[go: up one dir, main page]

 
 
ijms-logo

Journal Browser

Journal Browser

Big Data Analysis and QSAR/QSPR Research in Chemistry, Bio-Medical, and Network Sciences

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Physical Chemistry, Theoretical and Computational Chemistry".

Deadline for manuscript submissions: closed (30 April 2016) | Viewed by 152605

Special Issue Editors


E-Mail Website1 Website2
Guest Editor
Chair and Director of Department of Computer Science and Information Technology, University of Coruña (UDC), Coruña, Spain
Interests: big data analysis; machine learning, artificial intelligence; bioinformatics; cheminformatics
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Milano Chemometrics and QSAR Research Group, Department of Environmental Sciences, University of Milano-Bicocca, 20126 Milano, Italy
Interests: chemometric; QSAR/QSPR; multi-criteria decision making; molecular descriptors; software development
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Co-Guest Editor
Department of Organic Chemistry II, University of the Basque Country (UPV/EHU), Leioa, Sarriena w/n, Bizkaia
Interests: organic chemistry; organic synthesis; chemical catalysis; computational chemistry

Special Issue Information

Dear Colleagues,

There is a steady increasing necessity of multidisciplinary collaborations in molecular science between experimentalists and theoretical scientists, as well as among theoretical scientists from different fields. One of the more important forces driving this necessity is the accumulation of large amounts of data as results of important advances in Chemometrics and Molecular Sciences Experimental Techniques of data acquisition in general.

In this context, we decided to create[MD1] a new scientific conference to promote the scientific synergies expressed earlier. MOL2NET (the conference's running title) will be held from 15–30 December, 2015, on the SciForum platform. The official website of the conference is: http://sciforum.net/conference/mol2net-1. Represented disciplines will encompass the molecular and biomedical sciences, social networks analysis, and beyond. More specifically, this conference aims to promote scientific synergies between groups of experimental molecular and bio-medical scientists. Relevant fields include chemistry, pharmacology, cancer research, proteomics, the neurosciences, the nanosciences, and epidemiology. Moreover, the conference welcomes computational and social sciences experts from different areas, such as computational chemistry, bioinformatics, social networks analysis, big data predictive analytics, biostatistics, etc. The full title of this conference is the 1st International Conference on Synergies of Experimental Groups of Molecular and Biomedical Sciences with Data, Networks, and Social Sciences Experts. The conference per se is the result of the synergy between the Department of Organic Chemistry, University of Basque Country (UPV/EHU), and IKERBASQUE, Basque Foundation for Sciences, with the Faculty of Informatics, University of Coruña (UDC).

In order to strengthen and spread the outputs of MOL2NET, we decided to be the Guest Editors for one Special Issue. In consonance with the conference, the topic of the issue is: Big Data Analysis and QSAR/QSPR Research in Chemistry, Bio-Medical, and Network Sciences. The issue is focused on the development and application of different theoretical algorithms combining Chemoinformatics, Computational Chemistry, Bioinformatics, Data Analysis, and Network Science methods. Submissions of other authors that do not attend the conference are also welcome. Accepted papers will be published in the International Journal of Molecular Science (IJMS), which is an open access publication journal of MDPI, in the field of Molecular and Biomedical Sciences (https://www.mdpi.com/journal/ijms).

Prof. Dr. Humberto González-Díaz
(IKERBASQUE Senior Professor)
Prof. Dr. Alejandro Pazos Sierra
Prof. Dr. Roberto Todeschini
Guest Editors

Dr. Sonia Arrasate Gil
Co-Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Big Data Analysis in Chemometrics
  • QSPR Chemoinformatics models of Chemical Reactivity
  • Computer-Aided Drug Discovery (CADD) with QSAR models
  • DNA/Protein Quantitative Sequence-Activity Models (QSAM) in Bioinformatics
  • Structure-Property Relationships Analysis of Bio-Molecular Networks
  • Prediction of Drug-Target Interaction Networks
  • Computational Proteomics and Metabolomics
  • Protein Interaction Networks
  • Machine Learning in Cheminformatics

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (19 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

172 KiB  
Editorial
Data Analysis in Chemistry and Bio-Medical Sciences
by Roberto Todeschini, Alejandro Pazos, Sonia Arrasate and Humberto González-Díaz
Int. J. Mol. Sci. 2016, 17(12), 2105; https://doi.org/10.3390/ijms17122105 - 14 Dec 2016
Cited by 2 | Viewed by 4530
Abstract
There is an increasing necessity for multidisciplinary collaborations in molecular science between experimentalists and theoretical scientists, as well as among theoretical scientists from different fields.[...] Full article

Research

Jump to: Editorial, Review

717 KiB  
Article
Conformation-Independent QSPR Approach for the Soil Sorption Coefficient of Heterogeneous Compounds
by José F. Aranda, Juan C. Garro Martinez, Eduardo A. Castro and Pablo R. Duchowicz
Int. J. Mol. Sci. 2016, 17(8), 1247; https://doi.org/10.3390/ijms17081247 - 3 Aug 2016
Cited by 21 | Viewed by 4658
Abstract
We predict the soil sorption coefficient for a heterogeneous set of 643 organic non-ionic compounds by means of Quantitative Structure-Property Relationships (QSPR). A conformation-independent representation of the chemical structure is established. The 17,538 molecular descriptors derived with PaDEL and EPI Suite softwares are [...] Read more.
We predict the soil sorption coefficient for a heterogeneous set of 643 organic non-ionic compounds by means of Quantitative Structure-Property Relationships (QSPR). A conformation-independent representation of the chemical structure is established. The 17,538 molecular descriptors derived with PaDEL and EPI Suite softwares are simultaneously analyzed through linear regressions obtained with the Replacement Method variable subset selection technique. The best predictive three-descriptors QSPR is developed on a reduced training set of 93 chemicals, having an acceptable predictive capability on 550 test set compounds. We also establish a model with a single optimal descriptor derived from CORAL freeware. The present approach compares fairly well with a previously reported one that uses Dragon descriptors. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Predicted and experimental <math display="inline"> <semantics> <mrow> <mi>log</mi> <msub> <mi>K</mi> <mrow> <mi>o</mi> <mi>c</mi> </mrow> </msub> </mrow> </semantics> </math> values according to QSPR based on Equation (1).</p>
Full article ">Figure 2
<p>Predicted and experimental <math display="inline"> <semantics> <mrow> <mi>log</mi> <msub> <mi>K</mi> <mrow> <mi>o</mi> <mi>c</mi> </mrow> </msub> </mrow> </semantics> </math> values according to QSPR based on Equation (3).</p>
Full article ">
527 KiB  
Article
A Machine Learning Approach for Hot-Spot Detection at Protein-Protein Interfaces
by Rita Melo, Robert Fieldhouse, André Melo, João D. G. Correia, Maria Natália D. S. Cordeiro, Zeynep H. Gümüş, Joaquim Costa, Alexandre M. J. J. Bonvin and Irina S. Moreira
Int. J. Mol. Sci. 2016, 17(8), 1215; https://doi.org/10.3390/ijms17081215 - 27 Jul 2016
Cited by 45 | Viewed by 10079
Abstract
Understanding protein-protein interactions is a key challenge in biochemistry. In this work, we describe a more accurate methodology to predict Hot-Spots (HS) in protein-protein interfaces from their native complex structure compared to previous published Machine Learning (ML) techniques. Our model is trained on [...] Read more.
Understanding protein-protein interactions is a key challenge in biochemistry. In this work, we describe a more accurate methodology to predict Hot-Spots (HS) in protein-protein interfaces from their native complex structure compared to previous published Machine Learning (ML) techniques. Our model is trained on a large number of complexes and on a significantly larger number of different structural- and evolutionary sequence-based features. In particular, we added interface size, type of interaction between residues at the interface of the complex, number of different types of residues at the interface and the Position-Specific Scoring Matrix (PSSM), for a total of 79 features. We used twenty-seven algorithms from a simple linear-based function to support-vector machine models with different cost functions. The best model was achieved by the use of the conditional inference random forest (c-forest) algorithm with a dataset pre-processed by the normalization of features and with up-sampling of the minor class. The method has an overall accuracy of 0.80, an F1-score of 0.73, a sensitivity of 0.76 and a specificity of 0.82 for the independent test set. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The flowchart of the current work.</p>
Full article ">Figure 2
<p>Top 15 variables for the c-forest method. SASA, Solvent Accessible Surface Area; #, Number of residues</p>
Full article ">
927 KiB  
Article
Molecular Rearrangement of an Aza-Scorpiand Macrocycle Induced by pH: A Computational Study
by Jesus Vicente De Julián-Ortiz, Begoña Verdejo, Víctor Polo, Emili Besalú and Enrique García-España
Int. J. Mol. Sci. 2016, 17(7), 1131; https://doi.org/10.3390/ijms17071131 - 14 Jul 2016
Cited by 5 | Viewed by 4304
Abstract
Rearrangements and their control are a hot topic in supramolecular chemistry due to the possibilities that these phenomena open in the design of synthetic receptors and molecular machines. Macrocycle aza-scorpiands constitute an interesting system that can reorganize their spatial structure depending on pH [...] Read more.
Rearrangements and their control are a hot topic in supramolecular chemistry due to the possibilities that these phenomena open in the design of synthetic receptors and molecular machines. Macrocycle aza-scorpiands constitute an interesting system that can reorganize their spatial structure depending on pH variations or the presence of metal cations. In this study, the relative stabilities of these conformations were predicted computationally by semi-empirical and density functional theory approximations, and the reorganization from closed to open conformations was simulated by using the Monte Carlo multiple minimum method. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Conformations closed (<b>A</b>) and open (<b>B</b>) of the aza-scorpiand macrocycle studied.</p>
Full article ">Figure 2
<p>Distance between NH and N, which proves the presence of the H–bond in the monoprotonated <b>A</b>.</p>
Full article ">Figure 3
<p>Labels for the dihedral angles in the pendant arm.</p>
Full article ">Figure 4
<p>(<b>A</b>,<b>B</b>) Conformations from X-ray minimized in the periodic box. <b>1</b>–<b>5</b>, conformations obtained from MCMM in vacuum and further minimization in standard water density-constant TI3P box until their respective local minima, for different runs.</p>
Full article ">
1745 KiB  
Article
Molecular Dynamics Simulation Study of the Selectivity of a Silica Polymer for Ibuprofen
by Riccardo Concu and M. Natalia D. S. Cordeiro
Int. J. Mol. Sci. 2016, 17(7), 1083; https://doi.org/10.3390/ijms17071083 - 7 Jul 2016
Cited by 7 | Viewed by 6038
Abstract
In the past few years, the sol-gel polycondensation technique has been increasingly employed with great success as an alternative approach to the preparation of molecularly imprinted materials (MIMs). The main aim of this study was to study, through a series of molecular dynamics [...] Read more.
In the past few years, the sol-gel polycondensation technique has been increasingly employed with great success as an alternative approach to the preparation of molecularly imprinted materials (MIMs). The main aim of this study was to study, through a series of molecular dynamics (MD) simulations, the selectivity of an imprinted silica xerogel towards a new template—the (±)-2-(P-Isobutylphenyl) propionic acid (Ibuprofen, IBU). We have previously demonstrated the affinity of this silica xerogel toward a similar molecule. In the present study, we simulated the imprinting process occurring in a sol-gel mixture using the Optimized Potentials for Liquid Simulations-All Atom (OPLS-AA) force field, in order to evaluate the selectivity of this xerogel for a template molecule. In addition, for the first time, we have developed and verified a new parameterisation for the Ibuprofen® based on the OPLS-AA framework. To evaluate the selectivity of the polymer, we have employed both the radial distribution functions, interaction energies and cluster analyses. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of the simulated molecules. <b>A</b> = Ibuprofen, IBU; <b>B</b> = Dehidroimidazolium motif modified with silica trimers, ORMOSIL, DHI<sup>+</sup>; <b>C</b> = Cyclic silica trimer, SI3; <b>D</b> = Anionic form of the cyclic silica trimer, SI<sup>−</sup>. The circles show the atoms considered in the calculation of the RDF.</p>
Full article ">Figure 2
<p>Radial distribution functions (RDF) analysis using the H atom of DHI for the pairs IBU-DHI, DHI-SI3, IBU-SI<sup>−</sup>. (<b>A</b>–<b>C</b>) represent the RDF of each MD replica.</p>
Full article ">Figure 3
<p>RDF analysis using the C atom of DHI for the pairs IBU-DHI, DHI-SI3, IBU-SI<sup>−</sup>. (<b>A</b>–<b>C</b>) represent the RDF of each MD replica.</p>
Full article ">Figure 4
<p>Cluster analysis of the three MD replicas.</p>
Full article ">Figure 5
<p>MSD of the molecular species during the simulation. (<b>A</b>) MSD of all the compounds present in the mixture; (<b>B</b>) MSD of IBU, DHI, SI3 and SI<sup>−</sup>.</p>
Full article ">
201 KiB  
Article
Prognostic Value of Affective Symptoms in First-Admission Psychotic Patients
by Marta Arrasate, Itxaso González-Ortega, Adriana García-Alocén, Susana Alberich, Iñaki Zorrilla and Ana González-Pinto
Int. J. Mol. Sci. 2016, 17(7), 1039; https://doi.org/10.3390/ijms17071039 - 30 Jun 2016
Cited by 16 | Viewed by 5161
Abstract
Background: Very little research has been conducted in patients with first-episode psychosis using a dimensional approach. Affective dimensional representations might be useful to predict the clinical course and treatment needs in such patients. Methods: Weincluded 112 patients with first-episode psychosis in a longitudinal-prospective [...] Read more.
Background: Very little research has been conducted in patients with first-episode psychosis using a dimensional approach. Affective dimensional representations might be useful to predict the clinical course and treatment needs in such patients. Methods: Weincluded 112 patients with first-episode psychosis in a longitudinal-prospective study with a five-year follow-up (N = 82). Logistic analyses were performed to determine the predictive factors associated with depressive, manic, activation, and dysphoric dimensions. Results: High scores on the depressive dimension were associated with the best prognosis. On the other hand, high scores on the activation dimension and the manic dimension were associated with a poorer prognosis in terms of relapses. Only the dysphoric dimension was not associated with syndromic or functional prognosis. Conclusion: Ourresults suggest that the pattern of baseline affective symptoms helps to predict the course of psychotic illness. Therefore, the systematic assessment of affective symptoms would enable us to draw important conclusions regarding patients’ prognosis. Interventions for patients with high scores on manic or activation dimensions could be beneficial in decreasing relapses in first-episode psychosis. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">
5012 KiB  
Article
Genome-Wide Discriminatory Information Patterns of Cytosine DNA Methylation
by Robersy Sanchez and Sally A. Mackenzie
Int. J. Mol. Sci. 2016, 17(6), 938; https://doi.org/10.3390/ijms17060938 - 17 Jun 2016
Cited by 11 | Viewed by 6187
Abstract
Cytosine DNA methylation (CDM) is a highly abundant, heritable but reversible chemical modification to the genome. Herein, a machine learning approach was applied to analyze the accumulation of epigenetic marks in methylomes of 152 ecotypes and 85 silencing mutants of Arabidopsis thaliana. [...] Read more.
Cytosine DNA methylation (CDM) is a highly abundant, heritable but reversible chemical modification to the genome. Herein, a machine learning approach was applied to analyze the accumulation of epigenetic marks in methylomes of 152 ecotypes and 85 silencing mutants of Arabidopsis thaliana. In an information-thermodynamics framework, two measurements were used: (1) the amount of information gained/lost with the CDM changes I R and (2) the uncertainty of not observing a SNP L C R . We hypothesize that epigenetic marks are chromosomal footprints accounting for different ontogenetic and phylogenetic histories of individual populations. A machine learning approach is proposed to verify this hypothesis. Results support the hypothesis by the existence of discriminatory information (DI) patterns of CDM able to discriminate between individuals and between individual subpopulations. The statistical analyses revealed a strong association between the topologies of the structured population of Arabidopsis ecotypes based on I R and on LCR, respectively. A statistical-physical relationship between I R and L C R was also found. Results to date imply that the genome-wide distribution of CDM changes is not only part of the biological signal created by the methylation regulatory machinery, but ensures the stability of the DNA molecule, preserving the integrity of the genetic message under continuous stress from thermal fluctuations in the cell environment. Full article
Show Figures

Figure 1

Figure 1
<p>Methylation hotspots along chromosome 5 from 151 <span class="html-italic">Arabidopsis</span> <span class="html-italic">thaliana</span> ecotypes [<a href="#B14-ijms-17-00938" class="html-bibr">14</a>] (CG methylation context). The color bar indicates the magnitude of <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> values.</p>
Full article ">Figure 2
<p>Illustrative heatmap showing the classification of GRs into hypervariable (HMRs), variable (VMRs) and low-variable or constant (LMRs) methylated regions. (<b>A</b>) The maximum and minimum of the <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> values correspond to black and sky blue, respectively; (<b>B</b>) The same samples, but with inverted color scale, equivalent to the photograph negative; the maximum and minimum correspond to sky blue and black, respectively. The heatmap for all the ecotype samples is given in <a href="#app1-ijms-17-00938" class="html-app">Figure S16</a>. In general, HMRs are regions with <math display="inline"> <semantics> <mrow> <mrow> <mo>|</mo> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> <mo>|</mo> </mrow> <mo>&gt;</mo> <mo>&gt;</mo> <mn>0</mn> </mrow> </semantics> </math> . In both panels, A and B, the HMRs readily visible are those straight lines in orange to black colors. In panel A, HMRs are GRs with <math display="inline"> <semantics> <mrow> <mrow> <mo>|</mo> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> <mo>|</mo> </mrow> <mo>&gt;</mo> <mo>&gt;</mo> <mn>0</mn> </mrow> </semantics> </math> and in panel B are those GRs with <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>&lt;</mo> <mo>&lt;</mo> <mn>0</mn> </mrow> </semantics> </math> . In both panels, the arrows in red, green, and sky blue indicate that at least one HMR is found in the observed heatmap position. The arrows in orange and light green indicate that at least one VMR is found in the specified heatmap position, while arrows in yellow indicate that at least one LMR is present. It must be noticed that LMRs are the most abundant types of GRs. The apparent abundance of HMRs results from the compression of sample vectors for 13,370 GRs. As a result, some GRs are superimposed in the graphic. In the present example, only 12,971 from 13,370 × 9 = 120,330 GRs (11% ) have <math display="inline"> <semantics> <mrow> <mrow> <mo>|</mo> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> <mo>|</mo> </mrow> <mo>≥</mo> <mn>10</mn> </mrow> </semantics> </math> bit. A quantitative way to define the borders of each class can be set by applying fuzzy set and fuzzy logic theory, beyond the limit of the current work.</p>
Full article ">Figure 3
<p>Methylation hotspots along chromosome 5 from 83 <span class="html-italic">Arabidopsis</span> silencing mutants in CG context. The color bar indicates the magnitude of <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> values (Equation (1), Material and Methods).</p>
Full article ">Figure 4
<p>Annotation of several CHG methylation hotspots on chromosome 2 from eight <span class="html-italic">Arabidopsis</span> silencing mutants.</p>
Full article ">Figure 5
<p>Mutational hotspots along chromosome 5 from 83 <span class="html-italic">Arabidopsis</span> silencing mutants. The color bar indicates the magnitude of <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> values (Equation (3), Material and Methods).</p>
Full article ">Figure 6
<p>Classification of the <span class="html-italic">Arabidopsis</span> ecotypes according to their geographical distribution. (<b>A</b>,<b>B</b>) LDAs based on <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> (SNPs), respectively; (<b>C</b>,<b>D</b>) fan dendrograms based on the individual coordinates estimated from the linear discriminant (LD) functions. The dendrograms were built by applying hierarchical clustering with Euclidean distance and UPGMA as agglomeration method.</p>
Full article ">Figure 7
<p>Dependence between variables <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> in the ecotypes La-0 and Fr.2. (<b>A</b>) The 2D kernel density plots <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> <span class="html-italic">versus</span> <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> indicate that most <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> values are located in a narrow band around the vertical line <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>=</mo> <mn>0</mn> </mrow> </semantics> </math> . That is, the density plots expose a statistical tendency: most of the GRs with lower uncertainty variations (lower methylation changes) also experience, in accordance with Equation (5), a lower uncertainty level (SNP not observed), determined by a lower probability that an SNP is present within a GR; (<b>B</b>) The 3D kernel density plot indicates that, for example, with high joint probability <math display="inline"> <semantics> <mrow> <mi>P</mi> <mo> </mo> <mo> </mo> <mrow> <mo>(</mo> <mrow> <mo>−</mo> <mn>1</mn> <mo>≤</mo> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>1</mn> <mo>,</mo> <mo> </mo> <mo> </mo> <mn>0</mn> <mo>≤</mo> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>25</mn> </mrow> <mo>)</mo> </mrow> </mrow> </semantics> </math> (the volume of the prism with squared base formed by the intervals <math display="inline"> <semantics> <mrow> <mo>−</mo> <mn>2</mn> <mo>≤</mo> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>2</mn> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mn>0</mn> <mo>≤</mo> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>25</mn> </mrow> </semantics> </math> and truncated by the surface, which covers red to yellow region) genomic regions <span class="html-italic">R</span> with values <math display="inline"> <semantics> <mrow> <mo>−</mo> <mn>2</mn> <mo>≤</mo> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>2</mn> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mn>0</mn> <mo>≤</mo> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>25</mn> </mrow> </semantics> </math> are observed. For these regions there is a low probability of observing SNPs (in accordance with Equations (4) and (5) and a low value of normalized counts supporting SNPs in the regions Equation (3). In another example, with low joint probability <math display="inline"> <semantics> <mrow> <mi>P</mi> <mo> </mo> <mo> </mo> <mrow> <mo>(</mo> <mrow> <mo>−</mo> <mn>1</mn> <mo>≤</mo> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>1</mn> <mo>,</mo> <mo> </mo> <mo> </mo> <mn>150</mn> <mo>≤</mo> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>200</mn> <mo> </mo> </mrow> <mo>)</mo> </mrow> </mrow> </semantics> </math> (corresponding to the volume of the prism truncated by the surface with squared base in the intervals <math display="inline"> <semantics> <mrow> <mo>−</mo> <mn>1</mn> <mo>≤</mo> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>1</mn> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mn>150</mn> <mo>≤</mo> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>200</mn> </mrow> </semantics> </math>), genomic regions <span class="html-italic">R</span> with values <math display="inline"> <semantics> <mrow> <mo>−</mo> <mn>1</mn> <mo>≤</mo> <msub> <mi>I</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>1</mn> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mn>150</mn> <mo>≤</mo> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> <mo>≤</mo> <mn>200</mn> </mrow> </semantics> </math> are observed; (<b>C</b>) 3D plot of the density probability distribution of the Farlie–Gumbel–Morgenstern copula built from the non-linear fit of the marginal distributions estimated for <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> (a Weibull PDF) and <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> (a Skew–Laplace PDF). The existence of a structural dependence between the variables, <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> is suggested by the Farlie–Gumbel–Morgenstern copula distribution [<a href="#B17-ijms-17-00938" class="html-bibr">17</a>,<a href="#B18-ijms-17-00938" class="html-bibr">18</a>], which describes in an acceptable approach the empirical behavior shown in panel B. That is, the stochastic relationship between the uncertainty variation of methylation levels (Equation (1)) and the uncertainty of not observing a SNP (Equations (3) and (5)) in a GR is confirmed. These estimations were performed for several <span class="html-italic">Arabidopsis</span> ecotypes. The results for the ecotypes La-0 and Fr.2 are shown.</p>
Full article ">Figure 8
<p>Classification of silencing mutants based on DI regions. (<b>A</b>,<b>B</b>) LDAs based on <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> estimated for CG and CHG methylation contexts, respectively; (<b>C</b>,<b>D</b>) fan dendrograms based on the individual coordinates estimated from the LD functions. The dendrograms were built by applying hierarchical clustering with Euclidean distance and UPGMA as agglomeration method. Roman numbers identify the main clades.</p>
Full article ">Figure 9
<p>Relationship between the distance matrices estimated for variables <span class="html-italic">I<sub>R</sub></span> and <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math>. The distance matrices were estimated for the ecotypes represented as vectors of the selected GFs. (<b>A</b>) the selection of GFs was based on the classification performance of each GR expressed in terms of AUC. In this case the features selected for <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> do not overlap with those selected for <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math>; (<b>B</b>) the selection of GFs was based on the classification performance of each GR expressed in terms of Chi-squared statistic. In this case the matrices were built with the intersection of GR features selected for <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math>. However, similitudes between topologies derived for the population structure based on <math display="inline"> <semantics> <mrow> <msub> <mi>I</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> and <math display="inline"> <semantics> <mrow> <mi>L</mi> <msub> <mi>C</mi> <mi>R</mi> </msub> </mrow> </semantics> </math> remain consistently high independent of the GF set selected, as reflected in the graphic and Mantel test results. This explains the semblance between the dendrograms presented in <a href="#ijms-17-00938-f006" class="html-fig">Figure 6</a>.</p>
Full article ">Figure 10
<p>Signal detection in noise according to reference [<a href="#B57-ijms-17-00938" class="html-bibr">57</a>,<a href="#B58-ijms-17-00938" class="html-bibr">58</a>] and, here, applied to the detection of regulatory CDM signals.</p>
Full article ">
8849 KiB  
Article
In Silico Prediction of Cytochrome P450-Drug Interaction: QSARs for CYP3A4 and CYP2C9
by Serena Nembri, Francesca Grisoni, Viviana Consonni and Roberto Todeschini
Int. J. Mol. Sci. 2016, 17(6), 914; https://doi.org/10.3390/ijms17060914 - 9 Jun 2016
Cited by 56 | Viewed by 7917
Abstract
Cytochromes P450 (CYP) are the main actors in the oxidation of xenobiotics and play a crucial role in drug safety, persistence, bioactivation, and drug-drug/food-drug interaction. This work aims to develop Quantitative Structure-Activity Relationship (QSAR) models to predict the drug interaction with two of [...] Read more.
Cytochromes P450 (CYP) are the main actors in the oxidation of xenobiotics and play a crucial role in drug safety, persistence, bioactivation, and drug-drug/food-drug interaction. This work aims to develop Quantitative Structure-Activity Relationship (QSAR) models to predict the drug interaction with two of the most important CYP isoforms, namely 2C9 and 3A4. The presented models are calibrated on 9122 drug-like compounds, using three different modelling approaches and two types of molecular description (classical molecular descriptors and binary fingerprints). For each isoform, three classification models are presented, based on a different approach and with different advantages: (1) a very simple and interpretable classification tree; (2) a local (k-Nearest Neighbor) model based classical descriptors and; (3) a model based on a recently proposed local classifier (N-Nearest Neighbor) on binary fingerprints. The salient features of the work are (1) the thorough model validation and the applicability domain assessment; (2) the descriptor interpretation, which highlighted the crucial aspects of P450-drug interaction; and (3) the consensus aggregation of models, which largely increased the prediction accuracy. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Scheme of the data splitting.</p>
Full article ">Figure 2
<p>Representation of the molecular descriptor (MD)-based models for CYP3A4: (<b>a</b>) Classification and Regression Trees (CART) model; (<b>b</b>) Score plot of the training molecules described by the <span class="html-italic">k</span>-Nearest Neighbours (<span class="html-italic">k</span>-NN) descriptors, coloured according to their activity; (<b>c</b>) Loading plot of the <span class="html-italic">k</span>-NN descriptors.</p>
Full article ">Figure 3
<p>Occurrence frequency of the 19 selected fragments for CYP3A4 within the active/inactive compounds. Symbols associated with the fragments (according to SMARTS language) have the following meaning: <span class="html-italic">X</span> + number = number of total bonds in which the considered atom is involved; a = aromatic atom; A = aliphatic atom.</p>
Full article ">Figure 3 Cont.
<p>Occurrence frequency of the 19 selected fragments for CYP3A4 within the active/inactive compounds. Symbols associated with the fragments (according to SMARTS language) have the following meaning: <span class="html-italic">X</span> + number = number of total bonds in which the considered atom is involved; a = aromatic atom; A = aliphatic atom.</p>
Full article ">Figure 4
<p>Representation of the MD-based models for CYP2C9: (<b>a</b>) CART model; (<b>b</b>) Score plot of the <span class="html-italic">k</span>-NN descriptors, colored according to their activity; (<b>c</b>) Loading plot of the <span class="html-italic">k</span>-NN descriptors.</p>
Full article ">Figure 5
<p>Occurrence frequency of the 16 selected fragments for CYP2C9 within the active/inactive compounds. Symbols associated with the fragments (according to SMARTS language) have the following meaning: <span class="html-italic">X</span> + number = number of total bonds in which the considered atom is involved; a = aromatic atom; A = aliphatic atom.</p>
Full article ">
4200 KiB  
Article
Development of an in Silico Model of DPPH• Free Radical Scavenging Capacity: Prediction of Antioxidant Activity of Coumarin Type Compounds
by Elizabeth Goya Jorge, Anita Maria Rayar, Stephen J. Barigye, María Elisa Jorge Rodríguez and Maité Sylla-Iyarreta Veitía
Int. J. Mol. Sci. 2016, 17(6), 881; https://doi.org/10.3390/ijms17060881 - 7 Jun 2016
Cited by 18 | Viewed by 7655
Abstract
A quantitative structure-activity relationship (QSAR) study of the 2,2-diphenyl-l-picrylhydrazyl (DPPH•) radical scavenging ability of 1373 chemical compounds, using DRAGON molecular descriptors (MD) and the neural network technique, a technique based on the multilayer multilayer perceptron (MLP), was developed. The built model demonstrated a [...] Read more.
A quantitative structure-activity relationship (QSAR) study of the 2,2-diphenyl-l-picrylhydrazyl (DPPH•) radical scavenging ability of 1373 chemical compounds, using DRAGON molecular descriptors (MD) and the neural network technique, a technique based on the multilayer multilayer perceptron (MLP), was developed. The built model demonstrated a satisfactory performance for the training ( R 2 = 0.713 ) and test set ( Q ext 2 = 0.654 ) , respectively. To gain greater insight on the relevance of the MD contained in the MLP model, sensitivity and principal component analyses were performed. Moreover, structural and mechanistic interpretation was carried out to comprehend the relationship of the variables in the model with the modeled property. The constructed MLP model was employed to predict the radical scavenging ability for a group of coumarin-type compounds. Finally, in order to validate the model’s predictions, an in vitro assay for one of the compounds (4-hydroxycoumarin) was performed, showing a satisfactory proximity between the experimental and predicted pIC50 values. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Regression plane of relation between targets, output and standard residuals values of the analyzed variable (pIC<sub>50</sub>).</p>
Full article ">Figure 2
<p>Correlation between experimental and predicted pIC<sub>50.</sub></p>
Full article ">Figure 3
<p>Sensitivity analysis of the MD for the MLP 14-9-1 model.</p>
Full article ">Figure 4
<p>Diagram of variable importance according to the PCA.</p>
Full article ">
15608 KiB  
Article
Physico-Chemical and Structural Interpretation of Discrete Derivative Indices on N-Tuples Atoms
by Oscar Martínez-Santiago, Yovani Marrero-Ponce, Stephen J. Barigye, Huong Le Thi Thu, F. Javier Torres, Cesar H. Zambrano, Jorge L. Muñiz Olite, Maykel Cruz-Monteagudo, Ricardo Vivas-Reyes, Liliana Vázquez Infante and Luis M. Artiles Martínez
Int. J. Mol. Sci. 2016, 17(6), 812; https://doi.org/10.3390/ijms17060812 - 27 May 2016
Cited by 7 | Viewed by 6038
Abstract
This report examines the interpretation of the Graph Derivative Indices (GDIs) from three different perspectives (i.e., in structural, steric and electronic terms). It is found that the individual vertex frequencies may be expressed in terms of the geometrical and electronic reactivity [...] Read more.
This report examines the interpretation of the Graph Derivative Indices (GDIs) from three different perspectives (i.e., in structural, steric and electronic terms). It is found that the individual vertex frequencies may be expressed in terms of the geometrical and electronic reactivity of the atoms and bonds, respectively. On the other hand, it is demonstrated that the GDIs are sensitive to progressive structural modifications in terms of: size, ramifications, electronic richness, conjugation effects and molecular symmetry. Moreover, it is observed that the GDIs quantify the interaction capacity among molecules and codify information on the activation entropy. A structure property relationship study reveals that there exists a direct correspondence between the individual frequencies of atoms and Hückel’s Free Valence, as well as between the atomic GDIs and the chemical shift in NMR, which collectively validates the theory that these indices codify steric and electronic information of the atoms in a molecule. Taking in consideration the regularity and coherence found in experiments performed with the GDIs, it is possible to say that GDIs possess plausible interpretation in structural and physicochemical terms. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Example of graph G (<b>A</b>), fragmentation according to an event “S” (<b>B</b>) and the sub-graphs sets for vertexes a and b (<b>C</b>).</p>
Full article ">Figure 2
<p>Scheme of the analogy between derivatives and their mathematical development. (<b>A</b>) Obtaining of the discrete derivative over a pair of elements <span class="html-italic">i j</span> from a graph G; (<b>B</b>) Algebraic development of the process for obtaining the discrete derivative over pairs of vertexes; (<b>C</b>) Obtaining of the classical derivative from the mathematical analysis.</p>
Full article ">Figure 3
<p>(<b>A</b>) Molecular structure of 2-amino-5-vinylfurane; (<b>B</b>) Corresponding graph with arbitrary numeration.</p>
Full article ">Figure 4
<p>Graphical behavior of the steric reactivity based in LOVIs calculated by GDIs and the Relative Bond Accessibility Area (RBA) proposed by Estrada for evaluating the accessibility.</p>
Full article ">Figure 5
<p>Regularity in the Derivative (over exocyclic double bond) variation and the log<span class="html-italic">K</span> values.</p>
Full article ">Figure 6
<p>Own Frequency and duplex derivative in electronic terms.</p>
Full article ">Figure 7
<p>Behavior of the free valence, the E-States and GDIs for atoms from 19 conjugated molecules.</p>
Full article ">Figure 8
<p>Development of the one-variable linear regression models obtained for each event: (<b>A</b>) Ethers; (<b>B</b>) Aldehydes and Ketones.</p>
Full article ">Figure 8 Cont.
<p>Development of the one-variable linear regression models obtained for each event: (<b>A</b>) Ethers; (<b>B</b>) Aldehydes and Ketones.</p>
Full article ">Figure 9
<p>Methylbutane. (<b>A</b>) Equivalence between LOVIs values and the chemical shift in ppm; (<b>B</b>) Quantity of protons that provoke the signal <span class="html-italic">vs</span>. LOVIs and ppm.</p>
Full article ">Figure 10
<p>2,2,3-Trimethylpentane. (<b>A</b>) Equivalence between LOVIs values and the chemical shift in ppm; (<b>B</b>) Number of protons that provoke the signal <span class="html-italic">vs.</span> LOVIs and ppm.</p>
Full article ">Figure 11
<p>Regression and prediction graphs for the Equation (27).</p>
Full article ">
1247 KiB  
Article
Fast Modeling of Binding Affinities by Means of Superposing Significant Interaction Rules (SSIR) Method
by Emili Besalú
Int. J. Mol. Sci. 2016, 17(6), 827; https://doi.org/10.3390/ijms17060827 - 26 May 2016
Cited by 7 | Viewed by 4789
Abstract
The Superposing Significant Interaction Rules (SSIR) method is described. It is a general combinatorial and symbolic procedure able to rank compounds belonging to combinatorial analogue series. The procedure generates structure-activity relationship (SAR) models and also serves as an inverse SAR tool. The method [...] Read more.
The Superposing Significant Interaction Rules (SSIR) method is described. It is a general combinatorial and symbolic procedure able to rank compounds belonging to combinatorial analogue series. The procedure generates structure-activity relationship (SAR) models and also serves as an inverse SAR tool. The method is fast and can deal with large databases. SSIR operates from statistical significances calculated from the available library of compounds and according to the previously attached molecular labels of interest or non-interest. The required symbolic codification allows dealing with almost any combinatorial data set, even in a confidential manner, if desired. The application example categorizes molecules as binding or non-binding, and consensus ranking SAR models are generated from training and two distinct cross-validation methods: leave-one-out and balanced leave-two-out (BL2O), the latter being suited for the treatment of binary properties. Full article
Show Figures

Figure 1

Figure 1
<p>Distribution of <span class="html-italic">p</span>-values for all the definable rules of order 4 (negations allowed) for (<b>a</b>) FPR1; and (<b>b</b>) FPR2 properties. Note the logarithmic scale in both axes.</p>
Full article ">Figure 2
<p>Receiver operating characteristic (ROC) curve and the area under it (<span class="html-italic">AU-ROC</span>) value for the FPR2 property calculated with the balanced leave-two-out (BL2O) cross-validation procedure (SSIR model involves rules of order 2, <span class="html-italic">p<sub>c</sub></span> = 0.005).</p>
Full article ">Figure 3
<p>Randomization test leave-one-out (L1O) predictions of <span class="html-italic">AU-ROC</span> values that could be obtained for the (<b>a</b>) FPR1; and (<b>b</b>) FPR2 properties after 1000 cycles. Horizontal axes (logarithmic units) show the number of rules entering in each SSIR model.</p>
Full article ">Figure 4
<p>Representation of the toy model of molecular scaffolding having three substitution sites that admit 2, 3 and 4 residues, respectively.</p>
Full article ">
3156 KiB  
Article
3D-QSAR Studies on Barbituric Acid Derivatives as Urease Inhibitors and the Effect of Charges on the Quality of a Model
by Zaheer Ul-Haq, Sajda Ashraf, Abdullah Mohammed Al-Majid and Assem Barakat
Int. J. Mol. Sci. 2016, 17(5), 657; https://doi.org/10.3390/ijms17050657 - 30 Apr 2016
Cited by 14 | Viewed by 7190
Abstract
Urease enzyme (EC 3.5.1.5) has been determined as a virulence factor in pathogenic microorganisms that are accountable for the development of different diseases in humans and animals. In continuance of our earlier study on the helicobacter pylori urease inhibition by barbituric acid derivatives, [...] Read more.
Urease enzyme (EC 3.5.1.5) has been determined as a virulence factor in pathogenic microorganisms that are accountable for the development of different diseases in humans and animals. In continuance of our earlier study on the helicobacter pylori urease inhibition by barbituric acid derivatives, 3D-QSAR (three dimensional quantitative structural activity relationship) advance studies were performed by Comparative Molecular Field Analysis (CoMFA) and Comparative Molecular Similarity Indices Analysis (CoMSIA) methods. Different partial charges were calculated to examine their consequences on the predictive ability of the developed models. The finest developed model for CoMFA and CoMSIA were achieved by using MMFF94 charges. The developed CoMFA model gives significant results with cross-validation (q2) value of 0.597 and correlation coefficients (r2) of 0.897. Moreover, five different fields i.e., steric, electrostatic, and hydrophobic, H-bond acceptor and H-bond donors were used to produce a CoMSIA model, with q2 and r2 of 0.602 and 0.98, respectively. The generated models were further validated by using an external test set. Both models display good predictive power with r2pred ≥ 0.8. The analysis of obtained CoMFA and CoMSIA contour maps provided detailed insight for the promising modification of the barbituric acid derivatives with an enhanced biological activity. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Superimposed view of <b>1</b>–<b>44</b> compounds by structure-based alignment using the reference ligand of 1E9Y.pdb. Cyan, red, blue, green and white color represent Hydrogen, Oxygen, Nitrogen, Chlorine and Carbon atom, respectively.</p>
Full article ">Figure 2
<p>Graphical interpretation of experimental <span class="html-italic">vs.</span> predicted pIC<sub>50</sub> of compounds <b>1</b>–<b>44</b> developed by CoMFA (<b>a</b>) and CoMSIA (<b>b</b>) models.</p>
Full article ">Figure 3
<p>CoMFA steric and electrostatic contour maps. Electronegativity and electropositivity are represented by red and blue contours, while sterically-favored and -disfavored areas are depicted by green and yellow regions, respectively. Panel (<b>a</b>) and (<b>c</b>) are representative of steric and electrostatic contours of most active comp-4i, while (<b>b</b>) and (<b>d</b>) are representative of least active comp-<b>5s</b>, respectively.</p>
Full article ">Figure 3 Cont.
<p>CoMFA steric and electrostatic contour maps. Electronegativity and electropositivity are represented by red and blue contours, while sterically-favored and -disfavored areas are depicted by green and yellow regions, respectively. Panel (<b>a</b>) and (<b>c</b>) are representative of steric and electrostatic contours of most active comp-4i, while (<b>b</b>) and (<b>d</b>) are representative of least active comp-<b>5s</b>, respectively.</p>
Full article ">Figure 4
<p>CoMSIA contour maps. Figure (<b>a</b>,<b>c</b>,<b>e</b>,<b>g</b>) displayed steric, electrostatic, hydrophobic, as well as donor contour maps of compound 4i claimed as the most active; While, (<b>b</b>,<b>d</b>,<b>f</b>,<b>h</b>) are representative of compounds, <b>5s</b> claimed as one of the least active compounds within the series.</p>
Full article ">Figure 5
<p>The 3D scatter plot of three selected principle components (PCA1, PCA2, and PCA3), each point corresponds to a molecule and is colored according to its activity.</p>
Full article ">Scheme 1
<p>Synthesis of compounds 4a-z and 5a-s.</p>
Full article ">
412 KiB  
Article
Is It Reliable to Use Common Molecular Docking Methods for Comparing the Binding Affinities of Enantiomer Pairs for Their Protein Target?
by David Ramírez and Julio Caballero
Int. J. Mol. Sci. 2016, 17(4), 525; https://doi.org/10.3390/ijms17040525 - 20 Apr 2016
Cited by 117 | Viewed by 9329
Abstract
Molecular docking is a computational chemistry method which has become essential for the rational drug design process. In this context, it has had great impact as a successful tool for the study of ligand–receptor interaction modes, and for the exploration of large chemical [...] Read more.
Molecular docking is a computational chemistry method which has become essential for the rational drug design process. In this context, it has had great impact as a successful tool for the study of ligand–receptor interaction modes, and for the exploration of large chemical datasets through virtual screening experiments. Despite their unquestionable merits, docking methods are not reliable for predicting binding energies due to the simple scoring functions they use. However, comparisons between two or three complexes using the predicted binding energies as a criterion are commonly found in the literature. In the present work we tested how wise is it to trust the docking energies when two complexes between a target protein and enantiomer pairs are compared. For this purpose, a ligand library composed by 141 enantiomeric pairs was used, including compounds with biological activities reported against seven protein targets. Docking results using the software Glide (considering extra precision (XP), standard precision (SP), and high-throughput virtual screening (HTVS) modes) and AutoDock Vina were compared with the reported biological activities using a classification scheme. Our test failed for all modes and targets, demonstrating that an accurate prediction when binding energies of enantiomers are compared using docking may be due to chance. We also compared pairs of compounds with different molecular weights and found the same results. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Number of publications where molecular docking was used (search in Scopus).</p>
Full article ">
2231 KiB  
Article
Structural Investigation for Optimization of Anthranilic Acid Derivatives as Partial FXR Agonists by in Silico Approaches
by Meimei Chen, Xuemei Yang, Xinmei Lai, Jie Kang, Huijuan Gan and Yuxing Gao
Int. J. Mol. Sci. 2016, 17(4), 536; https://doi.org/10.3390/ijms17040536 - 8 Apr 2016
Cited by 13 | Viewed by 5805
Abstract
In this paper, a three level in silico approach was applied to investigate some important structural and physicochemical aspects of a series of anthranilic acid derivatives (AAD) newly identified as potent partial farnesoid X receptor (FXR) agonists. Initially, both two and three-dimensional quantitative [...] Read more.
In this paper, a three level in silico approach was applied to investigate some important structural and physicochemical aspects of a series of anthranilic acid derivatives (AAD) newly identified as potent partial farnesoid X receptor (FXR) agonists. Initially, both two and three-dimensional quantitative structure activity relationship (2D- and 3D-QSAR) studies were performed based on such AAD by a stepwise technology combined with multiple linear regression and comparative molecular field analysis. The obtained 2D-QSAR model gave a high predictive ability (R2train = 0.935, R2test = 0.902, Q2LOO = 0.899). It also uncovered that number of rotatable single bonds (b_rotN), relative negative partial charges (RPC), oprea's lead-like (opr_leadlike), subdivided van der Waal’s surface area (SlogP_VSA2) and accessible surface area (ASA) were important features in defining activity. Additionally, the derived3D-QSAR model presented a higher predictive ability (R2train = 0.944, R2test = 0.892, Q2LOO = 0.802). Meanwhile, the derived contour maps from the 3D-QSAR model revealed the significant structural features (steric and electronic effects) required for improving FXR agonist activity. Finally, nine newly designed AAD with higher predicted EC50 values than the known template compound were docked into the FXR active site. The excellent molecular binding patterns of these molecules also suggested that they can be robust and potent partial FXR agonists in agreement with the QSAR results. Overall, these derived models may help to identify and design novel AAD with better FXR agonist activity. Full article
Show Figures

Figure 1

Figure 1
<p>Plots of experimental against predicted pEC<sub>50</sub> values by (<b>A</b>) multiple linear regression (MLR) and (<b>B</b>) CoMFA models.</p>
Full article ">Figure 2
<p>The Williams plot for the MLR model.</p>
Full article ">Figure 3
<p>Contour maps of the CoMFA model: (<b>A</b>) steric field based on compound 30; (<b>B</b>) electrostatic field based on compound 30. Color values specify the CoMFA field levels that enclose volumes within which increase or decrease in bulk or positive charge favor higher dependent values.</p>
Full article ">Figure 4
<p>Structure of template compound (compound 30). The three regions A, B and C are depicted.</p>
Full article ">Figure 5
<p>The best docked conformations and poses of newly designed compounds in the ligand binding domain of FXR.</p>
Full article ">Figure 6
<p>The 2D representation of docking of compounds N9 (<b>A</b>) and complexed ligand (<b>B</b>) into the FXR active site.</p>
Full article ">Figure 7
<p>Alignment of training and test set compounds on compound 30. Baby blue, red, blue, green, gray and yellow signify hydrogen atom, oxygen atom, nitrogen atom, fluorine atom, carbon atom and sulfur atom, respectively.</p>
Full article ">
1608 KiB  
Article
Hyaluronidase Inhibitory Activity of Pentacylic Triterpenoids from Prismatomeris tetrandra (Roxb.) K. Schum: Isolation, Synthesis and QSAR Study
by Nor Hayati Abdullah, Noel Francis Thomas, Yasodha Sivasothy, Vannajan Sanghiran Lee, Sook Yee Liew, Ibrahim Ali Noorbatcha and Khalijah Awang
Int. J. Mol. Sci. 2016, 17(2), 143; https://doi.org/10.3390/ijms17020143 - 14 Feb 2016
Cited by 20 | Viewed by 7420
Abstract
The mammalian hyaluronidase degrades hyaluronic acid by the cleavage of the β-1,4-glycosidic bond furnishing a tetrasaccharide molecule as the main product which is a highly angiogenic and potent inducer of inflammatory cytokines. Ursolic acid 1, isolated from Prismatomeris tetrandra, was identified [...] Read more.
The mammalian hyaluronidase degrades hyaluronic acid by the cleavage of the β-1,4-glycosidic bond furnishing a tetrasaccharide molecule as the main product which is a highly angiogenic and potent inducer of inflammatory cytokines. Ursolic acid 1, isolated from Prismatomeris tetrandra, was identified as having the potential to develop inhibitors of hyaluronidase. A series of ursolic acid analogues were either synthesized via structure modification of ursolic acid 1 or commercially obtained. The evaluation of the inhibitory activity of these compounds on the hyaluronidase enzyme was conducted. Several structural, topological and quantum chemical descriptors for these compounds were calculated using semi empirical quantum chemical methods. A quantitative structure activity relationship study (QSAR) was performed to correlate these descriptors with the hyaluronidase inhibitory activity. The statistical characteristics provided by the best multi linear model (BML) (R2 = 0.9717, R2cv = 0.9506) indicated satisfactory stability and predictive ability of the developed model. The in silico molecular docking study which was used to determine the binding interactions revealed that the ursolic acid analog 22 had a strong affinity towards human hyaluronidase. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Synthesis of ursolic acid <b>1</b> derivatives with different substituents at C-3. ((C<sub>5</sub>H<sub>6</sub>N {ClCrNO<sub>3</sub>}) = Pyridinium chlorochromate; CH<sub>2</sub>Cl<sub>2</sub> = dichloromethane; (CH<sub>3</sub>)<sub>2</sub>CO = acetone; H<sub>2</sub>NOHHCl = hydroxylamine hydrochloride; (CH<sub>3</sub>CO)<sub>2</sub> = acetic anhydride; (CH<sub>3</sub>SiCH<sub>2</sub>N<sup>+</sup>N) = trimethylsilyl diazomethane (TMS)).</p>
Full article ">Figure 2
<p>Structure activity relationship of pentacyclic triterpenes (PTs).</p>
Full article ">Figure 3
<p>Comparison of the experimental hyaluronidase activity with the activity presented by the QSAR Equation (1), <span class="html-italic">n</span> = 20, with <span class="html-italic">R</span><sup>2</sup> = 0.8579; <span class="html-italic">s</span><sup>2</sup> = 0.0246; <span class="html-italic">F</span> = 21.13; four descriptors.</p>
Full article ">Figure 4
<p>Structure of new PTC A compound.</p>
Full article ">Figure 5
<p>Superimposed the complex structures of apigenin and 22 with human hyaluronidase (<b>left</b>). The interactions with residues interaction energy below −4 kcal/mol of apigenin (<b>right</b>, <b>top</b>) and 22 (<b>right</b>, <b>bottom</b>) with human hyaluronidase were illustrated. The π–π and hydrogen bonding interaction are depicted in orange and green dashed, respectively.</p>
Full article ">
6385 KiB  
Article
Computational Analysis of Structure-Based Interactions for Novel H1-Antihistamines
by Yinfeng Yang, Yan Li, Yanqiu Pan, Jinghui Wang, Feng Lin, Chao Wang, Shuwei Zhang and Ling Yang
Int. J. Mol. Sci. 2016, 17(1), 129; https://doi.org/10.3390/ijms17010129 - 19 Jan 2016
Cited by 18 | Viewed by 8740
Abstract
As a chronic disorder, insomnia affects approximately 10% of the population at some time during their lives, and its treatment is often challenging. Since the antagonists of the H1 receptor, a protein prevalent in human central nervous system, have been proven as [...] Read more.
As a chronic disorder, insomnia affects approximately 10% of the population at some time during their lives, and its treatment is often challenging. Since the antagonists of the H1 receptor, a protein prevalent in human central nervous system, have been proven as effective therapeutic agents for treating insomnia, the H1 receptor is quite possibly a promising target for developing potent anti-insomnia drugs. For the purpose of understanding the structural actors affecting the antagonism potency, presently a theoretical research of molecular interactions between 129 molecules and the H1 receptor is performed through three-dimensional quantitative structure-activity relationship (3D-QSAR) techniques. The ligand-based comparative molecular similarity indices analysis (CoMSIA) model (Q2 = 0.525, R2ncv = 0.891, R2pred = 0.807) has good quality for predicting the bioactivities of new chemicals. The cross-validated result suggests that the developed models have excellent internal and external predictability and consistency. The obtained contour maps were appraised for affinity trends for the investigated compounds, which provides significantly useful information in the rational drug design of novel anti-insomnia agents. Molecular docking was also performed to investigate the mode of interaction between the ligand and the active site of the receptor. Furthermore, as a supplementary tool to study the docking conformation of the antagonists in the H1 receptor binding pocket, molecular dynamics simulation was also applied, providing insights into the changes in the structure. All of the models and the derived information would, we hope, be of help for developing novel potent histamine H1 receptor antagonists, as well as exploring the H1-antihistamines interaction mechanism. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Distribution of activities (p<span class="html-italic">K<sub>i</sub></span>) for the training and the test sets <span class="html-italic">versus</span> the numbers of compounds. The training and the test sets are colored blue and orange, respectively.</p>
Full article ">Figure 2
<p>The ligand-based correlation plots of the predicted <span class="html-italic">versus</span> the actual p<span class="html-italic">K<sub>i</sub></span> values using the training (filled red triangles) and the test (filled black dots) set compounds based on the optimal CoMSIA model.</p>
Full article ">Figure 3
<p>Contour maps of CoMSIA combined with compound 49. (<b>A</b>) Contour maps in steric (green/yellow) fields. Green and yellow contours represent regions where bulky groups will increase and decrease the activity, respectively; (<b>B</b>) Contour maps in electrostatic (red/blue) fields. Red and blue contours represent regions where negative- and positive-charged substituents will decrease and increase the activity, respectively; (<b>C</b>) Contour maps in hydrophobic (yellow/gray) fields. Yellow and gray contours represent regions where the hydrophobic and hydrophilic groups will increase their activity; (<b>D</b>) Contour maps in H-bond (HB) donor (cyan/purple) fields. Cyan and purple contours represent regions where HB donor substituents will enhance and decrease the activity, respectively.</p>
Full article ">Figure 4
<p>(<b>A</b>) Binding poses of co-crystallized (magenta) and re-docked (green) compound doxepin; (<b>B</b>) overlap of the compound 49 (orange) and experimental doxepin (green; PDB code: 3RZE) conformation.</p>
Full article ">Figure 5
<p>Docked conformation of compound 49 into histamine H<sub>1</sub> receptor. The projection highlights the structure of the active site with compound 49, which is displayed in sticks.</p>
Full article ">Figure 6
<p>(<b>A</b>) Plot of the root-mean-square deviation (RMSD) of docked complex/ligand <span class="html-italic">versus</span> the MD simulation time in the MD-simulated structures; (<b>B</b>) view of the superimposed backbone atoms of the average structure for the MD simulations and the initial structure of the docking for the complex. Compound 49 is represented as a carbon-chain in green for the initial complex and a carbon-chain in orange for the average structure, respectively.</p>
Full article ">Figure 7
<p>Plot of the MD-simulated structures of the binding site with compound 49. H-bonds are shown as dotted black lines; amino acid residues in the active site are represented as sticks.</p>
Full article ">Figure 8
<p>(<b>A</b>) Compound 49 used as the template molecule for alignments, with the common framework marked in blue bold. The substituent containing a protonated –NMe<sub>2</sub> group at the position-18 is depicted in a red oval, which would be more desirable for potent antagonism activity; (<b>B</b>–<b>D</b>) show the results of Alignment-I, -II and -III of all molecules, respectively. All compounds in these panels are colored white for common carbon, blue for nitrogen, red for oxygen, yellow for sulfur and cyan for hydrogen atoms, respectively.</p>
Full article ">Figure 9
<p>Proposed hypothetical histamine H<sub>1</sub>-receptor active site models. The structure-activity relationship is taken from the results of 3D-QSAR, docking and MD simulation studies for compound 49.</p>
Full article ">

Review

Jump to: Editorial, Research

14758 KiB  
Review
Virtual Screening Approaches towards the Discovery of Toll-Like Receptor Modulators
by Lucía Pérez-Regidor, Malik Zarioh, Laura Ortega and Sonsoles Martín-Santamaría
Int. J. Mol. Sci. 2016, 17(9), 1508; https://doi.org/10.3390/ijms17091508 - 9 Sep 2016
Cited by 33 | Viewed by 12444
Abstract
This review aims to summarize the latest efforts performed in the search for novel chemical entities such as Toll-like receptor (TLR) modulators by means of virtual screening techniques. This is an emergent research field with only very recent (and successful) contributions. Identification of [...] Read more.
This review aims to summarize the latest efforts performed in the search for novel chemical entities such as Toll-like receptor (TLR) modulators by means of virtual screening techniques. This is an emergent research field with only very recent (and successful) contributions. Identification of drug-like molecules with potential therapeutic applications for the treatment of a variety of TLR-regulated diseases has attracted considerable interest due to the clinical potential. Additionally, the virtual screening databases and computational tools employed have been overviewed in a descriptive way, widening the scope for researchers interested in the field. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Summary of the virtual screening (VS) protocols applied for the search for novel Toll-like receptors (TLR) modulators: access to databases and preparation/filtering of small-molecules; pharmacophore generation; docking calculations; selection of candidates; experimental testing, and final identification of drug candidates.</p>
Full article ">Figure 2
<p>Representations of the 3D structures of TLR2/1 (<b>left</b>); and TLR2/6 (<b>right</b>) complexes with Pam3CSK4 and Pam2CSK4, respectively. The 2D structure of some compounds mentioned in the text are shown.</p>
Full article ">Figure 3
<p>Representation of the X-ray structure of TLR4/MD-2 system (PDB-ID: 3FXI) in complex with <span class="html-italic">Escherichia coli</span> LPS. Right view: detail of the LPS (<b>green</b>) bound to TLR4 (<b>yellow</b>) and MD-2 (<b>orange</b>). Partner TLR4*/MD-2* system is represented in violet colors.</p>
Full article ">Figure 4
<p>3D structure representation of the extracellular domain of the TLR4/MD-2 complex with Eritoran (PDB-ID: 2Z65) focused on the interaction surfaces between TLR4 (<b>yellow</b>), and MD-2 (<b>orange</b>). Polar amino-acid residues used to perform the docking procedure are shown in sticks.</p>
Full article ">
4536 KiB  
Review
Deep Artificial Neural Networks and Neuromorphic Chips for Big Data Analysis: Pharmaceutical and Bioinformatics Applications
by Lucas Antón Pastur-Romay, Francisco Cedrón, Alejandro Pazos and Ana Belén Porto-Pazos
Int. J. Mol. Sci. 2016, 17(8), 1313; https://doi.org/10.3390/ijms17081313 - 11 Aug 2016
Cited by 73 | Viewed by 21618
Abstract
Over the past decade, Deep Artificial Neural Networks (DNNs) have become the state-of-the-art algorithms in Machine Learning (ML), speech recognition, computer vision, natural language processing and many other tasks. This was made possible by the advancement in Big Data, Deep Learning (DL) and [...] Read more.
Over the past decade, Deep Artificial Neural Networks (DNNs) have become the state-of-the-art algorithms in Machine Learning (ML), speech recognition, computer vision, natural language processing and many other tasks. This was made possible by the advancement in Big Data, Deep Learning (DL) and drastically increased chip processing abilities, especially general-purpose graphical processing units (GPGPUs). All this has created a growing interest in making the most of the potential offered by DNNs in almost every field. An overview of the main architectures of DNNs, and their usefulness in Pharmacology and Bioinformatics are presented in this work. The featured applications are: drug design, virtual screening (VS), Quantitative Structure–Activity Relationship (QSAR) research, protein structure prediction and genomics (and other omics) data mining. The future need of neuromorphic hardware for DNNs is also discussed, and the two most advanced chips are reviewed: IBM TrueNorth and SpiNNaker. In addition, this review points out the importance of considering not only neurons, as DNNs and neuromorphic chips should also include glial cells, given the proven importance of astrocytes, a type of glial cell which contributes to information processing in the brain. The Deep Artificial Neuron–Astrocyte Networks (DANAN) could overcome the difficulties in architecture design, learning process and scalability of the current ML methods. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Big Data Workflow.</p>
Full article ">Figure 2
<p>Deep neural network architecture from Yanjun Qi et al. [<a href="#B45-ijms-17-01313" class="html-bibr">45</a>]. The input to the first layer is the protein sequence represented by the single-letter amino acid code, for example the letter “A” (in green) represents “Alanine”. This method uses a sliding window input {S<sub>1</sub>, S<sub>2</sub>… S<sub>k</sub>}, in this case <span class="html-italic">k</span> = 7. The first layer consists a PSI-Blast feature module and an amino acid embedding module, the green boxes represent the feature vector derived from the Alanine in both modules. In the second layer, the feature vectors are concatenated to facilitate identification of local sequence structure. Finally the derived vector is fed into the Deep Artificial Neural Network.</p>
Full article ">Figure 3
<p>Convolutional layers that extract features of the input to create a feature map. The artificial neurons are represented by the circles, and the weights by the narrows. Weights of the same color are shared, constrained to be identical [<a href="#B56-ijms-17-01313" class="html-bibr">56</a>].</p>
Full article ">Figure 4
<p>Architecture of a Deep Convolutional Neural Network (DCNN), alternating the convolutional layer and the max-pooling layer (or sub-sampling layer), and finally the fully-connected layer [<a href="#B56-ijms-17-01313" class="html-bibr">56</a>].</p>
Full article ">Figure 5
<p>This diagram represents a simplification of the structure of the epoxidation model, which was made up of one input layer, two hidden layers, and two output layers. The actual model had several additional nodes in the input and hidden layers. In the input layer, M represents the molecule input node, B is the bond input node, and two atom input nodes (for each atom associated with the bond). The bond epoxidation score (BES) quantifies the probability that the bond is a site of epoxidation based in the input from the nodes of the first hidden layer (H<sub>1</sub> and H<sub>2</sub>). The molecule epoxidation score (MES) reflects the probability that the molecule will be epoxidized. This score is calculated with the information from the all molecule-level descriptors and the BES. The bond network and the molecule network are represented in orange and purple respectively [<a href="#B57-ijms-17-01313" class="html-bibr">57</a>].</p>
Full article ">Figure 6
<p>Details of inner workings of DeepBind developed by Alipanahi et al. and its training procedure. In “<b>a</b>”, five independent sequences of DNA are being processed in parallel, each composed by a string of letters (C, G, A and T) which represent the nucleotides. The scores are represented in white and red tones, and the outputs are compared to the targets to improve the model using backpropagation; In “<b>b</b>”, The Calibration, training, and tasting procedure is represented in more detail [<a href="#B59-ijms-17-01313" class="html-bibr">59</a>].</p>
Full article ">Figure 7
<p>Different Recurrent Neural Networks architectures, the white circles represent the input layers, the black circles the hidden layers, and the grey circles the output layers [<a href="#B65-ijms-17-01313" class="html-bibr">65</a>].</p>
Full article ">Figure 8
<p>Schematic diagram of Youjun Xu et al. network encoding glycine, first using primary canonical SMILES strucuture. Then, each of the atoms in the SMILES structure is sequentially defined as a root node. Finally, information for all other atoms is transferred along the shortest possible paths, in which case is obtained following the narrows [<a href="#B67-ijms-17-01313" class="html-bibr">67</a>].</p>
Full article ">Figure 9
<p>Mapping a Deep Artificial Neural Network (DANN) (<b>a</b>) to a neuromorphic chip like the TrueNorth (<b>b</b>). The input neurons are represented with the red and white shapes (x and x’), and the output neurons with the grey shapes (z and z’). The weights (w) to the neuron z are approximated using a Pseudo Random Number Generator (PRNG), resulting in the weights (w’) to the neuron z’ in the neuromorphic chip [<a href="#B74-ijms-17-01313" class="html-bibr">74</a>].</p>
Full article ">Figure 10
<p>(<b>A</b>) The neurosynaptic core is loosely inspired by the idea of a canonical cortical microcircuit; (<b>B</b>) A network of neurosynaptic cores is inspired by the cortex’s two-dimensional sheet, the brain regions are represented in different colors; (<b>C</b>) The multichip network is inspired by the long-range connections between cortical regions shown from the macaque brain; (<b>D</b>–<b>F</b>) Structural scheme of the core, chip and multi-chip level. The white shapes represent axons (inputs) and the grey shapes the neurons (outputs); (<b>G</b>–<b>I</b>) Functional view at different level; (<b>J</b>–<b>L</b>) Image of the physical layout [<a href="#B77-ijms-17-01313" class="html-bibr">77</a>].</p>
Full article ">Figure 11
<p>Mapping of a CNN to TrueNorth. (<b>A</b>) Convolutional network features for one group at one topographic location are implemented using neurons on the same TrueNorth core, with their corresponding filter support region implemented using the core’s input lines, and filter weights implemented using the core’s synaptic array. The inputs are represented with white shapes, and the grey triangles represent the neurons. The filter used in each case is implemented mapping the matrix of weights (the numbers in the green boxes) into the synaptic array (grey circles); (<b>B</b>) For a neuron (blue points) to target multiple core inputs, its output (orange points) must be replicated by neuron copies, recruited from other neurons on the same core, or on extra cores if needed [<a href="#B76-ijms-17-01313" class="html-bibr">76</a>].</p>
Full article ">Figure 12
<p>Sparse coding applied to audio. In red 20 basis functions learned from unlabeled audio, in blue the functions from cat auditory nerve fibers [<a href="#B113-ijms-17-01313" class="html-bibr">113</a>].</p>
Full article ">Figure 13
<p>Tripartite synapse represented by a presynaptic neuron, postsynaptic neuron and perisynaptic astrocyte (astrocyte process). The presynaptic neuron release neurotransmitters that are received by the postsynaptic neuron or the perisynaptic astrocyte [<a href="#B129-ijms-17-01313" class="html-bibr">129</a>].</p>
Full article ">
363 KiB  
Review
A Brief Review of Computer-Assisted Approaches to Rational Design of Peptide Vaccines
by Ashesh Nandy and Subhash C. Basak
Int. J. Mol. Sci. 2016, 17(5), 666; https://doi.org/10.3390/ijms17050666 - 4 May 2016
Cited by 49 | Viewed by 10350
Abstract
The growing incidences of new viral diseases and increasingly frequent viral epidemics have strained therapeutic and preventive measures; the high mutability of viral genes puts additional strains on developmental efforts. Given the high cost and time requirements for new drugs development, vaccines remain [...] Read more.
The growing incidences of new viral diseases and increasingly frequent viral epidemics have strained therapeutic and preventive measures; the high mutability of viral genes puts additional strains on developmental efforts. Given the high cost and time requirements for new drugs development, vaccines remain as a viable alternative, but there too traditional techniques of live-attenuated or inactivated vaccines have the danger of allergenic reactions and others. Peptide vaccines have, over the last several years, begun to be looked on as more appropriate alternatives, which are economically affordable, require less time for development and hold the promise of multi-valent dosages. The developments in bioinformatics, proteomics, immunogenomics, structural biology and other sciences have spurred the growth of vaccinomics where computer assisted approaches serve to identify suitable peptide targets for eventual development of vaccines. In this mini-review we give a brief overview of some of the recent trends in computer assisted vaccine development with emphasis on the primary selection procedures of probable peptide candidates for vaccine development. Full article
Show Figures

Figure 1

Figure 1
<p>Work flow chart of peptide selection process, as in References [<a href="#B26-ijms-17-00666" class="html-bibr">26</a>,<a href="#B27-ijms-17-00666" class="html-bibr">27</a>,<a href="#B28-ijms-17-00666" class="html-bibr">28</a>].</p>
Full article ">
Back to TopTop