1
|
Akella M, Malla R. Molecular modeling and in vitro study on pyrocatechol as potential pharmacophore of CD151 inhibitor. J Mol Graph Model 2020; 100:107681. [PMID: 32738620 DOI: 10.1016/j.jmgm.2020.107681] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/25/2020] [Accepted: 06/24/2020] [Indexed: 11/24/2022]
Abstract
CD151 has been recognized as a prognostic marker, the therapeutic target of breast cancers, but less explored for small molecule inhibitors due to lack of a validated model. The 3-D structure of CD151 large extracellular loop (LEL) was modeled using the LOMETS server and validated by the Ramachandran plot. The validated structure was employed for molecular docking and structure-based pharmacophore analysis. Druglikeness was evaluated by the ADMET description protocol. Antiproliferative activity was evaluated by MTT, BrdU incorporation, flow cytometry, and cell death ELISAPLUS assay. This study predicted the best model for CD151-LEL with 94.1% residues in favored regions and Z score -2.79 kcal/mol using the threading method. The web-based receptor cavity method identified one functional target site, which was suitable for the binding of aromatic and heterocyclic compounds. Molecular docking study identified pyrocatechol (PCL) and 5-fluorouracil (FU) as potential leads of CD151-LEL. The pharmacophore model identified interaction points of modeled CD151-LEL with PCL and FU. Also, the analysis of ADMET properties revealed the drug-likeness of PCL and FU. The viability of MDA-MB 231 cells was significantly reduced with PCL and FU but less affected MCF-12A, normal healthy breast epithelial cell line. With 50% toxic concentration, both PCL and FU significantly inhibited 82.46 and 87.12% proliferation, respectively, of MDA-MB 231 cells by altering morphology and inducing G1 cell cycle arrest and apoptosis. In addition, PCL and FU inhibited the CD151 expression by 4.5-and 4.8-folds, respectively. This study suggests the further assessment of pyrocatechol as a potential lead of CD151 in breast cancer at the molecular level.
Collapse
Affiliation(s)
- Manasa Akella
- Cancer Biology Lab, Dept. of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to Be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - RamaRao Malla
- Cancer Biology Lab, Dept. of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to Be University), Visakhapatnam, 530045, Andhra Pradesh, India.
| |
Collapse
|
2
|
Eguida M, Rognan D. A Computer Vision Approach to Align and Compare Protein Cavities: Application to Fragment-Based Drug Design. J Med Chem 2020; 63:7127-7142. [DOI: 10.1021/acs.jmedchem.0c00422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Merveille Eguida
- UMR 7200 CNRS-Université de Strasbourg, Laboratoire d’Innovation Thérapeutique, 67400 Illkirch, France
| | - Didier Rognan
- UMR 7200 CNRS-Université de Strasbourg, Laboratoire d’Innovation Thérapeutique, 67400 Illkirch, France
| |
Collapse
|
3
|
Bartolowits M, Davisson VJ. Considerations of Protein Subpockets in Fragment-Based Drug Design. Chem Biol Drug Des 2015; 87:5-20. [PMID: 26307335 DOI: 10.1111/cbdd.12631] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
While the fragment-based drug design approach continues to gain importance, gaps in the tools and methods available in the identification and accurate utilization of protein subpockets have limited the scope. The importance of these features of small molecule-protein recognition is highlighted with several examples. A generalized solution for the identification of subpockets and corresponding chemical fragments remains elusive, but there are numerous advancements in methods that can be used in combination to address subpockets. Finally, additional examples of approaches that consider the relative importance of small-molecule co-dependence of protein conformations are highlighted to emphasize an increased significance of subpockets, especially at protein interfaces.
Collapse
Affiliation(s)
- Matthew Bartolowits
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47907, USA
| | - V Jo Davisson
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, 575 Stadium Mall Dr., West Lafayette, IN, 47907, USA
| |
Collapse
|
4
|
Vlachakis D, Fakourelis P, Megalooikonomou V, Makris C, Kossida S. DrugOn: a fully integrated pharmacophore modeling and structure optimization toolkit. PeerJ 2015; 3:e725. [PMID: 25648563 PMCID: PMC4304849 DOI: 10.7717/peerj.725] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 12/24/2014] [Indexed: 11/20/2022] Open
Abstract
During the past few years, pharmacophore modeling has become one of the key components in computer-aided drug design and in modern drug discovery. DrugOn is a fully interactive pipeline designed to exploit the advantages of modern programming and overcome the command line barrier with two friendly environments for the user (either novice or experienced in the field of Computer Aided Drug Design) to perform pharmacophore modeling through an efficient combination of the PharmACOphore, Gromacs, Ligbuilder and PDB2PQR suites. Our platform features a novel workflow that guides the user through each logical step of the iterative 3D structural optimization setup and drug design process. For the pharmacophore modeling we are focusing on either the characteristics of the receptor or the full molecular system, including a set of selected ligands. DrugOn can be freely downloaded from our dedicated server system at www.bioacademy.gr/bioinformatics/drugon/.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Bioinformatics & Medical Informatics Team, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Computer Engineering and Informatics Department, University of Patras, Patras, Greece
| | - Paraskevas Fakourelis
- Bioinformatics & Medical Informatics Team, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Computer Engineering and Informatics Department, University of Patras, Patras, Greece
| | | | - Christos Makris
- Computer Engineering and Informatics Department, University of Patras, Patras, Greece
| | - Sophia Kossida
- Bioinformatics & Medical Informatics Team, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,IMGT, Laboratoire d'ImmunoGénétique Moléculaire, Institut de Génétique Humaine, Montpellier, France
| |
Collapse
|
5
|
Abstract
The amount of known protein structures is continuously growing, exhibited in over 95,000 3D structures freely available via the PDB. Over the last decade, pharmaceutical research has sparked interest in computationally extracting information from this large data pool, resulting in a homology-driven knowledge transfer from annotated to new structures. Studying protein structures with respect to understanding and modulating their functional behavior means analyzing their centers of action. Therefore, the detection and description of potential binding sites on the protein surface is a major step towards protein classification and assessment. Subsequently, these representations can be incorporated to compare proteins, and to predict their druggability or function. Especially in the context of target identification and polypharmacology, automated tools for large-scale target comparisons are highly needed. In this article, developments for automated structure-based target assessment are reviewed and remaining challenges as well as future perspectives are discussed.
Collapse
|
6
|
Jalencas X, Mestres J. Identification of Similar Binding Sites to Detect Distant Polypharmacology. Mol Inform 2013; 32:976-90. [PMID: 27481143 DOI: 10.1002/minf.201300082] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/29/2013] [Indexed: 01/19/2023]
Abstract
The ability of small molecules to interact with multiple proteins is referred to as polypharmacology. This property is often linked to the therapeutic action of drugs but it is known also to be responsible for many of their side effects. Because of its importance, the development of computational methods that can predict drug polypharmacology has become an important line of research that led recently to the identification of many novel targets for known drugs. Nowadays, the majority of these methods are based on measuring the similarity of a query molecule against the hundreds of thousands of molecules for which pharmacological data on thousands of proteins are available in public sources. However, similarity-based methods are inherently biased by the chemical coverage offered by the active molecules present in those public repositories, which limits significantly their capacity to predict interactions with proteins structurally and functionally unrelated to any of the already known targets for drugs. It is in this respect that structure-based methods aiming at identifying similar binding sites may offer an alternative complementary means to ligand-based methods for detecting distant polypharmacology. The different existing approaches to binding site detection, representation, comparison, and fragmentation are reviewed and recent successful applications presented.
Collapse
Affiliation(s)
- Xavier Jalencas
- Systems Pharmacology, Research Program on Biomedical Informatics (GRIB), IMIM Hospital del Mar Research Institute & University Pompeu Fabra, Parc de Recerca Biomèdica, Doctor Aiguader 88, 08003 Barcelona, Catalonia, Spain fax: +34 93 3160550
| | - Jordi Mestres
- Systems Pharmacology, Research Program on Biomedical Informatics (GRIB), IMIM Hospital del Mar Research Institute & University Pompeu Fabra, Parc de Recerca Biomèdica, Doctor Aiguader 88, 08003 Barcelona, Catalonia, Spain fax: +34 93 3160550.
| |
Collapse
|
7
|
Chemogenomics in drug discovery: computational methods based on the comparison of binding sites. Future Med Chem 2013; 4:1971-9. [PMID: 23088277 DOI: 10.4155/fmc.12.147] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Novel computational methods for understanding relationships between ligands and all possible biological targets have emerged in recent years. Proteins are connected to each other based on the similarity of their ligands or based on the similarity of their binding sites. The assumption is that compounds sharing chemical similarity should share targets and that targets with a similar binding site should also share ligands. A large number of computational techniques have been developed to assess ligand and binding site similarity, which can be used to make quantitative predictions of the most probable biological target of a given compound. This review covers the recent advances in new computational methods for relating biological targets based on the similarity of their binding sites. Binding site comparisons are used for the prediction of their most likely ligands, their possible cross reactivity and selectivity. These comparisons can also be used to infer the function of novel uncharacterized proteins.
Collapse
|
8
|
Kalliokoski T, Olsson TSG, Vulpetti A. Subpocket analysis method for fragment-based drug discovery. J Chem Inf Model 2013; 53:131-41. [PMID: 23327721 DOI: 10.1021/ci300523r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Although two binding sites might be dissimilar overall, they might still bind the same fragments if they share suitable subpockets. Information about shared subpockets can be therefore used in fragment-based drug design to suggest new fragments or to replace existing fragments within an already known compound. A novel computational method called SubCav is described which allows the similarity searching and alignment of subpockets from a PDB-wide database against a user-defined query. The method is based on pharmacophoric fingerprints combined with a subpocket alignment algorithm. SubCav was shown to be effective in producing reasonable alignments for subpockets with low sequence similarity and be able to retrieve relevant subpockets from a large database of structures including those with different folds. It can also be used to analyze subpockets inside a protein family to facilitate drug design and to rationalize compound selectivity.
Collapse
Affiliation(s)
- Tuomo Kalliokoski
- Novartis Institutes for Biomedical Research, Postfach, CH-4002 Basel, Switzerland
| | | | | |
Collapse
|
9
|
Local functional descriptors for surface comparison based binding prediction. BMC Bioinformatics 2012; 13:314. [PMID: 23176080 PMCID: PMC3585919 DOI: 10.1186/1471-2105-13-314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 10/10/2012] [Indexed: 11/10/2022] Open
Abstract
Background Molecular recognition in proteins occurs due to appropriate arrangements of physical, chemical, and geometric properties of an atomic surface. Similar surface regions should create similar binding interfaces. Effective methods for comparing surface regions can be used in identifying similar regions, and to predict interactions without regard to the underlying structural scaffold that creates the surface. Results We present a new descriptor for protein functional surfaces and algorithms for using these descriptors to compare protein surface regions to identify ligand binding interfaces. Our approach uses descriptors of local regions of the surface, and assembles collections of matches to compare larger regions. Our approach uses a variety of physical, chemical, and geometric properties, adaptively weighting these properties as appropriate for different regions of the interface. Our approach builds a classifier based on a training corpus of examples of binding sites of the target ligand. The constructed classifiers can be applied to a query protein providing a probability for each position on the protein that the position is part of a binding interface. We demonstrate the effectiveness of the approach on a number of benchmarks, demonstrating performance that is comparable to the state-of-the-art, with an approach with more generality than these prior methods. Conclusions Local functional descriptors offer a new method for protein surface comparison that is sufficiently flexible to serve in a variety of applications.
Collapse
|
10
|
Wood DJ, Vlieg JD, Wagener M, Ritschel T. Pharmacophore Fingerprint-Based Approach to Binding Site Subpocket Similarity and Its Application to Bioisostere Replacement. J Chem Inf Model 2012; 52:2031-43. [DOI: 10.1021/ci3000776] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- David J. Wood
- Computational Drug Discovery,
CMBI 260, NCMLS, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Jacob de Vlieg
- Computational Drug Discovery,
CMBI 260, NCMLS, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands
- MSD, Department of Molecular Design and Informatics, Molenstraat 110,
5342 CC Oss, PO Box 20, 5340 BH Oss, The Netherlands
| | - Markus Wagener
- MSD, Department of Molecular Design and Informatics, Molenstraat 110,
5342 CC Oss, PO Box 20, 5340 BH Oss, The Netherlands
| | - Tina Ritschel
- Computational Drug Discovery,
CMBI 260, NCMLS, Radboud University Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
11
|
Structure-based computational analysis of protein binding sites for function and druggability prediction. J Biotechnol 2012; 159:123-34. [DOI: 10.1016/j.jbiotec.2011.12.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Revised: 12/02/2011] [Accepted: 12/06/2011] [Indexed: 11/19/2022]
|
12
|
Wallach I. Pharmacophore inference and its application to computational drug discovery. Drug Dev Res 2010. [DOI: 10.1002/ddr.20398] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Izhar Wallach
- Department of Computer Science and the Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Sonavane S, Chakrabarti P. Prediction of active site cleft using support vector machines. J Chem Inf Model 2010; 50:2266-73. [PMID: 21080689 DOI: 10.1021/ci1002922] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Computational tools are available today for the detection and delineation of the clefts and cavities in protein 3D structure and ranking them on the basis of probable binding site clefts. There is a need to improve the ranking of clefts and accuracy of predicting catalytic site clefts. Our results show that the distance of the clefts from protein centroid and sequence entropy of the lining residues, when used in conjunction with the volume, are valuable descriptors for predicting the catalytic site. We have applied the SVM approach for recognizing and ranking the active site clefts and tested its performance using different combinations of attributes. In both the ligand-bound and the unbound forms of structures, our method correctly predicts the active site clefts in 73% of cases at rank one. If we consider the results at rank 3 (i.e., the correct solution is among one of the top three solutions), the correctly predicted cases are 94% and 90% for the bound and the unbound forms of structures, respectively. Our approach improves the ranking of binding site clefts in comparison with CASTp and is comparable to other existing methods like Fpocket. Although the data set for training the SVM approach is rather small in size, the results are encouraging for the method to be used as complementary to other existing tools.
Collapse
Affiliation(s)
- Shrihari Sonavane
- Department of Biochemistry and Bioinformatics Centre, Bose Institute, P-1/12 CIT Scheme VIIM, Kolkata 700 054, India
| | | |
Collapse
|
14
|
De Franchi E, Schalon C, Messa M, Onofri F, Benfenati F, Rognan D. Binding of protein kinase inhibitors to synapsin I inferred from pair-wise binding site similarity measurements. PLoS One 2010; 5:e12214. [PMID: 20808948 PMCID: PMC2922380 DOI: 10.1371/journal.pone.0012214] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 07/26/2010] [Indexed: 11/18/2022] Open
Abstract
Predicting off-targets by computational methods is getting increasing importance in early drug discovery stages. We herewith present a computational method based on binding site three-dimensional comparisons, which prompted us to investigate the cross-reaction of protein kinase inhibitors with synapsin I, an ATP-binding protein regulating neurotransmitter release in the synapse. Systematic pair-wise comparison of the staurosporine-binding site of the proto-oncogene Pim-1 kinase with 6,412 druggable protein-ligand binding sites suggested that the ATP-binding site of synapsin I may recognize the pan-kinase inhibitor staurosporine. Biochemical validation of this hypothesis was realized by competition experiments of staurosporine with ATP-gamma(35)S for binding to synapsin I. Staurosporine, as well as three other inhibitors of protein kinases (cdk2, Pim-1 and casein kinase type 2), effectively bound to synapsin I with nanomolar affinities and promoted synapsin-induced F-actin bundling. The selective Pim-1 kinase inhibitor quercetagetin was shown to be the most potent synapsin I binder (IC50 = 0.15 microM), in agreement with the predicted binding site similarities between synapsin I and various protein kinases. Other protein kinase inhibitors (protein kinase A and chk1 inhibitor), kinase inhibitors (diacylglycerolkinase inhibitor) and various other ATP-competitors (DNA topoisomerase II and HSP-90alpha inhibitors) did not bind to synapsin I, as predicted from a lower similarity of their respective ATP-binding sites to that of synapsin I. The present data suggest that the observed downregulation of neurotransmitter release by some but not all protein kinase inhibitors may also be contributed by a direct binding to synapsin I and phosphorylation-independent perturbation of synapsin I function. More generally, the data also demonstrate that cross-reactivity with various targets may be detected by systematic pair-wise similarity measurement of ligand-annotated binding sites.
Collapse
Affiliation(s)
- Enrico De Franchi
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Genova, Italy
| | - Claire Schalon
- Structural Chemogenomics, Laboratory of Therapeutic Innovation, CNRS UMR 7200, Université de Strasbourg, Illkirch, France
| | - Mirko Messa
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Genova, Italy
| | - Franco Onofri
- Department of Experimental Medicine, University of Genova and Istituto Nazionale di Neuroscienze, Genova, Italy
| | - Fabio Benfenati
- Department of Neuroscience and Brain Technologies, The Italian Institute of Technology, Genova, Italy
- Department of Experimental Medicine, University of Genova and Istituto Nazionale di Neuroscienze, Genova, Italy
| | - Didier Rognan
- Structural Chemogenomics, Laboratory of Therapeutic Innovation, CNRS UMR 7200, Université de Strasbourg, Illkirch, France
| |
Collapse
|
15
|
Weill N, Rognan D. Alignment-free ultra-high-throughput comparison of druggable protein-ligand binding sites. J Chem Inf Model 2010; 50:123-35. [PMID: 20058856 DOI: 10.1021/ci900349y] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Inferring the biological function of a protein from its three-dimensional structure as well as explaining why a drug may bind to various targets is of crucial importance to modern drug discovery. Here we present a generic 4833-integer vector describing druggable protein-ligand binding sites that can be applied to any protein and any binding cavity. The fingerprint registers counts of pharmacophoric triplets from the Calpha atomic coordinates of binding-site-lining residues. Starting from a customized data set of diverse protein-ligand binding site pairs, the most appropriate metric and a similarity threshold could be defined for similar binding sites. The method (FuzCav) has been used in various scenarios: (i) screening a collection of 6000 binding sites for similarity to different queries; (ii) classifying protein families (serine endopeptidases, protein kinases) by binding site diversity; (iii) discriminating adenine-binding cavities from decoys. The fingerprint generation and comparison supports ultra-high throughput (ca. 1000 measures/s), does not require prior alignment of protein binding sites, and is able to detect local similarity among subpockets. It is thus particularly well suited to the functional annotation of novel genomic structures with low sequence identity to known X-ray templates.
Collapse
Affiliation(s)
- Nathanaël Weill
- Structural Chemogenomics, Laboratory of Therapeutic Innovation, UMR 7200 CNRS-UdS (Universite de Strasbourg), F-67400 Illkirch, France
| | | |
Collapse
|