Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
2.
Expert Opin Drug Discov ; 18(11): 1231-1243, 2023.
Article in English | MEDLINE | ID: mdl-37639708

ABSTRACT

INTRODUCTION: Drug discovery has provided modern societies with the means to fight against many diseases. In this sense, computational methods have been at the forefront, playing an important role in rationalizing the search for novel drugs. Yet, tackling phenomena such as the multi-genic nature of diseases and drug resistance are limitations of the current computational methods. Multi-tasking models for quantitative structure-biological effect relationships (mtk-QSBER) have emerged to overcome such limitations. AREAS COVERED: The present review describes an update on the fundamentals and applications of the mtk-QSBER models as tools to accelerate multiple stages/substages of the drug discovery process. EXPERT OPINION: Computational approaches are extremely important for the rationalization of the search for novel and efficacious therapeutic agents. However, they need to focus more on the multi-target drug discovery paradigm. In this sense, mtk-QSBER models are particularly suited for multi-target drug discovery, offering encouraging opportunities across multiple therapeutic areas and scientific disciplines associated with drug discovery.


Subject(s)
Drug Discovery , Quantitative Structure-Activity Relationship , Humans , Drug Discovery/methods , Drug Delivery Systems , Drug Design
4.
ACS Omega ; 7(36): 32119-32130, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36120024

ABSTRACT

Respiratory viruses are infectious agents, which can cause pandemics. Although nowadays the danger associated with respiratory viruses continues to be evidenced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as the virus responsible for the current COVID-19 pandemic, other viruses such as SARS-CoV-1, the influenza A and B viruses (IAV and IBV, respectively), and the respiratory syncytial virus (RSV) can lead to globally spread viral diseases. Also, from a biological point of view, most of these viruses can cause an organ-damaging hyperinflammatory response known as the cytokine storm (CS). Computational approaches constitute an essential component of modern drug development campaigns, and therefore, they have the potential to accelerate the discovery of chemicals able to simultaneously inhibit multiple molecular and nonmolecular targets. We report here the first multicondition model based on quantitative structure-activity relationships and an artificial neural network (mtc-QSAR-ANN) for the virtual design and prediction of molecules with dual pan-antiviral and anti-CS profiles. Our mtc-QSAR-ANN model exhibited an accuracy higher than 80%. By interpreting the different descriptors present in the mtc-QSAR-ANN model, we could retrieve several molecular fragments whose assembly led to new molecules with drug-like properties and predicted pan-antiviral and anti-CS activities.

5.
Biomedicines ; 10(2)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35203699

ABSTRACT

Pancreatic cancer (PANC) is a dangerous type of cancer that is a major cause of mortality worldwide and exhibits a remarkably poor prognosis. To date, discovering anti-PANC agents remains a very complex and expensive process. Computational approaches can accelerate the search for anti-PANC agents. We report for the first time two models that combined perturbation theory with machine learning via a multilayer perceptron network (PTML-MLP) to perform the virtual design and prediction of molecules that can simultaneously inhibit multiple PANC cell lines and PANC-related proteins, such as caspase-1, tumor necrosis factor-alpha (TNF-alpha), and the insulin-like growth factor 1 receptor (IGF1R). Both PTML-MLP models exhibited accuracies higher than 78%. Using the interpretation from one of the PTML-MLP models as a guideline, we extracted different molecular fragments desirable for the inhibition of the PANC cell lines and the aforementioned PANC-related proteins and then assembled some of those fragments to form three new molecules. The two PTML-MLP models predicted the designed molecules as potentially versatile anti-PANC agents through inhibition of the three PANC-related proteins and multiple PANC cell lines. Conclusions: This work opens new horizons for the application of the PTML modeling methodology to anticancer research.

6.
Mol Divers ; 26(5): 2523-2534, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34802116

ABSTRACT

Hypertension is a medical condition that affects millions of people worldwide. Despite the high efficacy of the current antihypertensive drugs, they are associated with serious side effects. Peptides constitute attractive options for chemical therapy against hypertension, and computational models can accelerate the design of antihypertensive peptides. Yet, to the best of our knowledge, all the in silico models predict only the antihypertensive activity of peptides while neglecting their inherent toxic potential to red blood cells. In this work, we report the first sequence-based model that combines perturbation theory and machine learning through multilayer perceptron networks (SB-PTML-MLP) to enable the simultaneous screening of antihypertensive activity and hemotoxicity of peptides. We have interpreted the molecular descriptors present in the model from a physicochemical and structural point of view. By strictly following such interpretations as guidelines, we performed two tasks. First, we selected amino acids with favorable contributions to both the increase of the antihypertensive activity and the diminution of hemotoxicity. Then, we assembled those suitable amino acids, virtually designing peptides that were predicted by the SB-PTML-MLP model as antihypertensive agents exhibiting low hemotoxicity. The potentiality of the SB-PTML-MLP model as a tool for designing potent and safe antihypertensive peptides was confirmed by predictions performed by online computational tools reported in the scientific literature. The methodology presented here can be extended to other pharmacological applications of peptides.


Subject(s)
Antihypertensive Agents , Hypertension , Amino Acids , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Humans , Hypertension/drug therapy , Machine Learning , Peptides/chemistry , Peptides/pharmacology
7.
Biomolecules ; 11(12)2021 12 04.
Article in English | MEDLINE | ID: mdl-34944476

ABSTRACT

Inflammation involves a complex biological response of the body tissues to damaging stimuli. When dysregulated, inflammation led by biomolecular mediators such as caspase-1 and tumor necrosis factor-alpha (TNF-alpha) can play a detrimental role in the progression of different medical conditions such as cancer, neurological disorders, autoimmune diseases, and cytokine storms caused by viral infections such as COVID-19. Computational approaches can accelerate the search for dual-target drugs able to simultaneously inhibit the aforementioned proteins, enabling the discovery of wide-spectrum anti-inflammatory agents. This work reports the first multicondition model based on quantitative structure-activity relationships and a multilayer perceptron neural network (mtc-QSAR-MLP) for the virtual screening of agency-regulated chemicals as versatile anti-inflammatory therapeutics. The mtc-QSAR-MLP model displayed accuracy higher than 88%, and was interpreted from a physicochemical and structural point of view. When using the mtc-QSAR-MLP model as a virtual screening tool, we could identify several agency-regulated chemicals as dual inhibitors of caspase-1 and TNF-alpha, and the experimental information later retrieved from the scientific literature converged with our computational results. This study supports the capabilities of our mtc-QSAR-MLP model in anti-inflammatory therapy with direct applications to current health issues such as the COVID-19 pandemic.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Caspase Inhibitors/pharmacology , Drug Repositioning/methods , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Anti-Inflammatory Agents/chemistry , Caspase 1/metabolism , Caspase Inhibitors/chemistry , Humans , Inflammation/drug therapy , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , COVID-19 Drug Treatment
8.
Curr Top Med Chem ; 21(30): 2687-2693, 2021.
Article in English | MEDLINE | ID: mdl-34636311

ABSTRACT

Respiratory viruses continue to afflict mankind. Among them, pathogens such as coronaviruses [including the current pandemic agent known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)] and the one causing influenza A (IAV) are highly contagious and deadly. These can evade the immune system defenses while causing a hyperinflammatory response that can damage different tissues/organs. Simultaneously targeting several immunomodulatory proteins is a plausible antiviral strategy since it could lead to the discovery of indirect-acting pan-antiviral (IAPA) agents for the treatment of diseases caused by respiratory viruses. In this context, computational approaches, which are an essential part of the modern drug discovery campaigns, could accelerate the identification of multi-target immunomodulators. This perspective discusses the usefulness of computational multi-target drug discovery for the virtual screening (drug repurposing) of IAPA agents capable of boosting the immune system through the activation of the toll-like receptor 7 (TLR7) and/or the stimulator of interferon genes (STING) while inhibiting key inflammation-related proteins such as caspase-1 and tumor necrosis factor-alpha (TNF-α).


Subject(s)
Antiviral Agents , Drug Discovery , Respiratory Tract Infections/drug therapy , Antiviral Agents/pharmacology , COVID-19 , Computational Biology , Drug Evaluation, Preclinical , Humans , Pandemics , Respiratory Tract Infections/virology , SARS-CoV-2/drug effects
9.
Antibiotics (Basel) ; 10(8)2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34439055

ABSTRACT

Tuberculosis remains the most afflicting infectious disease known by humankind, with one quarter of the population estimated to have it in the latent state. Discovering antituberculosis drugs is a challenging, complex, expensive, and time-consuming task. To overcome the substantial costs and accelerate drug discovery and development, drug repurposing has emerged as an attractive alternative to find new applications for "old" drugs and where computational approaches play an essential role by filtering the chemical space. This work reports the first multi-condition model based on quantitative structure-activity relationships and an ensemble of neural networks (mtc-QSAR-EL) for the virtual screening of potential antituberculosis agents able to act as multi-strain inhibitors. The mtc-QSAR-EL model exhibited an accuracy higher than 85%. A physicochemical and fragment-based structural interpretation of this model was provided, and a large dataset of agency-regulated chemicals was virtually screened, with the mtc-QSAR-EL model identifying already proven antituberculosis drugs while proposing chemicals with great potential to be experimentally repurposed as antituberculosis (multi-strain inhibitors) agents. Some of the most promising molecules identified by the mtc-QSAR-EL model as antituberculosis agents were also confirmed by another computational approach, supporting the capabilities of the mtc-QSAR-EL model as an efficient tool for computational drug repurposing.

10.
Front Chem ; 9: 634663, 2021.
Article in English | MEDLINE | ID: mdl-33777898

ABSTRACT

Parasitic diseases remain as unresolved health issues worldwide. While for some parasites the treatments involve drug combinations with serious side effects, for others, chemical therapies are inefficient due to the emergence of drug resistance. This urges the search for novel antiparasitic agents able to act through multiple mechanisms of action. Here, we report the first multi-target model based on quantitative structure-activity relationships and a multilayer perceptron neural network (mt-QSAR-MLP) to virtually design and predict versatile inhibitors of proteins involved in the survival and/or infectivity of different pathogenic parasites. The mt-QSAR-MLP model exhibited high accuracy (>80%) in both training and test sets for the classification/prediction of protein inhibitors. Several fragments were directly extracted from the physicochemical and structural interpretations of the molecular descriptors in the mt-QSAR-MLP model. Such interpretations enabled the generation of four molecules that were predicted as multi-target inhibitors against at least three of the five parasitic proteins reported here with two of the molecules being predicted to inhibit all the proteins. Docking calculations converged with the mt-QSAR-MLP model regarding the multi-target profile of the designed molecules. The designed molecules exhibited drug-like properties, complying with Lipinski's rule of five, as well as Ghose's filter and Veber's guidelines.

11.
Curr Top Med Chem ; 21(7): 661-675, 2021.
Article in English | MEDLINE | ID: mdl-33463472

ABSTRACT

BACKGROUND: Cyclin-dependent kinase 4 (CDK4) and the human epidermal growth factor receptor 2 (HER2) are two of the most promising targets in oncology research. Thus, a series of computational approaches have been applied to the search for more potent inhibitors of these cancerrelated proteins. However, current approaches have focused on chemical analogs while predicting the inhibitory activity against only one of these targets, but never against both. AIMS: We report the first perturbation model combined with machine learning (PTML) to enable the design and prediction of dual inhibitors of CDK4 and HER2. METHODS: Inhibition data for CDK4 and HER2 were extracted from ChEMBL. The PTML model relied on artificial neural networks to allow the classification/prediction of molecules as active or inactive against CDK4 and/or HER2. RESULTS: The PTML model displayed sensitivity and specificity higher than 80% in the training set. The same statistical metrics had values above 75% in the test set. We extracted several molecular fragments and estimated their quantitative contributions to the inhibitory activity against CDK4 and HER2. Guided by the physicochemical and structural interpretations of the molecular descriptors in the PTML model, we designed six molecules by assembling several fragments with positive contributions. Three of these molecules were predicted as potent dual inhibitors of CDK4 and HER2, while the other three were predicted as inhibitors of at least one of these proteins. All the molecules complied with Lipinski's rule of five and its variants. CONCLUSION: The present work represents an encouraging alternative for future anticancer chemotherapies.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/chemistry , Programming Languages , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Humans , Molecular Structure , Neural Networks, Computer , Receptor, ErbB-2/antagonists & inhibitors
12.
Curr Top Med Chem ; 20(19): 1661-1676, 2020.
Article in English | MEDLINE | ID: mdl-32515311

ABSTRACT

BACKGROUND: Alzheimer's disease is characterized by a progressive pattern of cognitive and functional impairment, which ultimately leads to death. Computational approaches have played an important role in the context of drug discovery for anti-Alzheimer's therapies. However, most of the computational models reported to date have been focused on only one protein associated with Alzheimer's, while relying on small datasets of structurally related molecules. OBJECTIVE: We introduce the first model combining perturbation theory and machine learning based on artificial neural networks (PTML-ANN) for simultaneous prediction and design of inhibitors of three Alzheimer's disease-related proteins, namely glycogen synthase kinase 3 beta (GSK3B), histone deacetylase 1 (HDAC1), and histone deacetylase 6 (HDAC6). METHODS: The PTML-ANN model was obtained from a dataset retrieved from ChEMBL, and it relied on a classification approach to predict chemicals as active or inactive. RESULTS: The PTML-ANN model displayed sensitivity and specificity higher than 85% in both training and test sets. The physicochemical and structural interpretation of the molecular descriptors in the model permitted the direct extraction of fragments suggested to favorably contribute to enhancing the multitarget inhibitory activity. Based on this information, we assembled ten molecules from several fragments with positive contributions. Seven of these molecules were predicted as triple target inhibitors while the remaining three were predicted as dual-target inhibitors. The estimated physicochemical properties of the designed molecules complied with Lipinski's rule of five and its variants. CONCLUSION: This work opens new horizons toward the design of multi-target inhibitors for anti- Alzheimer's therapies.


Subject(s)
Alzheimer Disease/drug therapy , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Machine Learning , Neural Networks, Computer , Protein Kinase Inhibitors/pharmacology , Alzheimer Disease/metabolism , Drug Design , Glycogen Synthase Kinase 3 beta/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry
13.
Mini Rev Med Chem ; 20(14): 1357-1374, 2020.
Article in English | MEDLINE | ID: mdl-32013845

ABSTRACT

Fragment-Based Drug Design (FBDD) has established itself as a promising approach in modern drug discovery, accelerating and improving lead optimization, while playing a crucial role in diminishing the high attrition rates at all stages in the drug development process. On the other hand, FBDD has benefited from the application of computational methodologies, where the models derived from the Quantitative Structure-Activity Relationships (QSAR) have become consolidated tools. This mini-review focuses on the evolution and main applications of the QSAR paradigm in the context of FBDD in the last five years. This report places particular emphasis on the QSAR models derived from fragment-based topological approaches to extract physicochemical and/or structural information, allowing to design potentially novel mono- or multi-target inhibitors from relatively large and heterogeneous databases. Here, we also discuss the need to apply multi-scale modeling, to exemplify how different datasets based on target inhibition can be simultaneously integrated and predicted together with other relevant endpoints such as the biological activity against non-biomolecular targets, as well as in vitro and in vivo toxicity and pharmacokinetic properties. In this context, seminal papers are briefly analyzed. As huge amounts of data continue to accumulate in the domains of the chemical, biological and biomedical sciences, it has become clear that drug discovery must be viewed as a multi-scale optimization process. An ideal multi-scale approach should integrate diverse chemical and biological data and also serve as a knowledge generator, enabling the design of potentially optimal chemicals that may become therapeutic agents.


Subject(s)
Drug Design , Models, Molecular , Quantitative Structure-Activity Relationship , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Gram-Negative Bacteria/drug effects , HIV Infections/drug therapy , Hepacivirus/drug effects , Humans , Neural Networks, Computer
14.
Nanotoxicology ; 11(7): 891-906, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28937298

ABSTRACT

Nanoparticles (NPs) are part of our daily life, having a wide range of applications in engineering, physics, chemistry, and biomedicine. However, there are serious concerns regarding the harmful effects that NPs can cause to the different biological systems and their ecosystems. Toxicity testing is an essential step for assessing the potential risks of the NPs, but the experimental assays are often very expensive and usually too slow to flag the number of NPs that may cause adverse effects. In silico models centered on quantitative structure-activity/toxicity relationships (QSAR/QSTR) are alternative tools that have become valuable supports to risk assessment, rationalizing the search for safer NPs. In this work, we develop a unified QSTR-perturbation model based on artificial neural networks, aimed at simultaneously predicting general toxicity profiles of NPs under diverse experimental conditions. The model is derived from 54,371 NP-NP pair cases generated by applying the perturbation theory to a set of 260 unique NPs, and showed an accuracy higher than 97% in both training and validation sets. Physicochemical interpretation of the different descriptors in the model are additionally provided. The QSTR-perturbation model is then employed to predict the toxic effects of several NPs not included in the original dataset. The theoretical results obtained for this independent set are strongly consistent with the experimental evidence found in the literature, suggesting that the present QSTR-perturbation model can be viewed as a promising and reliable computational tool for probing the toxicity of NPs.


Subject(s)
Computer Simulation , Machine Learning , Models, Theoretical , Nanoparticles/chemistry , Nanoparticles/toxicity , Neural Networks, Computer , Animals , Humans , Predictive Value of Tests , Quantitative Structure-Activity Relationship , Risk Assessment , Toxicity Tests
15.
ACS Comb Sci ; 18(8): 490-8, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27280735

ABSTRACT

Antimicrobial peptides (AMPs) represent promising alternatives to fight against bacterial pathogens. However, cellular toxicity remains one of the main concerns in the early development of peptide-based drugs. This work introduces the first multitasking (mtk) computational model focused on performing simultaneous predictions of antibacterial activities, and cytotoxicities of peptides. The model was created from a data set containing 3592 cases, and it displayed accuracy higher than 96% for classifying/predicting peptides in both training and prediction (test) sets. The technique known as alanine scanning was computationally applied to illustrate the calculation of the quantitative contributions of the amino acids (in their respective positions of the sequence) to the biological effects of a defined peptide. A small library formed by 10 peptides was generated, where peptides were designed by considering the interpretations of the different descriptors in the mtk-computational model. All the peptides were predicted to exhibit high antibacterial activities against multiple bacterial strains, and low cytotoxicity against various cell types. The present mtk-computational model can be considered a very useful tool to support high throughput research for the discovery of potent and safe AMPs.


Subject(s)
Anti-Bacterial Agents/chemistry , Peptides/chemistry , Amino Acids/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Cell Line , Drug Discovery , Gram-Negative Bacteria/drug effects , High-Throughput Screening Assays , Humans , Mice , Molecular Structure , Peptides/pharmacology , Peptides/toxicity , Quantitative Structure-Activity Relationship , Rats
16.
J Chem Inf Model ; 56(3): 588-98, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-26960000

ABSTRACT

Antimicrobial peptides (AMPs) have emerged as promising therapeutic alternatives to fight against the diverse infections caused by different pathogenic microorganisms. In this context, theoretical approaches in bioinformatics have paved the way toward the creation of several in silico models capable of predicting antimicrobial activities of peptides. All current models have several significant handicaps, which prevent the efficient search for highly active AMPs. Here, we introduce the first multitarget (mt) chemo-bioinformatic model devoted to performing alignment-free prediction of antibacterial activity of peptides against multiple Gram-positive bacterial strains. The model was constructed from a data set containing 2488 cases of AMPs sequences assayed against at least 1 out of 50 Gram-positive bacterial strains. This mt-chemo-bioinformatic model displayed percentages of correct classification higher than 90.00% in both training and prediction (test) sets. For the first time, two computational approaches derived from basic concepts in genetics and molecular biology were applied, allowing the calculations of the relative contributions of any amino acid (in a defined position) to the antibacterial activity of an AMP and depending on the bacterial strain used in the biological assay. The present mt-chemo-bioinformatic model constitutes a powerful tool to enable the discovery of potent and versatile AMPs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Computational Biology , Gram-Positive Bacteria/drug effects , Peptides/pharmacology , Microbial Sensitivity Tests
17.
Mini Rev Med Chem ; 15(8): 677-86, 2015.
Article in English | MEDLINE | ID: mdl-25694074

ABSTRACT

The assessment of acute toxicity is one of the most important stages to ensure the safety of chemicals with potential applications in pharmaceutical sciences, biomedical research, or any other industrial branch. A huge and indiscriminate number of toxicity assays have been carried out on laboratory animals. In this sense, computational approaches involving models based on quantitative-structure activity/toxicity relationships (QSAR/QSTR) can help to rationalize time and financial costs. Here, we discuss the most significant advances in the last 6 years focused on the use of QSAR/QSTR models to predict acute toxicity of drugs/chemicals in laboratory animals, employing large and heterogeneous datasets. The advantages and drawbacks of the different QSAR/QSTR models are analyzed. As a contribution to the field, we introduce the first multitasking (mtk) QSTR model for simultaneous prediction of acute toxicity of compounds by considering different routes of administration, diverse breeds of laboratory animals, and the reliability of the experimental conditions. The mtk-QSTR model was based on artificial neural networks (ANN), allowing the classification of compounds as toxic or non-toxic. This model correctly classified more than 94% of the 1646 cases present in the whole dataset, and its applicability was demonstrated by performing predictions of different chemicals such as drugs, dietary supplements, and molecules which could serve as nanocarriers for drug delivery. The predictions given by the mtk-QSTR model are in very good agreement with the experimental results.


Subject(s)
Computer Simulation , Drug Discovery , Models, Biological , Neural Networks, Computer , Quantitative Structure-Activity Relationship , Toxicological Phenomena , Animals , Anti-HIV Agents/toxicity , Antiprotozoal Agents/toxicity , Benzylamines , Cyclams , Databases, Pharmaceutical , Drug Discovery/methods , Fullerenes/toxicity , Heterocyclic Compounds/toxicity , Humans , Models, Molecular , Pharmaceutical Preparations/administration & dosage , Thiamine/toxicity , Tinidazole/toxicity , Vitamin B Complex/toxicity
18.
Nanomedicine (Lond) ; 10(2): 193-204, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25600965

ABSTRACT

AIMS: We introduce the first quantitative structure-activity relationship (QSAR) perturbation model for probing multiple antibacterial profiles of nanoparticles (NPs) under diverse experimental conditions. MATERIALS & METHODS: The dataset is based on 300 nanoparticles containing dissimilar chemical compositions, sizes, shapes and surface coatings. In general terms, the NPs were tested against different bacteria, by considering several measures of antibacterial activity and diverse assay times. The QSAR perturbation model was created from 69,231 nanoparticle-nanoparticle (NP-NP) pairs, which were randomly generated using a recently reported perturbation theory approach. RESULTS: The model displayed an accuracy rate of approximately 98% for classifying NPs as active or inactive, and a new copper-silver nanoalloy was correctly predicted by this model with consensus accuracy of 77.73%. CONCLUSION: Our QSAR perturbation model can be used as an efficacious tool for the virtual screening of antibacterial nanomaterials.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Nanomedicine/methods , Nanoparticles/chemistry , Nanostructures/chemistry , Quantitative Structure-Activity Relationship , Alloys/chemistry , Escherichia coli/drug effects , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects
19.
Environ Sci Technol ; 48(24): 14686-94, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25384130

ABSTRACT

Nanomaterials have revolutionized modern science and technology due to their multiple applications in engineering, physics, chemistry, and biomedicine. Nevertheless, the use and manipulation of nanoparticles (NPs) can bring serious damages to living organisms and their ecosystems. For this reason, ecotoxicity and cytotoxicity assays are of special interest in order to determine the potential harmful effects of NPs. Processes based on ecotoxicity and cytotoxicity tests can significantly consume time and financial resources. In this sense, alternative approaches such as quantitative structure-activity/toxicity relationships (QSAR/QSTR) modeling have provided important insights for the better understanding of the biological behavior of NPs that may be responsible for causing toxicity. Until now, QSAR/QSTR models have predicted ecotoxicity or cytotoxicity separately against only one organism (bioindicator species or cell line) and have not reported information regarding the quantitative influence of characteristics other than composition or size. In this work, we developed a unified QSTR-perturbation model to simultaneously probe ecotoxicity and cytotoxicity of NPs under different experimental conditions, including diverse measures of toxicities, multiple biological targets, compositions, sizes and conditions to measure those sizes, shapes, times during which the biological targets were exposed to NPs, and coating agents. The model was created from 36488 cases (NP-NP pairs) and exhibited accuracies higher than 98% in both training and prediction sets. The model was used to predict toxicities of several NPs that were not included in the original data set. The results of the predictions suggest that the present QSTR-perturbation model can be employed as a highly promising tool for the fast and efficient assessment of ecotoxicity and cytotoxicity of NPs.


Subject(s)
Nanostructures/chemistry , Nanostructures/toxicity , Quantitative Structure-Activity Relationship , Risk Assessment/methods , Animals , Ecotoxicology/methods , Nanoparticles/chemistry , Nanoparticles/toxicity
20.
Environ Int ; 73: 288-94, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25173945

ABSTRACT

Nanotechnology has brought great advances to many fields of modern science. A manifold of applications of nanoparticles have been found due to their interesting optical, electrical, and biological/chemical properties. However, the potential toxic effects of nanoparticles to different ecosystems are of special concern nowadays. Despite the efforts of the scientific community, the mechanisms of toxicity of nanoparticles are still poorly understood. Quantitative-structure activity/toxicity relationships (QSAR/QSTR) models have just started being useful computational tools for the assessment of toxic effects of nanomaterials. But most QSAR/QSTR models have been applied so far to predict ecotoxicity against only one organism/bio-indicator such as Daphnia magna. This prevents having a deeper knowledge about the real ecotoxic effects of nanoparticles, and consequently, there is no possibility to establish an efficient risk assessment of nanomaterials in the environment. In this work, a perturbation model for nano-QSAR problems is introduced with the aim of simultaneously predicting the ecotoxicity of different nanoparticles against several assay organisms (bio-indicators), by considering also multiple measures of ecotoxicity, as well as the chemical compositions, sizes, conditions under which the sizes were measured, shapes, and the time during which the diverse assay organisms were exposed to nanoparticles. The QSAR-perturbation model was derived from a database containing 5520 cases (nanoparticle-nanoparticle pairs), and it was shown to exhibit accuracies of ca. 99% in both training and prediction sets. In order to demonstrate the practical applicability of our model, three different nickel-based nanoparticles (Ni) with experimental values reported in the literature were predicted. The predictions were found to be in very good agreement with the experimental evidences, confirming that Ni-nanoparticles are not ecotoxic when compared with other nanoparticles. The results of this study thus provide a single valuable tool toward an efficient prediction of the ecotoxicity of nanoparticles under multiple experimental conditions.


Subject(s)
Nanoparticles/toxicity , Toxicity Tests/methods , Animals , Computational Biology , Daphnia/drug effects , Ecotoxicology/methods , Nanoparticles/chemistry , Nickel/toxicity , Quantitative Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...