans). We identified genes that were differentially expressed (DEGs) in rat in vivo, rat in vitro, and human in vitro studies in which rats or in vitro primary cell lines were exposed to the chemicals at different doses and durations. Using these DEGs, we performed pathway enrichment analysis, analyzed the molecular initiating events (MIEs) of the steatosis adverse outcome pathway (AOP), and predicted metabolite changes using metabolic network analysis. Genes indicative of oxidative stress were among the DEGs most frequently observed in the rat in vivo studies. Nox4, a pro-fibrotic gene, was down-regulated across these chemical exposure conditions. We identified eight genes (Cyp1a1, Egr1, Ccnb1, Gdf15, Cdk1, Pdk4, Ccna2, and Ns5atp9) and one pathway (retinol metabolism), associated with steatogenic chemicals and whose response was conserved across the three in vitro and in vivo systems. Similarly, we found the predicted metabolite changes, such as increases of saturated and unsaturated fatty acids, conserved across the three systems. Analysis of the target genes associated with the MIEs of the current steatosis AOP did not provide a clear association between these 18 chemicals and the MIEs, underlining the multi-factorial nature of this disease. Notably, our overall analysis implicated mitochondrial toxicity as an important and overlooked MIE for chemical-induced steatosis. The integrated toxicogenomics approach to identify genes, pathways, and metabolites based on known steatogenic chemicals, provide an important mean to assess development of AOPs and gauging the relevance of new testing strategies.

19.
J Phys Chem B ; 112(24): 7320-9, 2008 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-18476739

RESUMO

Microsomal prostaglandin E synthase-1 (mPGES-1) is a promising target for development of next-generation anti-inflammatory drugs. It is crucial for rational design of the next-generation anti-inflammatory drugs to know the three-dimensional (3D) structure of mPGES-1 trimer and to understand how mPGES-1 binds with substrates and inhibitors. In the current work, a 3D structural model of human mPGES-1 trimer has been developed, for the first time, by performing combined homology modeling, molecular docking, and molecular dynamics simulation. The 3D structural model enables us to understand how mPGES-1 binds with its substrates/inhibitors, and the key amino acid residues for the mPGES-1 binding with ligands have been identified. The detailed 3D structures and calculated binding free energies for mPGES-1's binding with substrates and inhibitors are all consistent with available experimental data, suggesting that the 3D model of the mPGES-1 trimer and the enzyme-ligand binding modes are reasonable. The new structural insights obtained from this study should be valuable for rational design of next-generation anti-inflammatory drugs.


Assuntos
Inibidores Enzimáticos/química , Oxirredutases Intramoleculares/química , Modelos Moleculares , Prostaglandina H2/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Domínio Catalítico , Inibidores Enzimáticos/metabolismo , Glutationa/química , Glutationa/metabolismo , Glutationa Transferase/química , Humanos , Oxirredutases Intramoleculares/antagonistas & inibidores , Oxirredutases Intramoleculares/metabolismo , Dados de Sequência Molecular , Prostaglandina H2/metabolismo , Prostaglandina-E Sintases , Ligação Proteica , Conformação Proteica , Ratos , Homologia de Sequência de Aminoácidos , Termodinâmica
20.
BMC Pharmacol Toxicol ; 18(1): 44, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28595649

RESUMO

BACKGROUND: The expanded use of multiple drugs has increased the occurrence of adverse drug reactions (ADRs) induced by drug-drug interactions (DDIs). However, such reactions are typically not observed in clinical drug-development studies because most of them focus on single-drug therapies. ADR reporting systems collect information on adverse health effects caused by both single drugs and DDIs. A major challenge is to unambiguously identify the effects caused by DDIs and to attribute them to specific drug interactions. A computational method that provides prospective predictions of potential DDI-induced ADRs will help to identify and mitigate these adverse health effects. METHOD: We hypothesize that drug-protein interactions can be used as independent variables in predicting ADRs. We constructed drug pair-protein interaction profiles for ~800 drugs using drug-protein interaction information in the public domain. We then constructed statistical models to score drug pairs for their potential to induce ADRs based on drug pair-protein interaction profiles. RESULTS: We used extensive clinical database information to construct categorical prediction models for drug pairs that are likely to induce ADRs via synergistic DDIs and showed that model performance deteriorated only slightly, with a moderate amount of false positives and false negatives in the training samples, as evaluated by our cross-validation analysis. The cross validation calculations showed an average prediction accuracy of 89% across 1,096 ADR models that captured the deleterious effects of synergistic DDIs. Because the models rely on drug-protein interactions, we made predictions for pairwise combinations of 764 drugs that are currently on the market and for which drug-protein interaction information is available. These predictions are publicly accessible at http://avoid-db.bhsai.org . We used the predictive models to analyze broader aspects of DDI-induced ADRs, showing that ~10% of all combinations have the potential to induce ADRs via DDIs. This allowed us to identify potential DDI-induced ADRs not yet clinically reported. The ability of the models to quantify adverse effects between drug classes also suggests that we may be able to select drug combinations that minimize the risk of ADRs. CONCLUSION: Almost all information on DDI-induced ADRs is generated after drug approval. This situation poses significant health risks for vulnerable patient populations with comorbidities. To help mitigate the risks, we developed a robust probabilistic approach to prospectively predict DDI-induced ADRs. Based on this approach, we developed prediction models for 1,096 ADRs and used them to predict the propensity of all pairwise combinations of nearly 800 drugs to be associated with these ADRs via DDIs. We made the predictions publicly available via internet access.


Assuntos
Interações Medicamentosas , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Modelos Estatísticos , Sistemas de Notificação de Reações Adversas a Medicamentos , Bases de Dados Factuais , Humanos , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa