1
|
Yang J, Li Z, Wu WKK, Yu S, Xu Z, Chu Q, Zhang Q. Deep learning identifies explainable reasoning paths of mechanism of action for drug repurposing from multilayer biological network. Brief Bioinform 2022; 23:6809964. [PMID: 36347526 DOI: 10.1093/bib/bbac469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/07/2022] [Accepted: 09/29/2022] [Indexed: 11/11/2022] Open
Abstract
The discovery and repurposing of drugs require a deep understanding of the mechanism of drug action (MODA). Existing computational methods mainly model MODA with the protein-protein interaction (PPI) network. However, the molecular interactions of drugs in the human body are far beyond PPIs. Additionally, the lack of interpretability of these models hinders their practicability. We propose an interpretable deep learning-based path-reasoning framework (iDPath) for drug discovery and repurposing by capturing MODA on by far the most comprehensive multilayer biological network consisting of the complex high-dimensional molecular interactions between genes, proteins and chemicals. Experiments show that iDPath outperforms state-of-the-art machine learning methods on a general drug repurposing task. Further investigations demonstrate that iDPath can identify explicit critical paths that are consistent with clinical evidence. To demonstrate the practical value of iDPath, we apply it to the identification of potential drugs for treating prostate cancer and hypertension. Results show that iDPath can discover new FDA-approved drugs. This research provides a novel interpretable artificial intelligence perspective on drug discovery.
Collapse
Affiliation(s)
- Jiannan Yang
- School of Data Science, City University of Hong Kong, Hong Kong SAR, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shi Yu
- The USC Norris Center for Cancer Drug Development, University of Southern California, Los Angeles, CA, USA.,Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhongzhi Xu
- School of Data Science, City University of Hong Kong, Hong Kong SAR, China
| | - Qian Chu
- Department of Thoracic Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qingpeng Zhang
- School of Data Science, City University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2
|
Zhang P, Meng X, Liu L, Li S, Li Y, Ali S, Li S, Xiong J, Liu X, Li S, Xia Q, Dong L. Identification of the Prognostic Signatures of Glioma With Different PTEN Status. Front Oncol 2021; 11:633357. [PMID: 34336645 PMCID: PMC8317988 DOI: 10.3389/fonc.2021.633357] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 06/25/2021] [Indexed: 12/17/2022] Open
Abstract
The high-grade glioma is characterized by cell heterogeneity, gene mutations, and poor prognosis. The deletions and mutations of the tumor suppressor gene PTEN (5%-40%) in glioma patients are associated with worse survival and therapeutic resistance. Characterization of unique prognosis molecular signatures by PTEN status in glioma is still unclear. This study established a novel risk model, screened optimal prognostic signatures, and calculated the risk score for the individual glioma patients with different PTEN status. Screening results revealed fourteen independent prognostic gene signatures in PTEN-wt and three in the -50PTEN-mut subgroup. Moreover, we verified risk score as an independent prognostic factor significantly correlated with tumor malignancy. Due to the higher malignancy of the PTEN-mut gliomas, we explored the independent prognostic signatures (CLCF1, AEBP1, and OS9) for a potential therapeutic target in PTEN-mut glioma. We further separated IDH wild-type glioma patients into GBM and LGG to verify the therapeutic target along with PTEN status, notably, the above screened therapeutic targets are also significant prognostic genes in both IDH-wt/PTEN-mut GBM and LGG patients. We further identified the small molecule compound (+)-JQ1 binds to all three targets, indicating a potential therapy for PTEN-mut glioma. In sum, gene signatures and risk scores in the novel risk model facilitate glioma diagnosis, prognosis prediction, and treatment.
Collapse
Affiliation(s)
- Pei Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xinyi Meng
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Liqun Liu
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Shengzhen Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yang Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Sakhawat Ali
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Shanhu Li
- Department of Cell Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jichuan Xiong
- School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Xuefeng Liu
- School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Shouwei Li
- Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Qin Xia
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Lei Dong
- School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
3
|
Puspo NA, Akter L, Siddique S, Paul BK, Ahmed K, Bhuiyan T, Islam MK. Analyzing the protein-protein interaction network and the topological properties of prostate cancer and allied diseases: A computational bioinformatics approach. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
4
|
Pinzi L, Rastelli G. Identification of Target Associations for Polypharmacology from Analysis of Crystallographic Ligands of the Protein Data Bank. J Chem Inf Model 2019; 60:372-390. [PMID: 31800237 DOI: 10.1021/acs.jcim.9b00821] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The design of a chemical entity that potently and selectively binds to a biological target of therapeutic relevance has dominated the scene of drug discovery so far. However, recent findings suggest that multitarget ligands may be endowed with superior efficacy and be less prone to drug resistance. The Protein Data Bank (PDB) provides experimentally validated structural information about targets and bound ligands. Therefore, it represents a valuable source of information to help identifying active sites, understanding pharmacophore requirements, designing novel ligands, and inferring structure-activity relationships. In this study, we performed a large-scale analysis of the PDB by integrating different ligand-based and structure-based approaches, with the aim of identifying promising target associations for polypharmacology based on reported crystal structure information. First, the 2D and 3D similarity profiles of the crystallographic ligands were evaluated using different ligand-based methods. Then, activity data of pairs of similar ligands binding to different targets were inspected by comparing structural information with bioactivity annotations reported in the ChEMBL, BindingDB, BindingMOAD, and PDBbind databases. Afterward, extensive docking screenings of ligands in the identified cross-targets were made in order to validate and refine the ligand-based results. Finally, the therapeutic relevance of the identified target combinations for polypharmacology was evaluated from comparison with information on therapeutic targets reported in the Therapeutic Target Database (TTD). The results led to the identification of several target associations with high therapeutic potential for polypharmacology.
Collapse
Affiliation(s)
- Luca Pinzi
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Giuseppe Campi 103 , 41125 Modena , Italy
| | - Giulio Rastelli
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Giuseppe Campi 103 , 41125 Modena , Italy
| |
Collapse
|
5
|
Pinzi L, Caporuscio F, Rastelli G. Selection of protein conformations for structure-based polypharmacology studies. Drug Discov Today 2018; 23:1889-1896. [PMID: 30099123 DOI: 10.1016/j.drudis.2018.08.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 11/29/2022]
Abstract
Several drugs exert their therapeutic effect through the modulation of multiple targets. Structure-based approaches hold great promise for identifying compounds with the desired polypharmacological profiles. These methods use knowledge of the protein binding sites to identify stereoelectronically complementary ligands. The selection of the most suitable protein conformations to be used in the design process is vital, especially for multitarget drug design in which the same ligand has to be accommodated in multiple binding pockets. Herein, we focus on currently available techniques for the selection of the most suitable protein conformations for multitarget drug design, compare the potential advantages and limitations of each method, and comment on how their combination could help in polypharmacology drug design.
Collapse
Affiliation(s)
- Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Fabiana Caporuscio
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy.
| |
Collapse
|
6
|
Ranking Enzyme Structures in the PDB by Bound Ligand Similarity to Biological Substrates. Structure 2018; 26:565-571.e3. [PMID: 29551288 PMCID: PMC5890617 DOI: 10.1016/j.str.2018.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/26/2018] [Accepted: 02/09/2018] [Indexed: 11/22/2022]
Abstract
There are numerous applications that use the structures of protein-ligand complexes from the PDB, such as 3D pharmacophore identification, virtual screening, and fragment-based drug design. The structures underlying these applications are potentially much more informative if they contain biologically relevant bound ligands, with high similarity to the cognate ligands. We present a study of ligand-enzyme complexes that compares the similarity of bound and cognate ligands, enabling the best matches to be identified. We calculate the molecular similarity scores using a method called PARITY (proportion of atoms residing in identical topology), which can conveniently be combined to give a similarity score for all cognate reactants or products in the reaction. Thus, we generate a rank-ordered list of related PDB structures, according to the biological similarity of the ligands bound in the structures. We present PARITY, matching atoms in identical topology to gauge ligand similarity Bound-cognate ligand similarity is a useful metric for ranking PDB structures Only 26% of enzyme structures in the PDB have bound-cognate ligand similarity ≥0.7 We provide rank-ordered lists of PDBs with the most biologically relevant ligands
Collapse
|
7
|
Sam E, Athri P. Web-based drug repurposing tools: a survey. Brief Bioinform 2017; 20:299-316. [DOI: 10.1093/bib/bbx125] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Elizabeth Sam
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| | - Prashanth Athri
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| |
Collapse
|
8
|
Chen J, Xie ZR, Wu Y. Understand protein functions by comparing the similarity of local structural environments. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1865:142-152. [PMID: 27884635 DOI: 10.1016/j.bbapap.2016.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/03/2016] [Accepted: 11/17/2016] [Indexed: 12/20/2022]
Abstract
The three-dimensional structures of proteins play an essential role in regulating binding between proteins and their partners, offering a direct relationship between structures and functions of proteins. It is widely accepted that the function of a protein can be determined if its structure is similar to other proteins whose functions are known. However, it is also observed that proteins with similar global structures do not necessarily correspond to the same function, while proteins with very different folds can share similar functions. This indicates that function similarity is originated from the local structural information of proteins instead of their global shapes. We assume that proteins with similar local environments prefer binding to similar types of molecular targets. In order to testify this assumption, we designed a new structural indicator to define the similarity of local environment between residues in different proteins. This indicator was further used to calculate the probability that a given residue binds to a specific type of structural neighbors, including DNA, RNA, small molecules and proteins. After applying the method to a large-scale non-redundant database of proteins, we show that the positive signal of binding probability calculated from the local structural indicator is statistically meaningful. In summary, our studies suggested that the local environment of residues in a protein is a good indicator to recognize specific binding partners of the protein. The new method could be a potential addition to a suite of existing template-based approaches for protein function prediction.
Collapse
Affiliation(s)
- Jiawen Chen
- Department of Systems and Computational Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| | - Zhong-Ru Xie
- Department of Systems and Computational Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| |
Collapse
|
9
|
Raimondi F, Singh G, Betts MJ, Apic G, Vukotic R, Andreone P, Stein L, Russell RB. Insights into cancer severity from biomolecular interaction mechanisms. Sci Rep 2016; 6:34490. [PMID: 27698488 PMCID: PMC5048291 DOI: 10.1038/srep34490] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/14/2016] [Indexed: 12/11/2022] Open
Abstract
To attain a deeper understanding of diseases like cancer, it is critical to couple genetics with biomolecular mechanisms. High-throughput sequencing has identified thousands of somatic mutations across dozens of cancers, and there is a pressing need to identify the few that are pathologically relevant. Here we use protein structure and interaction data to interrogate nonsynonymous somatic cancer mutations, identifying a set of 213 molecular interfaces (protein-protein, -small molecule or -nucleic acid) most often perturbed in cancer, highlighting several potentially novel cancer genes. Over half of these interfaces involve protein-small-molecule interactions highlighting their overall importance in cancer. We found distinct differences in the predominance of perturbed interfaces between cancers and histological subtypes and presence or absence of certain interfaces appears to correlate with cancer severity.
Collapse
Affiliation(s)
- Francesco Raimondi
- CellNetworks, Bioquant, Im Neuenheimer Feld 267, University of Heidelberg, 69120 Heidelberg, Germany
- Biochemie Zentrum Heidelberg, Im Neuenheimer Feld 328, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gurdeep Singh
- CellNetworks, Bioquant, Im Neuenheimer Feld 267, University of Heidelberg, 69120 Heidelberg, Germany
- Biochemie Zentrum Heidelberg, Im Neuenheimer Feld 328, University of Heidelberg, 69120 Heidelberg, Germany
| | - Matthew J. Betts
- CellNetworks, Bioquant, Im Neuenheimer Feld 267, University of Heidelberg, 69120 Heidelberg, Germany
- Biochemie Zentrum Heidelberg, Im Neuenheimer Feld 328, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gordana Apic
- CellNetworks, Bioquant, Im Neuenheimer Feld 267, University of Heidelberg, 69120 Heidelberg, Germany
- Cambridge Cell Networks, St. John’s Innovation Centre, Cowley Road, Cambridge CB4 0WS, UK
| | - Ranka Vukotic
- Department of Medical and Surgical Sciences, University of Bologna and Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant’Orsola Malpighi, 40138 Bologna, Italy
| | - Pietro Andreone
- Department of Medical and Surgical Sciences, University of Bologna and Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant’Orsola Malpighi, 40138 Bologna, Italy
| | - Lincoln Stein
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Robert B. Russell
- CellNetworks, Bioquant, Im Neuenheimer Feld 267, University of Heidelberg, 69120 Heidelberg, Germany
- Biochemie Zentrum Heidelberg, Im Neuenheimer Feld 328, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Chen YC, Tolbert R, Aronov AM, McGaughey G, Walters WP, Meireles L. Prediction of Protein Pairs Sharing Common Active Ligands Using Protein Sequence, Structure, and Ligand Similarity. J Chem Inf Model 2016; 56:1734-45. [PMID: 27559831 DOI: 10.1021/acs.jcim.6b00118] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We benchmarked the ability of comparative computational approaches to correctly discriminate protein pairs sharing a common active ligand (positive protein pairs) from protein pairs with no common active ligands (negative protein pairs). Since the target and the off-targets of a drug share at least a common ligand, i.e., the drug itself, the prediction of positive protein pairs may help identify off-targets. We evaluated representative protein-centric and ligand-centric approaches, including (1) 2D and 3D ligand similarity, (2) several measures of protein sequence similarity in conjunction with different sequence sources (e.g., full protein sequence versus binding site residues), and (3) a newly described pocket shape similarity and alignment program called SiteHopper. While the sequence-based alignment of pocket residues achieved the best overall performance, SiteHopper outperformed sequence-based approaches for unrelated proteins with only 20-30% pocket residue identity. Analogously, among ligand-centric approaches, path-based fingerprints achieved the best overall performance, but ROCS-based ligand shape similarity outperformed path-based fingerprints for structurally dissimilar ligands (Tanimoto 25%-40%). A significant drop in recognition performance was observed for ligand-centric approaches when PDB ligands were used instead of ChEMBL ligands. Finally, we analyzed the relationship between pocket shape and ligand shape in our data set and found that similar ligands tend to bind to similar pockets while similar pockets may accept a range of different-shaped ligands.
Collapse
Affiliation(s)
- Yu-Chen Chen
- Vertex Pharmaceuticals Incorporated , 11010 Torreyana Road, San Diego, California 92121, United States
| | - Robert Tolbert
- OpenEye Scientific Software , 9 Bisbee Court, Suite D, Santa Fe, New Mexico 87508, United States
| | - Alex M Aronov
- Vertex Pharmaceuticals Incorporated , 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Georgia McGaughey
- Vertex Pharmaceuticals Incorporated , 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - W Patrick Walters
- Vertex Pharmaceuticals Incorporated , 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Lidio Meireles
- Vertex Pharmaceuticals Incorporated , 11010 Torreyana Road, San Diego, California 92121, United States
| |
Collapse
|
11
|
Rebollo-Lopez MJ, Lelièvre J, Alvarez-Gomez D, Castro-Pichel J, Martínez-Jiménez F, Papadatos G, Kumar V, Colmenarejo G, Mugumbate G, Hurle M, Barroso V, Young RJ, Martinez-Hoyos M, González del Río R, Bates RH, Lopez-Roman EM, Mendoza-Losana A, Brown JR, Alvarez-Ruiz E, Marti-Renom MA, Overington JP, Cammack N, Ballell L, Barros-Aguire D. Release of 50 new, drug-like compounds and their computational target predictions for open source anti-tubercular drug discovery. PLoS One 2015; 10:e0142293. [PMID: 26642067 PMCID: PMC4671658 DOI: 10.1371/journal.pone.0142293] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/19/2015] [Indexed: 12/12/2022] Open
Abstract
As a follow up to the antimycobacterial screening exercise and the release of GSK´s first Tres Cantos Antimycobacterial Set (TCAMS-TB), this paper presents the results of a second antitubercular screening effort of two hundred and fifty thousand compounds recently added to the GSK collection. The compounds were further prioritized based on not only antitubercular potency but also on physicochemical characteristics. The 50 most attractive compounds were then progressed for evaluation in three different predictive computational biology algorithms based on structural similarity or GSK historical biological assay data in order to determine their possible mechanisms of action. This effort has resulted in the identification of novel compounds and their hypothesized targets that will hopefully fuel future TB drug discovery and target validation programs alike.
Collapse
Affiliation(s)
| | - Joël Lelièvre
- Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
- * E-mail: (JL); (MAMR)
| | | | - Julia Castro-Pichel
- Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Francisco Martínez-Jiménez
- Genome Biology Group, Centre Nacional d’Anàlisi Genòmica (CNAG), Barcelona, Spain
- Gene Regulation Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - George Papadatos
- European Molecular Biology Laboratory–European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, United Kingdom
| | - Vinod Kumar
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Gonzalo Colmenarejo
- Centro de Investigación Básica, CSci Computational Chemistry, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Grace Mugumbate
- European Molecular Biology Laboratory–European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, United Kingdom
| | - Mark Hurle
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Vanessa Barroso
- Centro de Investigación Básica, Platform Technology & Science, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Rob J. Young
- CSC Medicinal Chemistry, Medicines Research Centre, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
| | | | | | - Robert H. Bates
- Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | | | | | - James R. Brown
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Emilio Alvarez-Ruiz
- Centro de Investigación Básica, Platform Technology & Science, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Marc A. Marti-Renom
- Genome Biology Group, Centre Nacional d’Anàlisi Genòmica (CNAG), Barcelona, Spain
- Gene Regulation Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- * E-mail: (JL); (MAMR)
| | - John P. Overington
- European Molecular Biology Laboratory–European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, United Kingdom
| | - Nicholas Cammack
- Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Lluís Ballell
- Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - David Barros-Aguire
- Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| |
Collapse
|
12
|
A fast topological analysis algorithm for large-scale similarity evaluations of ligands and binding pockets. J Cheminform 2015; 7:42. [PMID: 26561508 PMCID: PMC4631714 DOI: 10.1186/s13321-015-0091-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/22/2015] [Indexed: 11/10/2022] Open
Abstract
Motivation With the rapid increase of the structural data of biomolecular complexes, novel structural analysis methods have to be devised with high-throughput capacity to handle immense data input and to construct massive networks at the minimal computational cost. Moreover, novel methods should be capable of handling a broad range of molecular structural sizes and chemical natures, cognisant of the conformational and electrostatic bases of molecular recognition, and sufficiently accurate to enable contextually relevant biological inferences. Results A novel molecular topology comparison method was developed and tested. The method was tested for both ligand and binding pocket similarity analyses and a PDB-wide ligand topological similarity map was computed. Conclusion The unprecedentedly wide scope of ligand definition and large-scale topological similarity mapping can provide very robust tools, of performance unmatched by the present alignment-based methods. The method remarkably shows potential for application for scaffold hopping purposes. It also opens new frontiers in the areas of ligand-mediated protein connectivity, ligand-based molecular phylogeny, target fishing, and off-target predictions. Electronic supplementary material The online version of this article (doi:10.1186/s13321-015-0091-5) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Martínez-Jiménez F, Marti-Renom MA. Ligand-target prediction by structural network biology using nAnnoLyze. PLoS Comput Biol 2015; 11:e1004157. [PMID: 25816344 PMCID: PMC4376866 DOI: 10.1371/journal.pcbi.1004157] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/27/2015] [Indexed: 11/24/2022] Open
Abstract
Target identification is essential for drug design, drug-drug interaction prediction, dosage adjustment and side effect anticipation. Specifically, the knowledge of structural details is essential for understanding the mode of action of a compound on a target protein. Here, we present nAnnoLyze, a method for target identification that relies on the hypothesis that structurally similar binding sites bind similar ligands. nAnnoLyze integrates structural information into a bipartite network of interactions and similarities to predict structurally detailed compound-protein interactions at proteome scale. The method was benchmarked on a dataset of 6,282 pairs of known interacting ligand-target pairs reaching a 0.96 of area under the Receiver Operating Characteristic curve (AUC) when using the drug names as an input feature for the classifier, and a 0.70 of AUC for “anonymous” compounds or compounds not present in the training set. nAnnoLyze resulted in higher accuracies than its predecessor, AnnoLyze. We applied the method to predict interactions for all the compounds in the DrugBank database with each human protein structure and provide examples of target identification for known drugs against human diseases. The accuracy and applicability of our method to any compound indicate that a comparative docking approach such as nAnnoLyze enables large-scale annotation and analysis of compound–protein interactions and thus may benefit drug development. Description of the “mode-of-action” of a small chemical compound against a protein target is essential for the drug discovery process. Such description relies on three main steps: i) the identification of the target protein within the thousands of proteins in an organism, ii) the localization of the binding interaction site in the identified target protein, and iii) the molecular characterization of the compound’s binding mode in the binding site of the target protein. Here, we introduce a new computational method, called nAnnoLyze, which uses graph theory principles to relate compounds and target proteins based on comparative principles. nAnnoLyze aims at addressing two of the three previous steps, that is, target identification and binding site localization. Our results suggest that the nAnnoLyze accuracy and proteome-wide applicability enables the large-scale annotation and analysis of compound–protein interaction and thus may benefit drug development.
Collapse
Affiliation(s)
- Francisco Martínez-Jiménez
- Genome Biology Group, Centre Nacional d’Aanàlisi Genòmica (CNAG), Barcelona, Spain
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Marc A. Marti-Renom
- Genome Biology Group, Centre Nacional d’Aanàlisi Genòmica (CNAG), Barcelona, Spain
- Gene Regulation, Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- * E-mail:
| |
Collapse
|
14
|
Cortés-Ciriano I, Ain QU, Subramanian V, Lenselink EB, Méndez-Lucio O, IJzerman AP, Wohlfahrt G, Prusis P, Malliavin TE, van Westen GJP, Bender A. Polypharmacology modelling using proteochemometrics (PCM): recent methodological developments, applications to target families, and future prospects. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00216d] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Proteochemometric (PCM) modelling is a computational method to model the bioactivity of multiple ligands against multiple related protein targets simultaneously.
Collapse
Affiliation(s)
- Isidro Cortés-Ciriano
- Unité de Bioinformatique Structurale
- Institut Pasteur and CNRS UMR 3825
- Structural Biology and Chemistry Department
- 75 724 Paris
- France
| | - Qurrat Ul Ain
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| | | | - Eelke B. Lenselink
- Division of Medicinal Chemistry
- Leiden Academic Centre for Drug Research
- Leiden
- The Netherlands
| | - Oscar Méndez-Lucio
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry
- Leiden Academic Centre for Drug Research
- Leiden
- The Netherlands
| | - Gerd Wohlfahrt
- Computer-Aided Drug Design
- Orion Pharma
- FIN-02101 Espoo
- Finland
| | - Peteris Prusis
- Computer-Aided Drug Design
- Orion Pharma
- FIN-02101 Espoo
- Finland
| | - Thérèse E. Malliavin
- Unité de Bioinformatique Structurale
- Institut Pasteur and CNRS UMR 3825
- Structural Biology and Chemistry Department
- 75 724 Paris
- France
| | - Gerard J. P. van Westen
- European Molecular Biology Laboratory
- European Bioinformatics Institute
- Wellcome Trust Genome Campus
- Hinxton
- UK
| | - Andreas Bender
- Unilever Centre for Molecular Informatics
- Department of Chemistry
- CB2 1EW Cambridge
- UK
| |
Collapse
|
15
|
Betts MJ, Lu Q, Jiang Y, Drusko A, Wichmann O, Utz M, Valtierra-Gutiérrez IA, Schlesner M, Jaeger N, Jones DT, Pfister S, Lichter P, Eils R, Siebert R, Bork P, Apic G, Gavin AC, Russell RB. Mechismo: predicting the mechanistic impact of mutations and modifications on molecular interactions. Nucleic Acids Res 2014; 43:e10. [PMID: 25392414 PMCID: PMC4333368 DOI: 10.1093/nar/gku1094] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Systematic interrogation of mutation or protein modification data is important to identify sites with functional consequences and to deduce global consequences from large data sets. Mechismo (mechismo.russellab.org) enables simultaneous consideration of thousands of 3D structures and biomolecular interactions to predict rapidly mechanistic consequences for mutations and modifications. As useful functional information often only comes from homologous proteins, we benchmarked the accuracy of predictions as a function of protein/structure sequence similarity, which permits the use of relatively weak sequence similarities with an appropriate confidence measure. For protein–protein, protein–nucleic acid and a subset of protein–chemical interactions, we also developed and benchmarked a measure of whether modifications are likely to enhance or diminish the interactions, which can assist the detection of modifications with specific effects. Analysis of high-throughput sequencing data shows that the approach can identify interesting differences between cancers, and application to proteomics data finds potential mechanistic insights for how post-translational modifications can alter biomolecular interactions.
Collapse
Affiliation(s)
- Matthew J Betts
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Qianhao Lu
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - YingYing Jiang
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Armin Drusko
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Oliver Wichmann
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Mathias Utz
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Ilse A Valtierra-Gutiérrez
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Matthias Schlesner
- Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Natalie Jaeger
- Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - David T Jones
- Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Stefan Pfister
- Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Peter Lichter
- Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Roland Eils
- Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology (IPMB), University of Heidelberg, Heidelberg, Germany
| | - Reiner Siebert
- Institut für Humangenetik, Universitätsklinikum Schleswig-Holstein, Christian-Albrechts-Universität zu Kiel, Arnold Heller Straße 3, 24105 Kiel, Germany
| | - Peer Bork
- EMBL, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Gordana Apic
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Cambridge Cell Networks Ltd, St John's Innovation Centre, Cowley Road, CB3 0WS, Cambridge, UK
| | | | - Robert B Russell
- Cell Networks, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany Bioquant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Acharya C, Kufareva I, Ilatovskiy AV, Abagyan R. PeptiSite: a structural database of peptide binding sites in 4D. Biochem Biophys Res Commun 2014; 445:717-23. [PMID: 24406170 DOI: 10.1016/j.bbrc.2013.12.132] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/26/2013] [Indexed: 12/11/2022]
Abstract
We developed PeptiSite, a comprehensive and reliable database of biologically and structurally characterized peptide-binding sites, in which each site is represented by an ensemble of its complexes with protein, peptide and small molecule partners. The unique features of the database include: (1) the ensemble site representation that provides a fourth dimension to the otherwise three dimensional data, (2) comprehensive characterization of the binding site architecture that may consist of a multimeric protein assembly with cofactors and metal ions and (3) analysis of consensus interaction motifs within the ensembles and identification of conserved determinants of these interactions. Currently the database contains 585 proteins with 650 peptide-binding sites. http://peptisite.ucsd.edu/ link allows searching for the sites of interest and interactive visualization of the ensembles using the ActiveICM web-browser plugin. This structural database for protein-peptide interactions enables understanding of structural principles of these interactions and may assist the development of an efficient peptide docking benchmark.
Collapse
Affiliation(s)
- Chayan Acharya
- UCSD, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA
| | - Irina Kufareva
- UCSD, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA
| | - Andrey V Ilatovskiy
- UCSD, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA; Division of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute, Gatchina 188300, Russia; Research and Education Center "Biophysics", PNPI and St. Petersburg State Polytechnical University, St. Petersburg 195251, Russia
| | - Ruben Abagyan
- UCSD, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA.
| |
Collapse
|
17
|
Casado-Vela J, Fuentes M, Franco-Zorrilla JM. Screening of Protein–Protein and Protein–DNA Interactions Using Microarrays. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 95:231-81. [DOI: 10.1016/b978-0-12-800453-1.00008-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Martínez-Jiménez F, Papadatos G, Yang L, Wallace IM, Kumar V, Pieper U, Sali A, Brown JR, Overington JP, Marti-Renom MA. Target prediction for an open access set of compounds active against Mycobacterium tuberculosis. PLoS Comput Biol 2013; 9:e1003253. [PMID: 24098102 PMCID: PMC3789770 DOI: 10.1371/journal.pcbi.1003253] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/11/2013] [Indexed: 01/01/2023] Open
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), infects an estimated two billion people worldwide and is the leading cause of mortality due to infectious disease. The development of new anti-TB therapeutics is required, because of the emergence of multi-drug resistance strains as well as co-infection with other pathogens, especially HIV. Recently, the pharmaceutical company GlaxoSmithKline published the results of a high-throughput screen (HTS) of their two million compound library for anti-mycobacterial phenotypes. The screen revealed 776 compounds with significant activity against the M. tuberculosis H37Rv strain, including a subset of 177 prioritized compounds with high potency and low in vitro cytotoxicity. The next major challenge is the identification of the target proteins. Here, we use a computational approach that integrates historical bioassay data, chemical properties and structural comparisons of selected compounds to propose their potential targets in M. tuberculosis. We predicted 139 target - compound links, providing a necessary basis for further studies to characterize the mode of action of these compounds. The results from our analysis, including the predicted structural models, are available to the wider scientific community in the open source mode, to encourage further development of novel TB therapeutics. Mycobacterium tuberculosis is a major worldwide pathogen infecting millions individuals every year. Additionally, the number of antibiotic resistant strains has dramatically increased over the last decades. Trying to address this challenge, the pharmaceutical company GlaxoSmithKline has recently published the results of a large-scale high-throughput screen (HTS) that resulted in the release of 776 chemical compound structures active against tuberculosis. We have used this dataset of compounds as input to our computational approach that integrates historical bioassay data, chemical properties and structural comparisons. We propose 139 targets alongside their respective hit compounds and made them open to the wider scientific community. Our hope is that the availability of the experimental data from GSK and our computational analysis will encourage further research providing validated therapeutically targets against this devastating disease.
Collapse
Affiliation(s)
- Francisco Martínez-Jiménez
- Genome Biology Group, Centre Nacional d'Anàlisi Genòmica (CNAG), Barcelona, Spain
- Gene Regulation Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - George Papadatos
- European Molecular Biology Laboratory – European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Lun Yang
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Iain M. Wallace
- European Molecular Biology Laboratory – European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Vinod Kumar
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
| | - Ursula Pieper
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, United States of America
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, United States of America
| | - James R. Brown
- Computational Biology, Quantitative Sciences, GlaxoSmithKline, Collegeville, Pennsylvania, United States of America
- * E-mail: (JRB); (JPO); (MAMR)
| | - John P. Overington
- European Molecular Biology Laboratory – European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- * E-mail: (JRB); (JPO); (MAMR)
| | - Marc A. Marti-Renom
- Genome Biology Group, Centre Nacional d'Anàlisi Genòmica (CNAG), Barcelona, Spain
- Gene Regulation Stem Cells and Cancer Program, Centre for Genomic Regulation (CRG), Barcelona, Spain
- * E-mail: (JRB); (JPO); (MAMR)
| |
Collapse
|
19
|
Haupt VJ, Daminelli S, Schroeder M. Drug Promiscuity in PDB: Protein Binding Site Similarity Is Key. PLoS One 2013; 8:e65894. [PMID: 23805191 PMCID: PMC3689763 DOI: 10.1371/journal.pone.0065894] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 04/30/2013] [Indexed: 11/19/2022] Open
Abstract
Drug repositioning applies established drugs to new disease indications with increasing success. A pre-requisite for drug repurposing is drug promiscuity (polypharmacology) – a drug’s ability to bind to several targets. There is a long standing debate on the reasons for drug promiscuity. Based on large compound screens, hydrophobicity and molecular weight have been suggested as key reasons. However, the results are sometimes contradictory and leave space for further analysis. Protein structures offer a structural dimension to explain promiscuity: Can a drug bind multiple targets because the drug is flexible or because the targets are structurally similar or even share similar binding sites? We present a systematic study of drug promiscuity based on structural data of PDB target proteins with a set of 164 promiscuous drugs. We show that there is no correlation between the degree of promiscuity and ligand properties such as hydrophobicity or molecular weight but a weak correlation to conformational flexibility. However, we do find a correlation between promiscuity and structural similarity as well as binding site similarity of protein targets. In particular, 71% of the drugs have at least two targets with similar binding sites. In order to overcome issues in detection of remotely similar binding sites, we employed a score for binding site similarity: LigandRMSD measures the similarity of the aligned ligands and uncovers remote local similarities in proteins. It can be applied to arbitrary structural binding site alignments. Three representative examples, namely the anti-cancer drug methotrexate, the natural product quercetin and the anti-diabetic drug acarbose are discussed in detail. Our findings suggest that global structural and binding site similarity play a more important role to explain the observed drug promiscuity in the PDB than physicochemical drug properties like hydrophobicity or molecular weight. Additionally, we find ligand flexibility to have a minor influence.
Collapse
Affiliation(s)
| | | | - Michael Schroeder
- Biotechnology Center (BIOTEC), TU Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
20
|
Koch U, Hamacher M, Nussbaumer P. Cheminformatics at the interface of medicinal chemistry and proteomics. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1844:156-61. [PMID: 23707564 DOI: 10.1016/j.bbapap.2013.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 04/26/2013] [Accepted: 05/13/2013] [Indexed: 10/26/2022]
Abstract
Multiple factors have to be optimized in the course of a drug discovery project. Traditionally this includes potency on a single target, eventually specificity as well as the pharmacokinetic, physicochemical and the safety profile. Recently an additional dimension has been added by realizing that the therapeutic outcome of a drug is often determined not only by its activity on a single target but also by its activity profile across a variety of biological targets. To address the polypharmacology of drug candidates many compounds are tested on a set of targets or in phenotypic screens generating a tremendous amount of data. To extract useful information computational methods at the interface of proteomics and cheminformatics are indispensable. This review will focus on some recent developments in this field. This article is part of a Special Issue entitled: Computational Proteomics in the Post-Identification Era. Guest Editors: Martin Eisenacher and Christian Stephan.
Collapse
Affiliation(s)
- Uwe Koch
- Lead Discovery Center GmbH, Otto-Hahn-Str. 15, D-44227 Dortmund, Germany.
| | | | | |
Collapse
|
21
|
Abstract
The concept of chemoisosterism of protein environments is introduced as the complementary property to bioisosterism of chemical fragments. In the same way that two chemical fragments are considered bioisosteric if they can bind to the same protein environment, two protein environments will be considered chemoisosteric if they can interact with the same chemical fragment. The basis for the identification of chemoisosteric relationships among protein environments was the increasing amount of crystal structures available currently for protein-ligand complexes. It is shown that one can recover the right location and orientation of chemical fragments constituting the native ligand in a nuclear receptor structure by using only chemoisosteric environments present in enzyme structures. Examples of the potential applicability of chemoisosterism in fragment-based drug discovery are provided.
Collapse
Affiliation(s)
- Xavier Jalencas
- Chemogenomics Laboratory, Research Programme on Biomedical Informatics (GRIB), IMIM Hospital del Mar Research Institute and University Pompeu Fabra, Doctor Aiguader 88, 08003 Barcelona, Catalonia, Spain
| | | |
Collapse
|
22
|
Kufareva I, Ilatovskiy AV, Abagyan R. Pocketome: an encyclopedia of small-molecule binding sites in 4D. Nucleic Acids Res 2012; 40:D535-40. [PMID: 22080553 PMCID: PMC3245087 DOI: 10.1093/nar/gkr825] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 09/18/2011] [Indexed: 11/22/2022] Open
Abstract
The importance of binding site plasticity in protein-ligand interactions is well-recognized, and so are the difficulties in predicting the nature and the degree of this plasticity by computational means. To assist in understanding the flexible protein-ligand interactions, we constructed the Pocketome, an encyclopedia of about one thousand experimentally solved conformational ensembles of druggable binding sites in proteins, grouped by location and consistent chain/cofactor composition. The multiplicity of pockets within the ensembles adds an extra, fourth dimension to the Pocketome entry data. Within each ensemble, the pockets were carefully classified by the degree of their pairwise similarity and compatibility with different ligands. The core of the Pocketome is derived regularly and automatically from the current releases of the Protein Data Bank and the Uniprot Knowledgebase; this core is complemented by entries built from manually provided seed ligand locations. The Pocketome website (www.pocketome.org) allows searching for the sites of interest, analysis of conformational clusters, important residues, binding compatibility matrices and interactive visualization of the ensembles using the ActiveICM web browser plugin. The Pocketome collection can be used to build multi-conformational docking and 3D activity models as well as to design cross-docking and virtual ligand screening benchmarks.
Collapse
Affiliation(s)
- Irina Kufareva
- UCSD Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, 92093, USA, Division of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute, Russian Academy of Sciences, Gatchina, 188300 and Research and Education Center ‘Biophysics’, PNPI RAS and St. Petersburg State Polytechnical University, St. Petersburg, 194064, Russia
| | - Andrey V. Ilatovskiy
- UCSD Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, 92093, USA, Division of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute, Russian Academy of Sciences, Gatchina, 188300 and Research and Education Center ‘Biophysics’, PNPI RAS and St. Petersburg State Polytechnical University, St. Petersburg, 194064, Russia
| | - Ruben Abagyan
- UCSD Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, 92093, USA, Division of Molecular and Radiation Biophysics, Petersburg Nuclear Physics Institute, Russian Academy of Sciences, Gatchina, 188300 and Research and Education Center ‘Biophysics’, PNPI RAS and St. Petersburg State Polytechnical University, St. Petersburg, 194064, Russia
| |
Collapse
|
23
|
Kalinina OV, Wichmann O, Apic G, Russell RB. ProtChemSI: a network of protein-chemical structural interactions. Nucleic Acids Res 2011; 40:D549-53. [PMID: 22110041 PMCID: PMC3245083 DOI: 10.1093/nar/gkr1049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Progress in structure determination methods means that the set of experimentally determined 3D structures of proteins in complex with small molecules is growing exponentially. ProtChemSI exploits and extends this useful set of structures by both collecting and annotating the existing data as well as providing models of potential complexes inferred by protein or chemical structure similarity. The database currently includes 7704 proteins from 1803 organisms, 11 324 chemical compounds and 202 289 complexes including 178 974 predicted. It is publicly available at http://pcidb.russelllab.org.
Collapse
Affiliation(s)
- Olga V Kalinina
- Cell Networks, BioQuant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
24
|
Kuhn M, Szklarczyk D, Franceschini A, von Mering C, Jensen LJ, Bork P. STITCH 3: zooming in on protein-chemical interactions. Nucleic Acids Res 2011; 40:D876-80. [PMID: 22075997 PMCID: PMC3245073 DOI: 10.1093/nar/gkr1011] [Citation(s) in RCA: 202] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
To facilitate the study of interactions between proteins and chemicals, we have created STITCH, an aggregated database of interactions connecting over 300 000 chemicals and 2.6 million proteins from 1133 organisms. Compared to the previous version, the number of chemicals with interactions and the number of high-confidence interactions both increase 4-fold. The database can be accessed interactively through a web interface, displaying interactions in an integrated network view. It is also available for computational studies through downloadable files and an API. As an extension in the current version, we offer the option to switch between two levels of detail, namely whether stereoisomers of a given compound are shown as a merged entity or as separate entities. Separate display of stereoisomers is necessary, for example, for carbohydrates and chiral drugs. Combining the isomers increases the coverage, as interaction databases and publications found through text mining will often refer to compounds without specifying the stereoisomer. The database is accessible at http://stitch.embl.de/.
Collapse
Affiliation(s)
- Michael Kuhn
- Biotechnology Center, TU Dresden, 01062 Dresden, Germany.
| | | | | | | | | | | |
Collapse
|