Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Hazard Mater ; 471: 134297, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38677119

RESUMO

Developing mechanistic non-animal testing methods based on the adverse outcome pathway (AOP) framework must incorporate molecular and cellular key events associated with target toxicity. Using data from an in vitro assay and chemical structures, we aimed to create a hybrid model to predict hepatotoxicants. We first curated a reference dataset of 869 compounds for hepatotoxicity modeling. Then, we profiled them against PubChem for existing in vitro toxicity data. Of the 2560 resulting assays, we selected the mitochondrial membrane potential (MMP) assay, a high-throughput screening (HTS) tool that can test chemical disruptors for mitochondrial function. Machine learning was applied to develop quantitative structure-activity relationship (QSAR) models with 2536 compounds tested in the MMP assay for screening new compounds. The MMP assay results, including QSAR model outputs, yielded hepatotoxicity predictions for reference set compounds with a Correct Classification Ratio (CCR) of 0.59. The predictivity improved by including 37 structural alerts (CCR = 0.8). We validated our model by testing 37 reference set compounds in human HepG2 hepatoma cells, and reliably predicting them for hepatotoxicity (CCR = 0.79). This study introduces a novel AOP modeling strategy that combines public HTS data, computational modeling, and experimental testing to predict chemical hepatotoxicity.


Assuntos
Alternativas aos Testes com Animais , Doença Hepática Induzida por Substâncias e Drogas , Aprendizado de Máquina , Potencial da Membrana Mitocondrial , Relação Quantitativa Estrutura-Atividade , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Testes de Toxicidade , Ensaios de Triagem em Larga Escala , Fígado/efeitos dos fármacos , Células Hep G2
2.
Environ Sci Technol ; 57(16): 6573-6588, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37040559

RESUMO

Traditional methodologies for assessing chemical toxicity are expensive and time-consuming. Computational modeling approaches have emerged as low-cost alternatives, especially those used to develop quantitative structure-activity relationship (QSAR) models. However, conventional QSAR models have limited training data, leading to low predictivity for new compounds. We developed a data-driven modeling approach for constructing carcinogenicity-related models and used these models to identify potential new human carcinogens. To this goal, we used a probe carcinogen dataset from the US Environmental Protection Agency's Integrated Risk Information System (IRIS) to identify relevant PubChem bioassays. Responses of 25 PubChem assays were significantly relevant to carcinogenicity. Eight assays inferred carcinogenicity predictivity and were selected for QSAR model training. Using 5 machine learning algorithms and 3 types of chemical fingerprints, 15 QSAR models were developed for each PubChem assay dataset. These models showed acceptable predictivity during 5-fold cross-validation (average CCR = 0.71). Using our QSAR models, we can correctly predict and rank 342 IRIS compounds' carcinogenic potentials (PPV = 0.72). The models predicted potential new carcinogens, which were validated by a literature search. This study portends an automated technique that can be applied to prioritize potential toxicants using validated QSAR models based on extensive training sets from public data resources.


Assuntos
Algoritmos , Relação Quantitativa Estrutura-Atividade , Humanos , Simulação por Computador , Carcinógenos/toxicidade , Bioensaio
3.
Environ Sci Technol ; 56(9): 5984-5998, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35451820

RESUMO

For hazard identification, classification, and labeling purposes, animal testing guidelines are required by law to evaluate the developmental toxicity potential of new and existing chemical products. However, guideline developmental toxicity studies are costly, time-consuming, and require many laboratory animals. Computational modeling has emerged as a promising, animal-sparing, and cost-effective method for evaluating the developmental toxicity potential of chemicals, such as endocrine disruptors, without the use of animals. We aimed to develop a predictive and explainable computational model for developmental toxicants. To this end, a comprehensive dataset of 1244 chemicals with developmental toxicity classifications was curated from public repositories and literature sources. Data from 2140 toxicological high-throughput screening assays were extracted from PubChem and the ToxCast program for this dataset and combined with information about 834 chemical fragments to group assays based on their chemical-mechanistic relationships. This effort revealed two assay clusters containing 83 and 76 assays, respectively, with high positive predictive rates for developmental toxicants identified with animal testing guidelines (PPV = 72.4 and 77.3% during cross-validation). These two assay clusters can be used as developmental toxicity models and were applied to predict new chemicals for external validation. This study provides a new strategy for constructing alternative chemical developmental toxicity evaluations that can be replicated for other toxicity modeling studies.


Assuntos
Ensaios de Triagem em Larga Escala , Testes de Toxicidade , Animais , Bioensaio , Feminino , Substâncias Perigosas , Ensaios de Triagem em Larga Escala/métodos , Gravidez , Medição de Risco , Testes de Toxicidade/métodos
4.
Methods Mol Biol ; 2474: 169-187, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35294765

RESUMO

Advances in high-throughput screening (HTS) revolutionized the environmental and health sciences data landscape. However, new compounds still need to be experimentally synthesized and tested to obtain HTS data, which will still be costly and time-consuming when a large set of new compounds need to be studied against many tests. Quantitative structure-activity relationship (QSAR) modeling is a standard method to fill data gaps for new compounds. The major challenge for many toxicologists, especially those with limited computational backgrounds, is efficiently developing optimized QSAR models for each assay with missing data for certain test compounds. This chapter aims to introduce a freely available and user-friendly QSAR modeling workflow, which trains and optimizes models using five algorithms without the need for a programming background.


Assuntos
Ensaios de Triagem em Larga Escala , Relação Quantitativa Estrutura-Atividade , Algoritmos , Bioensaio
6.
ACS Sustain Chem Eng ; 9(10): 3909-3919, 2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-34239782

RESUMO

Compared to traditional experimental approaches, computational modeling is a promising strategy to efficiently prioritize new candidates with low cost. In this study, we developed a novel data mining and computational modeling workflow proven to be applicable by screening new analgesic opioids. To this end, a large opioid data set was used as the probe to automatically obtain bioassay data from the PubChem portal. There were 114 PubChem bioassays selected to build quantitative structure-activity relationship (QSAR) models based on the testing results across the probe compounds. The compounds tested in each bioassay were used to develop 12 models using the combination of three machine learning approaches and four types of chemical descriptors. The model performance was evaluated by the coefficient of determination (R 2) obtained from 5-fold cross-validation. In total, 49 models developed for 14 bioassays were selected based on the criteria and were identified to be mainly associated with binding affinities to different opioid receptors. The models for these 14 bioassays were further used to fill data gaps in the probe opioids data set and to predict general drug compounds in the DrugBank data set. This study provides a universal modeling strategy that can take advantage of large public data sets for computer-aided drug design (CADD).

8.
Environ Sci Technol ; 55(15): 10875-10887, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34304572

RESUMO

Traditional experimental testing to identify endocrine disruptors that enhance estrogenic signaling relies on expensive and labor-intensive experiments. We sought to design a knowledge-based deep neural network (k-DNN) approach to reveal and organize public high-throughput screening data for compounds with nuclear estrogen receptor α and ß (ERα and ERß) binding potentials. The target activity was rodent uterotrophic bioactivity driven by ERα/ERß activations. After training, the resultant network successfully inferred critical relationships among ERα/ERß target bioassays, shown as weights of 6521 edges between 1071 neurons. The resultant network uses an adverse outcome pathway (AOP) framework to mimic the signaling pathway initiated by ERα and identify compounds that mimic endogenous estrogens (i.e., estrogen mimetics). The k-DNN can predict estrogen mimetics by activating neurons representing several events in the ERα/ERß signaling pathway. Therefore, this virtual pathway model, starting from a compound's chemistry initiating ERα activation and ending with rodent uterotrophic bioactivity, can efficiently and accurately prioritize new estrogen mimetics (AUC = 0.864-0.927). This k-DNN method is a potential universal computational toxicology strategy to utilize public high-throughput screening data to characterize hazards and prioritize potentially toxic compounds.


Assuntos
Rotas de Resultados Adversos , Receptor beta de Estrogênio , Receptor alfa de Estrogênio , Estrogênios , Ensaios de Triagem em Larga Escala , Redes Neurais de Computação
9.
Environ Health Perspect ; 129(4): 47013, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33929906

RESUMO

BACKGROUND: Humans are exposed to tens of thousands of chemical substances that need to be assessed for their potential toxicity. Acute systemic toxicity testing serves as the basis for regulatory hazard classification, labeling, and risk management. However, it is cost- and time-prohibitive to evaluate all new and existing chemicals using traditional rodent acute toxicity tests. In silico models built using existing data facilitate rapid acute toxicity predictions without using animals. OBJECTIVES: The U.S. Interagency Coordinating Committee on the Validation of Alternative Methods (ICCVAM) Acute Toxicity Workgroup organized an international collaboration to develop in silico models for predicting acute oral toxicity based on five different end points: Lethal Dose 50 (LD50 value, U.S. Environmental Protection Agency hazard (four) categories, Globally Harmonized System for Classification and Labeling hazard (five) categories, very toxic chemicals [LD50 (LD50≤50mg/kg)], and nontoxic chemicals (LD50>2,000mg/kg). METHODS: An acute oral toxicity data inventory for 11,992 chemicals was compiled, split into training and evaluation sets, and made available to 35 participating international research groups that submitted a total of 139 predictive models. Predictions that fell within the applicability domains of the submitted models were evaluated using external validation sets. These were then combined into consensus models to leverage strengths of individual approaches. RESULTS: The resulting consensus predictions, which leverage the collective strengths of each individual model, form the Collaborative Acute Toxicity Modeling Suite (CATMoS). CATMoS demonstrated high performance in terms of accuracy and robustness when compared with in vivo results. DISCUSSION: CATMoS is being evaluated by regulatory agencies for its utility and applicability as a potential replacement for in vivo rat acute oral toxicity studies. CATMoS predictions for more than 800,000 chemicals have been made available via the National Toxicology Program's Integrated Chemical Environment tools and data sets (ice.ntp.niehs.nih.gov). The models are also implemented in a free, standalone, open-source tool, OPERA, which allows predictions of new and untested chemicals to be made. https://doi.org/10.1289/EHP8495.


Assuntos
Órgãos Governamentais , Animais , Simulação por Computador , Ratos , Testes de Toxicidade Aguda , Estados Unidos , United States Environmental Protection Agency
10.
Lab Invest ; 101(4): 490-502, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32778734

RESUMO

As defined by the World Health Organization, an endocrine disruptor is an exogenous substance or mixture that alters function(s) of the endocrine system and consequently causes adverse health effects in an intact organism, its progeny, or (sub)populations. Traditional experimental testing regimens to identify toxicants that induce endocrine disruption can be expensive and time-consuming. Computational modeling has emerged as a promising and cost-effective alternative method for screening and prioritizing potentially endocrine-active compounds. The efficient identification of suitable chemical descriptors and machine-learning algorithms, including deep learning, is a considerable challenge for computational toxicology studies. Here, we sought to apply classic machine-learning algorithms and deep-learning approaches to a panel of over 7500 compounds tested against 18 Toxicity Forecaster assays related to nuclear estrogen receptor (ERα and ERß) activity. Three binary fingerprints (Extended Connectivity FingerPrints, Functional Connectivity FingerPrints, and Molecular ACCess System) were used as chemical descriptors in this study. Each descriptor was combined with four machine-learning and two deep- learning (normal and multitask neural networks) approaches to construct models for all 18 ER assays. The resulting model performance was evaluated using the area under the receiver- operating curve (AUC) values obtained from a fivefold cross-validation procedure. The results showed that individual models have AUC values that range from 0.56 to 0.86. External validation was conducted using two additional sets of compounds (n = 592 and n = 966) with established interactions with nuclear ER demonstrated through experimentation. An agonist, antagonist, or binding score was determined for each compound by averaging its predicted probabilities in relevant assay models as an external validation, yielding AUC values ranging from 0.63 to 0.91. The results suggest that multitask neural networks offer advantages when modeling mechanistically related endpoints. Consensus predictions based on the average values of individual models remain the best modeling strategy for computational toxicity evaluations.


Assuntos
Aprendizado de Máquina , Modelos Estatísticos , Receptores de Estrogênio , Algoritmos , Animais , Biologia Computacional , Bases de Dados de Compostos Químicos , Aprendizado Profundo , Disruptores Endócrinos/metabolismo , Disruptores Endócrinos/toxicidade , Humanos , Camundongos , Ligação Proteica , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo
11.
Front Chem ; 8: 597726, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33304885

RESUMO

Understanding the stability of drugs in a forensic toxicology setting is critical for the evaluation of drug concentrations. Synthetic cathinones are new psychoactive substances structurally derived from cathinone, the psychoactive component of Catha edulis ("khat"), a shrub that is indigenous to the Middle East and East Africa. Previous research has evaluated the stability of synthetic cathinones in biological matrices, including blood preserved with the combination of NaF and K2C2O4 used in gray-top tubes. However, it does not assess their stability in blood preserved with Na2EDTA, used for some clinical samples. Further, stability in unpreserved urine samples was only studied for two weeks. This research evaluates the stabilities of four Schedule I synthetic cathinones: mephedrone, MDPV (3,4-methylenedioxypyrovalerone), naphyrone, and α-PVP (alpha-pyrrolidinopentiophenone) at 20°C (room temperature), 4°C (refrigerator), and -20°C (freezer). Stability was assessed in methanolic and acetonitrile solutions, as well as in Na2EDTA-preserved blood and unpreserved urine. Solutions (1 mg/L) of each drug in each matrix stored in aliquots (100 µL, solvents; 1.2 mL, biological samples; n = 12) at each of the three temperatures for triplicate analysis on days 3, 7, 14, and 30. On day 0 of each study, three additional aliquots of each solution were analyzed. Biological samples underwent solid-phase extraction before analysis. All samples were analyzed in full-scan by gas chromatography-mass spectrometry (GC-MS). The results of this study show that under room temperature and refrigerator storage conditions, mephedrone, naphyrone, and MDPV will degrade in methanol. This degradation starts are early as day 3. Additionally, all four drugs will degrade in Na2EDTA-preserved human whole blood samples in at least one evaluated storage environment. However, when in acetonitrile-based working solutions and unpreserved urine samples, they proved to be more stable. Methanolic working solutions and samples of Na2EDTA-preserved blood containing these cathinones should be stored in the freezer and used or tested with urgency to ensure that quantitative sample analysis is as accurate as possible in forensic casework.

12.
Drug Discov Today ; 25(9): 1624-1638, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32663517

RESUMO

Advancing a new drug to market requires substantial investments in time as well as financial resources. Crucial bioactivities for drug candidates, including their efficacy, pharmacokinetics (PK), and adverse effects, need to be investigated during drug development. With advancements in chemical synthesis and biological screening technologies over the past decade, a large amount of biological data points for millions of small molecules have been generated and are stored in various databases. These accumulated data, combined with new machine learning (ML) approaches, such as deep learning, have shown great potential to provide insights into relevant chemical structures to predict in vitro, in vivo, and clinical outcomes, thereby advancing drug discovery and development in the big data era.


Assuntos
Desenho de Fármacos , Descoberta de Drogas , Animais , Big Data , Humanos , Aprendizado de Máquina , Modelos Teóricos
13.
Chem Res Toxicol ; 32(4): 536-547, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30907586

RESUMO

In 2016, the Frank R. Lautenberg Chemical Safety for the 21st Century Act became the first US legislation to advance chemical safety evaluations by utilizing novel testing approaches that reduce the testing of vertebrate animals. Central to this mission is the advancement of computational toxicology and artificial intelligence approaches to implementing innovative testing methods. In the current big data era, the terms volume (amount of data), velocity (growth of data), and variety (the diversity of sources) have been used to characterize the currently available chemical, in vitro, and in vivo data for toxicity modeling purposes. Furthermore, as suggested by various scientists, the variability (internal consistency or lack thereof) of publicly available data pools, such as PubChem, also presents significant computational challenges. The development of novel artificial intelligence approaches based on public massive toxicity data is urgently needed to generate new predictive models for chemical toxicity evaluations and make the developed models applicable as alternatives for evaluating untested compounds. In this procedure, traditional approaches (e.g., QSAR) purely based on chemical structures have been replaced by newly designed data-driven and mechanism-driven modeling. The resulting models realize the concept of adverse outcome pathway (AOP), which can not only directly evaluate toxicity potentials of new compounds, but also illustrate relevant toxicity mechanisms. The recent advancement of computational toxicology in the big data era has paved the road to future toxicity testing, which will significantly impact on the public health.


Assuntos
Inteligência Artificial , Biologia Computacional , Testes de Toxicidade , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...