Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
PLoS One ; 19(1): e0296727, 2024.
Article in English | MEDLINE | ID: mdl-38277388

ABSTRACT

Carbapenem-resistant Enterobacterales (CRE) is considered a paramount threat due to its rapid spread and high mortality rate. Klebsiella pneumoniae carbapenemases (KPCs), specifically KPC-2, are prevalent enzymes responsible for carbapenem resistance in many countries. While combinations of antibiotics are commonly used, they must be tailored to match the remaining susceptibility of the infecting strains. Therefore, there is a need to develop the ß-lactamase inhibitor to effectively address this issue. ß-lactamase inhibitor protein (BLIP) and its variants, BLIP-I and BLIP-II, have demonstrated the ability to inhibit class A ß-lactamases. In particular, BLIP-II shows strong binding to the KPC-2 carbapenemase, making it a potential candidate for inhibition. To improve the intracellular penetration of BLIP-II, a cell-penetrating peptide (CPP) was employed. In this study, a KRK-rich peptide was introduced at either the N-terminal or C-terminal region of tBLIP-II, excluding the signal sequence of the BLIP-II protein. tBLIP-II, tBLIP-II-CPP, and CPP-BLIP-II were successfully expressed, and the chimeric proteins retained inhibitory activity compared to tBLIP-II alone. It is apparent that homology modeling demonstrated neither the poly-histidine tag nor the CPP interfered with the essential interaction residues of tBLIP-II. Interestingly, BLIP-II-CPP exhibited the highest inhibitory activity, reducing the minimal inhibitory concentration (MIC) of meropenem by 8 folds. Moreover, the combination of tBLIP-CPP with meropenem significantly decreased the viable bacterial cell count compared to the combination of tBLIP-II with meropenem or meropenem alone. These findings suggest that tBLIP-CPP is a promising candidate for restoring carbapenem susceptibility against CRE and provides a valuable therapeutic option for infections caused by CRE.


Subject(s)
Cell-Penetrating Peptides , beta-Lactamase Inhibitors , Meropenem/pharmacology , beta-Lactamase Inhibitors/pharmacology , Klebsiella pneumoniae , Cell-Penetrating Peptides/pharmacology , Cephalosporinase , Anti-Bacterial Agents/pharmacology , beta-Lactamases/metabolism , Bacterial Proteins/metabolism , Carbapenems/pharmacology , Microbial Sensitivity Tests
2.
ACS Omega ; 8(46): 43500-43510, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38027387

ABSTRACT

Angiotensin-converting enzyme inhibitors (ACEIs) play a crucial role in treating conditions such as hypertension, heart failure, and kidney diseases. Nevertheless, the ACEIs currently available on the market are linked to a variety of adverse effects including renal insufficiency, which restricts their usage. There is thus an urgent need to optimize the currently available ACEIs. This study represents a structure-activity relationship investigation of ACEIs, employing machine learning to analyze data sets sourced from the ChEMBL database. Exploratory data analysis was performed to visualize the physicochemical properties of compounds by investigating the distributions, patterns, and statistical significance among the different bioactivity groups. Further scaffold analysis has identified 9 representative Murcko scaffolds with frequencies ≥10. Scaffold diversity has revealed that active ACEIs had more scaffold diversity than their intermediate and inactive counterparts, thereby indicating the significance of performing lead optimization on scaffolds of active ACEIs. Scaffolds 1, 3, 6, and 8 are unfavorable in comparison with scaffolds 2, 3, 5, 7, and 9. QSAR investigation of compiled data sets consisting of 549 compounds led to the selection of Mordred descriptor and Random Forest algorithm as the best model, which afforded robust model performance (accuracy: 0.981, 0.77, and 0.745; MCC: 0.972, 0.658, and 0.617 for the training set, 10-fold cross-validation set, and testing set, respectively). To enhance the model's robustness and predictability, we reduced the chemical diversity of the input compounds by using the 9 most prevalent Murcko scaffold-matched compounds (comprising a total of 168) followed by a subsequent QSAR model investigation using Mordred descriptor and extremely gradient boost algorithm (accuracy: 0.973, 0.849, and 0.823; MCC: 0.959, 0.786, and 0.742 for the training set, 10-fold cross-validation set, and testing set, respectively). Further illustration of the structure-activity relationship using SALI plots has enabled the identification of clusters of compounds that create activity cliffs. These findings, as presented in this study, contribute to the advancement of drug discovery and the optimization of ACEIs.

3.
EXCLI J ; 22: 975-991, 2023.
Article in English | MEDLINE | ID: mdl-38023567

ABSTRACT

Antimicrobial resistance (AMR) has emerged as one of the global threats to human health in the 21st century. Drug discovery of inhibitors against novel targets rather than conventional bacterial targets has been considered an inevitable strategy for the growing threat of AMR infections. In this study, we applied quantitative structure-activity relationship (QSAR) modeling to the LpxC inhibitors to predict the inhibitory activity. In addition, we performed various cheminformatics analysis consisting of the exploration of the chemical space, identification of chemotypes, performing structure-activity landscape and activity cliffs as well as construction of the Structure-Activity Similarity (SAS) map. We built a total of 24 QSAR classification models using PubChem and MACCS fingerprint with 12 various machine learning algorithms. The best model with PubChem fingerprint is the Extremely Gradient Boost model (accuracy on the training set: 0.937; accuracy on the 10-fold cross-validation set: 0.795; accuracy on the test set: 0.799). Furthermore, it was found that the best model using the MACCS fingerprint was the Random Forest model (accuracy on the training set: 0.955; accuracy on the 10-fold cross-validation set: 0.803; accuracy on the test set: 0.785). In addition, we have identified eight consensus activity cliff generators that are highly informative for further SAR investigations. It is hoped that findings presented herein can provide guidance for further lead optimization of LpxC inhibitors.

4.
J Cheminform ; 15(1): 50, 2023 May 06.
Article in English | MEDLINE | ID: mdl-37149650

ABSTRACT

Drug resistance represents a major obstacle to therapeutic innovations and is a prevalent feature in prostate cancer (PCa). Androgen receptors (ARs) are the hallmark therapeutic target for prostate cancer modulation and AR antagonists have achieved great success. However, rapid emergence of resistance contributing to PCa progression is the ultimate burden of their long-term usage. Hence, the discovery and development of AR antagonists with capability to combat the resistance, remains an avenue for further exploration. Therefore, this study proposes a novel deep learning (DL)-based hybrid framework, named DeepAR, to accurately and rapidly identify AR antagonists by using only the SMILES notation. Specifically, DeepAR is capable of extracting and learning the key information embedded in AR antagonists. Firstly, we established a benchmark dataset by collecting active and inactive compounds against AR from the ChEMBL database. Based on this dataset, we developed and optimized a collection of baseline models by using a comprehensive set of well-known molecular descriptors and machine learning algorithms. Then, these baseline models were utilized for creating probabilistic features. Finally, these probabilistic features were combined and used for the construction of a meta-model based on a one-dimensional convolutional neural network. Experimental results indicated that DeepAR is a more accurate and stable approach for identifying AR antagonists in terms of the independent test dataset, by achieving an accuracy of 0.911 and MCC of 0.823. In addition, our proposed framework is able to provide feature importance information by leveraging a popular computational approach, named SHapley Additive exPlanations (SHAP). In the meanwhile, the characterization and analysis of potential AR antagonist candidates were achieved through the SHAP waterfall plot and molecular docking. The analysis inferred that N-heterocyclic moieties, halogenated substituents, and a cyano functional group were significant determinants of potential AR antagonists. Lastly, we implemented an online web server by using DeepAR (at http://pmlabstack.pythonanywhere.com/DeepAR ). We anticipate that DeepAR could be a useful computational tool for community-wide facilitation of AR candidates from a large number of uncharacterized compounds.

5.
Molecules ; 28(4)2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36838665

ABSTRACT

Cytochrome P450 17A1 (CYP17A1) is one of the key enzymes in steroidogenesis that produces dehydroepiandrosterone (DHEA) from cholesterol. Abnormal DHEA production may lead to the progression of severe diseases, such as prostatic and breast cancers. Thus, CYP17A1 is a druggable target for anti-cancer molecule development. In this study, cheminformatic analyses and quantitative structure-activity relationship (QSAR) modeling were applied on a set of 962 CYP17A1 inhibitors (i.e., consisting of 279 steroidal and 683 nonsteroidal inhibitors) compiled from the ChEMBL database. For steroidal inhibitors, a QSAR classification model built using the PubChem fingerprint along with the extra trees algorithm achieved the best performance, reflected by the accuracy values of 0.933, 0.818, and 0.833 for the training, cross-validation, and test sets, respectively. For nonsteroidal inhibitors, a systematic cheminformatic analysis was applied for exploring the chemical space, Murcko scaffolds, and structure-activity relationships (SARs) for visualizing distributions, patterns, and representative scaffolds for drug discoveries. Furthermore, seven total QSAR classification models were established based on the nonsteroidal scaffolds, and two activity cliff (AC) generators were identified. The best performing model out of these seven was model VIII, which is built upon the PubChem fingerprint along with the random forest algorithm. It achieved a robust accuracy across the training set, the cross-validation set, and the test set, i.e., 0.96, 0.92, and 0.913, respectively. It is anticipated that the results presented herein would be instrumental for further CYP17A1 inhibitor drug discovery efforts.


Subject(s)
Cheminformatics , Enzyme Inhibitors , Steroid 17-alpha-Hydroxylase , Dehydroepiandrosterone , Enzyme Inhibitors/pharmacology , Machine Learning , Quantitative Structure-Activity Relationship , Steroids/chemistry , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors
6.
ACS Omega ; 8(7): 6729-6742, 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36844574

ABSTRACT

Prostate cancer (PCa) is a major leading cause of mortality of cancer among males. There have been numerous studies to develop antagonists against androgen receptor (AR), a crucial therapeutic target for PCa. This study is a systematic cheminformatic analysis and machine learning modeling to study the chemical space, scaffolds, structure-activity relationship, and landscape of human AR antagonists. There are 1678 molecules as final data sets. Chemical space visualization by physicochemical property visualization has demonstrated that molecules from the potent/active class generally have a mildly smaller molecular weight (MW), octanol-water partition coefficient (log P), number of hydrogen-bond acceptors (nHA), number of rotatable bonds (nRot), and topological polar surface area (TPSA) than molecules from intermediate/inactive class. The chemical space visualization in the principal component analysis (PCA) plot shows significant overlapping distributions between potent/active class molecules and intermediate/inactive class molecules; potent/active class molecules are intensively distributed, while intermediate/inactive class molecules are widely and sparsely distributed. Murcko scaffold analysis has shown low scaffold diversity in general, and scaffold diversity of potent/active class molecules is even lower than intermediate/inactive class molecules, indicating the necessity for developing molecules with novel scaffolds. Furthermore, scaffold visualization has identified 16 representative Murcko scaffolds. Among them, scaffolds 1, 2, 3, 4, 7, 8, 10, 11, 15, and 16 are highly favorable scaffolds due to their high scaffold enrichment factor values. Based on scaffold analysis, their local structure-activity relationships (SARs) were investigated and summarized. In addition, the global SAR landscape was explored by quantitative structure-activity relationship (QSAR) modelings and structure-activity landscape visualization. A QSAR classification model incorporating all of the 1678 molecules stands out as the best model from a total of 12 candidate models for AR antagonists (built on PubChem fingerprint, extra trees algorithm, accuracy for training set: 0.935, 10-fold cross-validation set: 0.735 and test set: 0.756). Deeper insights into the structure-activity landscape highlighted a total of seven significant activity cliff (AC) generators (ChEMBL molecule IDs: 160257, 418198, 4082265, 348918, 390728, 4080698, and 6530), which provide valuable SAR information for medicinal chemistry. The findings in this study provide new insights and guidelines for hit identification and lead optimization for the development of novel AR antagonists.

7.
EXCLI J ; 21: 1331-1351, 2022.
Article in English | MEDLINE | ID: mdl-36540675

ABSTRACT

The emergence of New Delhi metallo-beta-lactamase-1 (NDM-1) has conferred enteric bacteria resistance to almost all beta-lactam antibiotics. Its capability of horizontal transfer through plasmids, amongst humans, animal reservoirs and the environment, has added up to the totality of antimicrobial resistance control, animal husbandry and food safety. Thus far, there have been no effective drugs for neutralizing NDM-1. This study explores the structure-activity relationship of NDM-1 inhibitors. IC50 values of NDM-1 inhibitors were compiled from both the ChEMBL database and literature. After curation, a final set of 686 inhibitors were used for machine learning model building using the random forest algorithm against 12 sets of molecular fingerprints. Benchmark results indicated that the KlekotaRothCount fingerprint provided the best overall performance with an accuracy of 0.978 and 0.778 for the training and testing set, respectively. Model interpretation revealed that nitrogen-containing features (KRFPC 4080, KRFPC 3882, KRFPC 677, KRFPC 3608, KRFPC 3750, KRFPC 4287 and KRFPC 3943), sulfur-containing substructures (KRFPC 2855 and KRFPC 4843), aromatic features (KRFPC 1566, KRFPC 1564, KRFPC 1642, KRFPC 3608, KRFPC 4287 and KRFPC 3943), carbonyl features (KRFPC 1193 and KRFPC 3025), aliphatic features (KRFPC 2975, KRFPC 297, KRFPC 3224 and KRFPC 669) are features contributing to NDM-1 inhibitory activity. It is anticipated that findings from this study would help facilitate the drug discovery of NDM-1 inhibitors by providing guidelines for further lead optimization.

8.
Sci Rep ; 12(1): 16435, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36180453

ABSTRACT

Progesterone receptors (PRs) are implicated in various cancers since their presence/absence can determine clinical outcomes. The overstimulation of progesterone can facilitate oncogenesis and thus, its modulation through PR inhibition is urgently needed. To address this issue, a novel stacked ensemble learning approach (termed StackPR) is presented for fast, accurate, and large-scale identification of PR antagonists using only SMILES notation without the need for 3D structural information. We employed six popular machine learning (ML) algorithms (i.e., logistic regression, partial least squares, k-nearest neighbor, support vector machine, extremely randomized trees, and random forest) coupled with twelve conventional molecular descriptors to create 72 baseline models. Then, a genetic algorithm in conjunction with the self-assessment-report approach was utilized to determine m out of the 72 baseline models as means of developing the final meta-predictor using the stacking strategy and tenfold cross-validation test. Experimental results on the independent test dataset show that StackPR achieved impressive predictive performance with an accuracy of 0.966 and Matthew's coefficient correlation of 0.925. In addition, analysis based on the SHapley Additive exPlanation algorithm and molecular docking indicates that aliphatic hydrocarbons and nitrogen-containing substructures were the most important features for having PR antagonist activity. Finally, we implemented an online webserver using StackPR, which is freely accessible at http://pmlabstack.pythonanywhere.com/StackPR . StackPR is anticipated to be a powerful computational tool for the large-scale identification of unknown PR antagonist candidates for follow-up experimental validation.


Subject(s)
Progesterone , Receptors, Progesterone , Algorithms , Computational Biology , Molecular Docking Simulation , Nitrogen , Support Vector Machine
9.
Curr Pharm Des ; 27(18): 2180-2188, 2021.
Article in English | MEDLINE | ID: mdl-33138759

ABSTRACT

In light of the growing resistance toward current antiviral drugs, efforts to discover novel and effective antiviral therapeutic agents remain a pressing scientific effort. Antiviral peptides (AVPs) represent promising therapeutic agents due to their extraordinary advantages in terms of potency, efficacy and pharmacokinetic properties. The growing volume of newly discovered peptide sequences in the post-genomic era requires computational approaches for timely and accurate identification of AVPs. Machine learning (ML) methods such as random forest and support vector machine represent robust learning algorithms that are instrumental in successful peptide-based drug discovery. Therefore, this review summarizes the current state-of-the-art application of ML methods for identifying AVPs directly from the sequence information. We compare the efficiency of these methods in terms of the underlying characteristics of the dataset used along with feature encoding methods, ML algorithms, cross-validation methods and prediction performance. Finally, guidelines for the development of robust AVP models are also discussed. It is anticipated that this review will serve as a useful guide for the design and development of robust AVP and related therapeutic peptide predictors in the future.


Subject(s)
Antiviral Agents , Peptides , Algorithms , Antiviral Agents/pharmacology , Computer Simulation , Humans , Machine Learning
10.
Bioorg Chem ; 79: 171-178, 2018 09.
Article in English | MEDLINE | ID: mdl-29758407

ABSTRACT

A three series of thioureas, monothiourea type I (4a-g), 1,4-bisthiourea type II (5a-h) and 1,3-bisthiourea type III (6a-h) were synthesized. Their aromatase inhibitory activities have been evaluated. Interestingly, eight thiourea derivatives (4e, 5f-h, 6d, 6f-h) exhibited the aromatase inhibitory activities with IC50 range of 0.6-10.2 µM. The meta-bisthiourea bearing 4-NO2 group (6f) and 3,5-diCF3 groups (6h) were shown to be the most potent compounds with sub-micromolar IC50 values of 0.8 and 0.6 µM, respectively. Molecular docking also revealed that one of the thiourea moieties of these two compounds could mimic steroidal backbone of the natural androstenedione (ASD) via hydrophobic interactions with enzyme residues (Val370, Leu477, Thr310, and Phe221 for 6f, Val370, Leu477, Ser478, and Ile133 for 6h). This is the first time that the bisthioureas have been reported for their potential to be developed as aromatase inhibitors, in which the 4-NO2 and 3,5-diCF3 analogs have been highlighted as promising candidates.


Subject(s)
Aromatase Inhibitors/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aromatase/chemistry , Aromatase Inhibitors/chemical synthesis , Aromatase Inhibitors/chemistry , Binding Sites , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Molecular Docking Simulation , Molecular Structure , Thiourea/chemical synthesis
11.
RSC Adv ; 8(11): 5920-5935, 2018 Feb 02.
Article in English | MEDLINE | ID: mdl-35539618

ABSTRACT

Sickle cell disease (SCD), an autosomal recessive genetic disorder, has been recognized by the World Health Organization (WHO) as a major public health problem as it affects 300 000 individuals worldwide. Complications arising from SCD include anemia, microvascular occlusion, severe pain, stokes, renal dysfunction and infections. A lucrative therapeutic strategy is to employ anti-sickling agents that can disrupt the formation of the HbS polymer. This study therefore employed cheminformatic approaches, encompassing classification structure-activity relationship (CSAR) modeling, to deduce the privileged substructures giving rise to the anti-sickling activity of an investigated set of 115 compounds, followed by substructure analysis. Briefly, the compiled compounds were described by fingerprint descriptors and used in the construction of CSAR models via several machine learning algorithms. The modelability of the data set, as exemplified by the MODI index, was determined to be in the range of 0.70-0.84. The predictive performance was deduced by the accuracy, sensitivity, specificity and Matthews correlation coefficient, which was found to be statistically robust, whereby the former three parameters afforded values in excess of 0.7 while the latter statistical parameter provided a value greater than 0.5. An analysis of the top 20 important substructure descriptors for anti-sickling activity revealed that 10 important features were significant in the differentiation of actives from inactives, as illustrated by aromaticity/conjugation (e.g. SubFPC287, SubFPC171 and SubFPC5), carbonyl groups (e.g. SubFPC137, SubFPC139, SubFPC49 and SubFPC135) and miscellaneous groups (e.g. SubFPC303, SubFPC302 and SubFPC275). Furthermore, an analysis of the structure-activity relationship revealed that the length of alkyl chains, choice of functional moiety and position of substitution on the benzene ring may affect the anti-sickling activity of these compounds. Thus, this knowledge is anticipated to be useful for guiding the design of robust compounds against the gelling activity of HbS, as preliminarily demonstrated in the data-driven compound design presented herein.

12.
RSC Adv ; 8(15): 8233, 2018 Feb 19.
Article in English | MEDLINE | ID: mdl-35543880

ABSTRACT

[This corrects the article DOI: 10.1039/C7RA12079F.].

13.
PeerJ ; 4: e2322, 2016.
Article in English | MEDLINE | ID: mdl-27602288

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disease which leads to the gradual loss of neuronal cells. Several hypotheses for AD exists (e.g., cholinergic, amyloid, tau hypotheses, etc.). As per the cholinergic hypothesis, the deficiency of choline is responsible for AD; therefore, the inhibition of AChE is a lucrative therapeutic strategy for the treatment of AD. Acetylcholinesterase (AChE) is an enzyme that catalyzes the breakdown of the neurotransmitter acetylcholine that is essential for cognition and memory. A large non-redundant data set of 2,570 compounds with reported IC50 values against AChE was obtained from ChEMBL and employed in quantitative structure-activity relationship (QSAR) study so as to gain insights on their origin of bioactivity. AChE inhibitors were described by a set of 12 fingerprint descriptors and predictive models were constructed from 100 different data splits using random forest. Generated models afforded R (2), [Formula: see text] and [Formula: see text] values in ranges of 0.66-0.93, 0.55-0.79 and 0.56-0.81 for the training set, 10-fold cross-validated set and external set, respectively. The best model built using the substructure count was selected according to the OECD guidelines and it afforded R (2), [Formula: see text] and [Formula: see text] values of 0.92 ± 0.01, 0.78 ± 0.06 and 0.78 ± 0.05, respectively. Furthermore, Y-scrambling was applied to evaluate the possibility of chance correlation of the predictive model. Subsequently, a thorough analysis of the substructure fingerprint count was conducted to provide informative insights on the inhibitory activity of AChE inhibitors. Moreover, Kennard-Stone sampling of the actives were applied to select 30 diverse compounds for further molecular docking studies in order to gain structural insights on the origin of AChE inhibition. Site-moiety mapping of compounds from the diversity set revealed three binding anchors encompassing both hydrogen bonding and van der Waals interaction. Molecular docking revealed that compounds 13, 5 and 28 exhibited the lowest binding energies of -12.2, -12.0 and -12.0 kcal/mol, respectively, against human AChE, which is modulated by hydrogen bonding, π-π stacking and hydrophobic interaction inside the binding pocket. These information may be used as guidelines for the design of novel and robust AChE inhibitors.

14.
Int J Mol Sci ; 17(7)2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27376281

ABSTRACT

Host defense peptides (HDPs) are positively-charged and amphipathic components of the innate immune system that have demonstrated great potential to become the next generation of broad spectrum therapeutic agents effective against a vast array of pathogens and tumor. As such, many approaches have been taken to improve the therapeutic efficacy of HDPs. Amongst these methods, the incorporation of d-amino acids (d-AA) is an approach that has demonstrated consistent success in improving HDPs. Although, virtually all HDP review articles briefly mentioned about the role of d-AA, however it is rather surprising that no systematic review specifically dedicated to this topic exists. Given the impact that d-AA incorporation has on HDPs, this review aims to fill that void with a systematic discussion of the impact of d-AA on HDPs.


Subject(s)
Amino Acids/metabolism , Peptides/metabolism , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Bacteria/drug effects , Gramicidin/chemistry , Gramicidin/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Structure, Secondary
15.
PeerJ ; 4: e1979, 2016.
Article in English | MEDLINE | ID: mdl-27190705

ABSTRACT

Aromatase, the rate-limiting enzyme that catalyzes the conversion of androgen to estrogen, plays an essential role in the development of estrogen-dependent breast cancer. Side effects due to aromatase inhibitors (AIs) necessitate the pursuit of novel inhibitor candidates with high selectivity, lower toxicity and increased potency. Designing a novel therapeutic agent against aromatase could be achieved computationally by means of ligand-based and structure-based methods. For over a decade, we have utilized both approaches to design potential AIs for which quantitative structure-activity relationships and molecular docking were used to explore inhibitory mechanisms of AIs towards aromatase. However, such approaches do not consider the effects that aromatase variants have on different AIs. In this study, proteochemometrics modeling was applied to analyze the interaction space between AIs and aromatase variants as a function of their substructural and amino acid features. Good predictive performance was achieved, as rigorously verified by 10-fold cross-validation, external validation, leave-one-compound-out cross-validation, leave-one-protein-out cross-validation and Y-scrambling tests. The investigations presented herein provide important insights into the mechanisms of aromatase inhibitory activity that could aid in the design of novel potent AIs as breast cancer therapeutic agents.

16.
PeerJ ; 4: e1958, 2016.
Article in English | MEDLINE | ID: mdl-27114890

ABSTRACT

The fight against the emergence of mutant influenza strains has led to the screening of an increasing number of compounds for inhibitory activity against influenza neuraminidase. This study explores the chemical space of neuraminidase inhibitors (NAIs), which provides an opportunity to obtain further molecular insights regarding the underlying basis of their bioactivity. In particular, a large set of 347 and 175 NAIs against influenza A and B, respectively, was compiled from the literature. Molecular and quantum chemical descriptors were obtained from low-energy conformational structures geometrically optimized at the PM6 level. The bioactivities of NAIs were classified as active or inactive according to their half maximum inhibitory concentration (IC50) value in which IC50 < 1µM and ≥ 10µM were defined as active and inactive compounds, respectively. Interpretable decision rules were derived from a quantitative structure-activity relationship (QSAR) model established using a set of substructure descriptors via decision tree analysis. Univariate analysis, feature importance analysis from decision tree modeling and molecular scaffold analysis were performed on both data sets for discriminating important structural features amongst active and inactive NAIs. Good predictive performance was achieved as deduced from accuracy and Matthews correlation coefficient values in excess of 81% and 0.58, respectively, for both influenza A and B NAIs. Furthermore, molecular docking was employed to investigate the binding modes and their moiety preferences of active NAIs against both influenza A and B neuraminidases. Moreover, novel NAIs with robust binding fitness towards influenza A and B neuraminidase were generated via combinatorial library enumeration and their binding fitness was on par or better than FDA-approved drugs. The results from this study are anticipated to be beneficial for guiding the rational drug design of novel NAIs for treating influenza infections.

17.
J Cheminform ; 8: 72, 2016.
Article in English | MEDLINE | ID: mdl-28053671

ABSTRACT

BACKGROUND: Currently, monomeric fluorescent proteins (FP) are ideal markers for protein tagging. The prediction of oligomeric states is helpful for enhancing live biomedical imaging. Computational prediction of FP oligomeric states can accelerate the effort of protein engineering efforts of creating monomeric FPs. To the best of our knowledge, this study represents the first computational model for predicting and analyzing FP oligomerization directly from the amino acid sequence. RESULTS: After data curation, an exhaustive data set consisting of 397 non-redundant FP oligomeric states was compiled from the literature. Results from benchmarking of the protein descriptors revealed that the model built with amino acid composition descriptors was the top performing model with accuracy, sensitivity and specificity in excess of 80% and MCC greater than 0.6 for all three data subsets (e.g. training, tenfold cross-validation and external sets). The model provided insights on the important residues governing the oligomerization of FP. To maximize the benefit of the generated predictive model, it was implemented as a web server under the R programming environment. CONCLUSION: osFP affords a user-friendly interface that can be used to predict the oligomeric state of FP using the protein sequence. The advantage of osFP is that it is platform-independent meaning that it can be accessed via a web browser on any operating system and device. osFP is freely accessible at http://codes.bio/osfp/ while the source code and data set is provided on GitHub at https://github.com/chaninn/osFP/.Graphical Abstract.

18.
Springerplus ; 4: 571, 2015.
Article in English | MEDLINE | ID: mdl-26543706

ABSTRACT

Considerable attention has been given on the search for novel anticancer drugs with respect to the disease sequelae on human health and well-being. Triazole is considered to be an attractive scaffold possessing diverse biological activities. Structural modification on the privileged structures is noted as an effective strategy towards successful design and development of novel drugs. The quantitative structure-activity relationships (QSAR) is well-known as a powerful computational tool to facilitate the discovery of potential compounds. In this study, a series of thirty-two 1,2,3-triazole derivatives (1-32) together with their experimentally measured cytotoxic activities against four cancer cell lines i.e., HuCCA-1, HepG2, A549 and MOLT-3 were used for QSAR analysis. Four QSAR models were successfully constructed with acceptable predictive performance affording R CV ranging from 0.5958 to 0.8957 and RMSECV ranging from 0.2070 to 0.4526. An additional set of 64 structurally modified triazole compounds (1A-1R, 2A-2R, 7A-7R and 8A-8R) were constructed in silico and their predicted cytotoxic activities were obtained using the constructed QSAR models. The study suggested crucial moieties and certain properties essential for potent anticancer activity and highlighted a series of promising compounds (21, 28, 32, 1P, 8G, 8N and 8Q) for further development as novel triazole-based anticancer agents.

19.
Drug Des Devel Ther ; 9: 4515-49, 2015.
Article in English | MEDLINE | ID: mdl-26309399

ABSTRACT

This study represents the first large-scale study on the chemical space of inhibitors of dipeptidyl peptidase-4 (DPP4), which is a potential therapeutic protein target for the treatment of diabetes mellitus. Herein, a large set of 2,937 compounds evaluated for their ability to inhibit DPP4 was compiled from the literature. Molecular descriptors were generated from the geometrically optimized low-energy conformers of these compounds at the semiempirical AM1 level. The origins of DPP4 inhibitory activity were elucidated from computed molecular descriptors that accounted for the unique physicochemical properties inherently present in the active and inactive sets of compounds as defined by their respective half maximal inhibitory concentration values of less than 1 µM and greater than 10 µM, respectively. Decision tree analysis revealed the importance of molecular weight, total energy of a molecule, topological polar surface area, lowest unoccupied molecular orbital, and number of hydrogen-bond donors, which correspond to molecular size, energy, surface polarity, electron acceptors, and hydrogen bond donors, respectively. The prediction model was subjected to rigorous independent testing via three external sets. Scaffold and chemical fragment analysis was also performed on these active and inactive sets of compounds to shed light on the distinguishing features of the functional moieties. Docking of representative active DPP4 inhibitors was also performed to unravel key interacting residues. The results of this study are anticipated to be useful in guiding the rational design of novel and robust DPP4 inhibitors for the treatment of diabetes.


Subject(s)
Computer-Aided Design , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Design , Binding Sites , Decision Trees , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/metabolism , Dose-Response Relationship, Drug , Humans , Hydrogen Bonding , Molecular Docking Simulation , Molecular Weight , Principal Component Analysis , Protein Binding , Protein Conformation , Quantitative Structure-Activity Relationship , Surface Properties , Workflow
20.
Curr Top Med Chem ; 15(18): 1814-26, 2015.
Article in English | MEDLINE | ID: mdl-25961524

ABSTRACT

Inhibition of non-structural protein 5B (NS5B) represents an attractive strategy for the therapeutic treatment of hepatitis C virus (HCV). In this study, machine learning classifiers such as artificial neural network (ANN), support vector machine (SVM), random forest (RF) and decision tree (DT) analyses were used to classify 970 compounds based on their physicochemical properties, including quantum chemical descriptors, constitutional descriptors, functional groups and molecular properties. Good predictive performance was obtained from all classifiers, providing accuracies ranging from 82.47-89.61% for external validation set. SVM was noted as the best classifier, indicated by its highest accuracy of 89.61%. The analyses were performed on data sets stratified by structural scaffolds (nucleoside and non-nucleoside) and bioactivities (active and inactive properties). In addition, a molecular fragment analysis was performed to investigate molecular substructures corresponding to biological activities. Furthermore, common substructures and potential functional groups governing the activities of active and inactive inhibitors were noted for the benefit of rational design and high-throughput screening towards potential HCV NS5B inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Machine Learning , Viral Nonstructural Proteins/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Molecular Structure , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...