1
|
Milon TI, Sarkar T, Chen Y, Grider JM, Chen F, Ji JY, Jois SD, Kousoulas KG, Raghavan V, Xu W. Development of the TSR-based computational method to investigate spike and monoclonal antibody interactions. Front Chem 2025; 13:1395374. [PMID: 40177350 PMCID: PMC11962798 DOI: 10.3389/fchem.2025.1395374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Monoclonal antibody (mAb) drug treatments have proven effective in reducing COVID-19-related hospitalizations or fatalities, particularly among high-risk patients. Numerous experimental studies have explored the structures of spike proteins and their complexes with ACE2 or mAbs. These 3D structures provide crucial insights into the interactions between spike proteins and ACE2 or mAb, forming a basis for the development of diagnostic tools and therapeutics. However, the field of computational biology has faced substantial challenges due to the lack of methods for precise protein structural comparisons and accurate prediction of molecular interactions. In our previous studies, we introduced the Triangular Spatial Relationship (TSR)-based algorithm, which represents a protein's 3D structure using a vector of integers (keys). These earlier studies, however, were limited to individual proteins. Purpose This study introduces new extensions of the TSR-based algorithm, enhancing its ability to study interactions between two molecules. We apply these extensions to gain a mechanistic understanding of spike - mAb interactions. Method We expanded the basic TSR method in three novel ways: (1) TSR keys encompassing all atoms, (2) cross keys for interactions between two molecules, and (3) intra-residual keys for amino acids. This TSR-based representation of 3D structures offers a unique advantage by simplifying the search for similar substructures within structural datasets. Results The study's key findings include: (i) The method effectively quantified and interpreted conformational changes and steric effects using the newly introduced TSR keys. (ii) Six clusters for CDRH3 and three clusters for CDRL3 were identified using all-atom keys. (iii) We constructed the TSR-STRSUM (TSR-STRucture SUbstitution Matrix), a matrix that represents pairwise similarities between amino acid structures, providing valuable applications in protein sequence and structure comparison. (iv) Intra-residual keys revealed two distinct Tyr clusters characterized by specific triangle geometries. Conclusion This study presents an advanced computational approach that not only quantifies and interprets conformational changes in protein backbones, entire structures, or individual amino acids, but also facilitates the search for substructures induced by molecular binding across protein datasets. In some instances, a direct correlation between structures and functions was successfully established.
Collapse
Affiliation(s)
- Tarikul I. Milon
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, United States
| | - Titli Sarkar
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, United States
- The Center for Advanced Computer Studies, University of Louisiana at Lafayette, Lafayette, LA, United States
| | - Yixin Chen
- Department of Computer and Information Science, The University of Mississippi, University, MS, United States
| | - Jordan M. Grider
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, United States
| | - Feng Chen
- High Performance Computing, 329 Frey Computing Services Center, Louisiana State University, Baton Rouge, LA, United States
| | - Jun-Yuan Ji
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA, United States
| | - Seetharama D. Jois
- Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Konstantin G. Kousoulas
- Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Vijay Raghavan
- The Center for Advanced Computer Studies, University of Louisiana at Lafayette, Lafayette, LA, United States
| | - Wu Xu
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, United States
| |
Collapse
|
2
|
Olechnovič K, Banciul R, Dapkūnas J, Venclovas Č. FTDMP: A Framework for Protein-Protein, Protein-DNA, and Protein-RNA Docking and Scoring. Proteins 2025. [PMID: 39748638 DOI: 10.1002/prot.26792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
FTDMP is a software framework for biomolecular docking and scoring. It can perform docking of subunits containing one or more protein, DNA, or RNA chains, followed by subsequent scoring of the resulting models. FTDMP can also be used for the ranking of user-provided models of biomolecular complexes, generated by any structure prediction method. FTDMP evaluates models according to the consensus-based method VoroIF-jury, which combines individual scores derived from the Voronoi tessellation of biomolecular structures. In addition to the default scoring mode, FTDMP can easily adopt additional scores; thus, it may be used as a tool to assess newly developed scoring functions. FTDMP was evaluated during blind testing in recent CAPRI experiments and using protein-protein, protein-DNA, and protein-RNA docking benchmarks. It proved to be a useful tool for different research tasks, related to modeling biomolecular interactions. The software, cleaned docking benchmarks, and benchmarking results are available at https://bioinformatics.lt/software/ftdmp/.
Collapse
Affiliation(s)
- Kliment Olechnovič
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Université Grenoble Alpes, CNRS, Grenoble INP, LJK, Grenoble, France
| | - Rita Banciul
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Justas Dapkūnas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Česlovas Venclovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
3
|
Viswanathan R, Carroll M, Roffe A, Fajardo JE, Fiser A. Computational prediction of multiple antigen epitopes. Bioinformatics 2024; 40:btae556. [PMID: 39271143 PMCID: PMC11453099 DOI: 10.1093/bioinformatics/btae556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 08/08/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024] Open
Abstract
MOTIVATION Identifying antigen epitopes is essential in medical applications, such as immunodiagnostic reagent discovery, vaccine design, and drug development. Computational approaches can complement low-throughput, time-consuming, and costly experimental determination of epitopes. Currently available prediction methods, however, have moderate success predicting epitopes, which limits their applicability. Epitope prediction is further complicated by the fact that multiple epitopes may be located on the same antigen and complete experimental data is often unavailable. RESULTS Here, we introduce the antigen epitope prediction program ISPIPab that combines information from two feature-based methods and a docking-based method. We demonstrate that ISPIPab outperforms each of its individual classifiers as well as other state-of-the-art methods, including those designed specifically for epitope prediction. By combining the prediction algorithm with hierarchical clustering, we show that we can effectively capture epitopes that align with available experimental data while also revealing additional novel targets for future experimental investigations.
Collapse
Affiliation(s)
- Rajalakshmi Viswanathan
- Department of Chemistry and Biochemistry, Yeshiva College, New York, NY 10033, United States
| | - Moshe Carroll
- Department of Chemistry and Biochemistry, Yeshiva College, New York, NY 10033, United States
| | - Alexandra Roffe
- Department of Chemistry and Biochemistry, Stern College for Women, New York, NY 10016, United States
| | - Jorge E Fajardo
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| |
Collapse
|
4
|
Carroll M, Rosenbaum E, Viswanathan R. Computational Methods to Predict Conformational B-Cell Epitopes. Biomolecules 2024; 14:983. [PMID: 39199371 PMCID: PMC11352882 DOI: 10.3390/biom14080983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
Accurate computational prediction of B-cell epitopes can greatly enhance biomedical research and rapidly advance efforts to develop therapeutics, monoclonal antibodies, vaccines, and immunodiagnostic reagents. Previous research efforts have primarily focused on the development of computational methods to predict linear epitopes rather than conformational epitopes; however, the latter is much more biologically predominant. Several conformational B-cell epitope prediction methods have recently been published, but their predictive performances are weak. Here, we present a review of the latest computational methods and assess their performances on a diverse test set of 29 non-redundant unbound antigen structures. Our results demonstrate that ISPIPab performs better than most methods and compares favorably with other recent antigen-specific methods. Finally, we suggest new strategies and opportunities to improve computational predictions of conformational B-cell epitopes.
Collapse
Affiliation(s)
| | | | - R. Viswanathan
- Department of Chemistry and Biochemistry, Yeshiva College, Yeshiva University, New York, NY 10033, USA; (M.C.); (E.R.)
| |
Collapse
|
5
|
Viswanathan R, Carroll M, Roffe A, Fajardo JE, Fiser A. Computational Prediction of Multiple Antigen Epitopes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607232. [PMID: 39211281 PMCID: PMC11360938 DOI: 10.1101/2024.08.08.607232] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Motivation Identifying antigen epitopes is essential in medical applications, such as immunodiagnostic reagent discovery, vaccine design, and drug development. Computational approaches can complement low-throughput, time-consuming, and costly experimental determination of epitopes. Currently available prediction methods, however, have moderate success predicting epitopes, which limits their applicability. Epitope prediction is further complicated by the fact that multiple epitopes may be located on the same antigen and complete experimental data is often unavailable. Results Here, we introduce the antigen epitope prediction program ISPIPab that combines information from two feature-based methods and a docking-based method. We demonstrate that ISPIPab outperforms each of its individual classifiers as well as other state-of-the-art methods, including those designed specifically for epitope prediction. By combining the prediction algorithm with hierarchical clustering, we show that we can effectively capture epitopes that align with available experimental data while also revealing additional novel targets for future experimental investigations. Contact raji@yu.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
|
6
|
Nandigrami P, Fiser A. Assessing the functional impact of protein binding site definition. Protein Sci 2024; 33:e5026. [PMID: 38757384 PMCID: PMC11099757 DOI: 10.1002/pro.5026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024]
Abstract
Many biomedical applications, such as classification of binding specificities or bioengineering, depend on the accurate definition of protein binding interfaces. Depending on the choice of method used, substantially different sets of residues can be classified as belonging to the interface of a protein. A typical approach used to verify these definitions is to mutate residues and measure the impact of these changes on binding. Besides the lack of exhaustive data, this approach also suffers from the fundamental problem that a mutation introduces an unknown amount of alteration into an interface, which potentially alters the binding characteristics of the interface. In this study we explore the impact of alternative binding site definitions on the ability of a protein to recognize its cognate ligand using a pharmacophore approach, which does not affect the interface. The study also shows that methods for protein binding interface predictions should perform above approximately F-score = 0.7 accuracy level to capture the biological function of a protein.
Collapse
Affiliation(s)
- Prithviraj Nandigrami
- Departments of Systems and Computational Biology, and BiochemistryAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Andras Fiser
- Departments of Systems and Computational Biology, and BiochemistryAlbert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
7
|
Grudman S, Fajardo JE, Fiser A. Optimal selection of suitable templates in protein interface prediction. Bioinformatics 2023; 39:btad510. [PMID: 37603727 PMCID: PMC10491951 DOI: 10.1093/bioinformatics/btad510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/11/2023] [Accepted: 08/18/2023] [Indexed: 08/23/2023] Open
Abstract
MOTIVATION Molecular-level classification of protein-protein interfaces can greatly assist in functional characterization and rational drug design. The most accurate protein interface predictions rely on finding homologous proteins with known interfaces since most interfaces are conserved within the same protein family. The accuracy of these template-based prediction approaches depends on the correct choice of suitable templates. Choosing the right templates in the immunoglobulin superfamily (IgSF) is challenging because its members share low sequence identity and display a wide range of alternative binding sites despite structural homology. RESULTS We present a new approach to predict protein interfaces. First, template-specific, informative evolutionary profiles are established using a mutual information-based approach. Next, based on the similarity of residue level conservation scores derived from the evolutionary profiles, a query protein is hierarchically clustered with all available template proteins in its superfamily with known interface definitions. Once clustered, a subset of the most closely related templates is selected, and an interface prediction is made. These initial interface predictions are subsequently refined by extensive docking. This method was benchmarked on 51 IgSF proteins and can predict nontrivial interfaces of IgSF proteins with an average and median F-score of 0.64 and 0.78, respectively. We also provide a way to assess the confidence of the results. The average and median F-scores increase to 0.8 and 0.81, respectively, if 27% of low confidence cases and 17% of medium confidence cases are removed. Lastly, we provide residue level interface predictions, protein complexes, and confidence measurements for singletons in the IgSF. AVAILABILITY AND IMPLEMENTATION Source code is freely available at: https://gitlab.com/fiserlab.org/interdct_with_refinement.
Collapse
Affiliation(s)
- Steven Grudman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - J Eduardo Fajardo
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
8
|
Nandigrami P, Fiser A. Assessing the functional impact of protein binding site definition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525812. [PMID: 36747792 PMCID: PMC9900911 DOI: 10.1101/2023.01.26.525812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Many biomedical applications, such as classification of binding specificities or bioengineering, depend on the accurate definition of protein binding interfaces. Depending on the choice of method used, substantially different sets of residues can be classified as belonging to the interface of a protein. A typical approach used to verify these definitions is to mutate residues and measure the impact of these changes on binding. Besides the lack of exhaustive data this approach generates, it also suffers from the fundamental problem that a mutation introduces an unknown amount of alteration into an interface, which potentially alters the binding characteristics of the interface. In this study we explore the impact of alternative binding site definitions on the ability of a protein to recognize its cognate ligand using a pharmacophore approach, which does not affect the interface. The study also provides guidance on the minimum expected accuracy of interface definition that is required to capture the biological function of a protein.
Collapse
Affiliation(s)
- Prithviraj Nandigrami
- Departments of Systems & Computational Biology, and Biochemistry, Albert Einstein College of Medicine 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - Andras Fiser
- Departments of Systems & Computational Biology, and Biochemistry, Albert Einstein College of Medicine 1300 Morris Park Ave, Bronx, NY 10461, USA
| |
Collapse
|
9
|
Walder M, Edelstein E, Carroll M, Lazarev S, Fajardo JE, Fiser A, Viswanathan R. Integrated structure-based protein interface prediction. BMC Bioinformatics 2022; 23:301. [PMID: 35879651 PMCID: PMC9316365 DOI: 10.1186/s12859-022-04852-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022] Open
Abstract
Background Identifying protein interfaces can inform how proteins interact with their binding partners, uncover the regulatory mechanisms that control biological functions and guide the development of novel therapeutic agents. A variety of computational approaches have been developed for predicting a protein’s interfacial residues from its known sequence and structure. Methods using the known three-dimensional structures of proteins can be template-based or template-free. Template-based methods have limited success in predicting interfaces when homologues with known complex structures are not available to use as templates. The prediction performance of template-free methods that only rely only upon proteins’ intrinsic properties is limited by the amount of biologically relevant features that can be included in an interface prediction model. Results We describe the development of an integrated method for protein interface prediction (ISPIP) to explore the hypothesis that the efficacy of a computational prediction method of protein binding sites can be enhanced by using a combination of methods that rely on orthogonal structure-based properties of a query protein, combining and balancing both template-free and template-based features. ISPIP is a method that integrates these approaches through simple linear or logistic regression models and more complex decision tree models. On a diverse test set of 156 query proteins, ISPIP outperforms each of its individual classifiers in identifying protein binding interfaces. Conclusions The integrated method captures the best performance of individual classifiers and delivers an improved interface prediction. The method is robust and performs well even when one of the individual classifiers performs poorly on a particular query protein. This work demonstrates that integrating orthogonal methods that depend on different structural properties of proteins performs better at interface prediction than any individual classifier alone. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04852-2.
Collapse
Affiliation(s)
- M Walder
- Department of Chemistry, Yeshiva College, Yeshiva University, New York, NY, 10033, USA
| | - E Edelstein
- Department of Chemistry, Yeshiva College, Yeshiva University, New York, NY, 10033, USA
| | - M Carroll
- Department of Chemistry, Yeshiva College, Yeshiva University, New York, NY, 10033, USA
| | - S Lazarev
- Department of Chemistry, Yeshiva College, Yeshiva University, New York, NY, 10033, USA
| | - J E Fajardo
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - A Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - R Viswanathan
- Department of Chemistry, Yeshiva College, Yeshiva University, New York, NY, 10033, USA.
| |
Collapse
|
10
|
Li S, Wu S, Wang L, Li F, Jiang H, Bai F. Recent advances in predicting protein-protein interactions with the aid of artificial intelligence algorithms. Curr Opin Struct Biol 2022; 73:102344. [PMID: 35219216 DOI: 10.1016/j.sbi.2022.102344] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/02/2022] [Accepted: 01/17/2022] [Indexed: 12/15/2022]
Abstract
Protein-protein interactions (PPIs) are essential in the regulation of biological functions and cell events, therefore understanding PPIs have become a key issue to understanding the molecular mechanism and investigating the design of drugs. Here we highlight the major developments in computational methods developed for predicting PPIs by using types of artificial intelligence algorithms. The first part introduces the source of experimental PPI data. The second part is devoted to the PPI prediction methods based on sequential information. The third part covers representative methods using structural information as the input feature. The last part is methods designed by combining different types of features. For each part, the state-of-the-art computational PPI prediction methods are reviewed in an inclusive view. Finally, we discuss the flaws existing in this area and future directions of next-generation algorithms.
Collapse
Affiliation(s)
- Shiwei Li
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Sanan Wu
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lin Wang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Fenglei Li
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China; School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hualiang Jiang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Pudong, Shanghai, 201203, China
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, China; School of Information Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
11
|
Dapkūnas J, Olechnovič K, Venclovas Č. Modeling of protein complexes in CASP14 with emphasis on the interaction interface prediction. Proteins 2021; 89:1834-1843. [PMID: 34176161 PMCID: PMC9292421 DOI: 10.1002/prot.26167] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/08/2023]
Abstract
The goal of CASP experiments is to monitor the progress in the protein structure prediction field. During the 14th CASP edition we aimed to test our capabilities of predicting structures of protein complexes. Our protocol for modeling protein assemblies included both template‐based modeling and free docking. Structural templates were identified using sensitive sequence‐based searches. If sequence‐based searches failed, we performed structure‐based template searches using selected CASP server models. In the absence of reliable templates we applied free docking starting from monomers generated by CASP servers. We evaluated and ranked models of protein complexes using an improved version of our protein structure quality assessment method, VoroMQA, taking into account both interaction interface and global structure scores. If reliable templates could be identified, generally accurate models of protein assemblies were generated with the exception of an antibody‐antigen interaction. The success of free docking mainly depended on the accuracy of initial subunit models and on the scoring of docking solutions. To put our overall results in perspective, we analyzed our performance in the context of other CASP groups. Although the subunits in our assembly models often were not of the top quality, these models had, overall, the best‐predicted intersubunit interfaces according to several accuracy measures. We attribute our relative success primarily to the emphasis on the interaction interface when modeling and scoring.
Collapse
Affiliation(s)
- Justas Dapkūnas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Kliment Olechnovič
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Česlovas Venclovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
12
|
Residue-based pharmacophore approaches to study protein-protein interactions. Curr Opin Struct Biol 2021; 67:205-211. [PMID: 33486430 DOI: 10.1016/j.sbi.2020.12.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/04/2020] [Accepted: 12/28/2020] [Indexed: 01/22/2023]
Abstract
This review focuses on pharmacophore approaches in researching protein interfaces that bind protein ligands. Pharmacophore descriptions of binding interfaces that employ molecular dynamics simulation can account for effects of solvation and conformational flexibility. In addition, these calculations provide an approximation to entropic considerations and as such, a better approximation of the free energy of binding. Residue-based pharmacophore approaches can facilitate a variety of drug discovery tasks such as the identification of receptor-ligand partners, identifying their binding poses, designing protein interfaces for selectivity, or defining a reduced mutational combinatorial exploration for subsequent experimental engineering techniques by orders of magnitudes.
Collapse
|
13
|
Das S, Chakrabarti S. Classification and prediction of protein-protein interaction interface using machine learning algorithm. Sci Rep 2021; 11:1761. [PMID: 33469042 PMCID: PMC7815773 DOI: 10.1038/s41598-020-80900-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/15/2020] [Indexed: 01/29/2023] Open
Abstract
Structural insight of the protein-protein interaction (PPI) interface can provide knowledge about the kinetics, thermodynamics and molecular functions of the complex while elucidating its role in diseases and further enabling it as a potential therapeutic target. However, owing to experimental lag in solving protein-protein complex structures, three-dimensional (3D) knowledge of the PPI interfaces can be gained via computational approaches like molecular docking and post-docking analyses. Despite development of numerous docking tools and techniques, success in identification of native like interfaces based on docking score functions is limited. Hence, we employed an in-depth investigation of the structural features of the interface that might successfully delineate native complexes from non-native ones. We identify interface properties, which show statistically significant difference between native and non-native interfaces belonging to homo and hetero, protein-protein complexes. Utilizing these properties, a support vector machine (SVM) based classification scheme has been implemented to differentiate native and non-native like complexes generated using docking decoys. Benchmarking and comparative analyses suggest very good performance of our SVM classifiers. Further, protein interactions, which are proven via experimental findings but not resolved structurally, were subjected to this approach where 3D-models of the complexes were generated and most likely interfaces were predicted. A web server called Protein Complex Prediction by Interface Properties (PCPIP) is developed to predict whether interface of a given protein-protein dimer complex resembles known protein interfaces. The server is freely available at http://www.hpppi.iicb.res.in/pcpip/ .
Collapse
Affiliation(s)
- Subhrangshu Das
- grid.417635.20000 0001 2216 5074Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB India
| | - Saikat Chakrabarti
- grid.417635.20000 0001 2216 5074Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB India
| |
Collapse
|
14
|
Aderinwale T, Christoffer CW, Sarkar D, Alnabati E, Kihara D. Computational structure modeling for diverse categories of macromolecular interactions. Curr Opin Struct Biol 2020; 64:1-8. [PMID: 32599506 PMCID: PMC7665979 DOI: 10.1016/j.sbi.2020.05.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 01/23/2023]
Abstract
Computational protein-protein docking is one of the most intensively studied topics in structural bioinformatics. The field has made substantial progress through over three decades of development. The development began with methods for rigid-body docking of two proteins, which have now been extended in different directions to cover the various macromolecular interactions observed in a cell. Here, we overview the recent developments of the variations of docking methods, including multiple protein docking, peptide-protein docking, and disordered protein docking methods.
Collapse
Affiliation(s)
- Tunde Aderinwale
- Department of Computer Science, Purdue University, West Lafayette, IN, 47907, USA
| | | | - Daipayan Sarkar
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Eman Alnabati
- Department of Computer Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Daisuke Kihara
- Department of Computer Science, Purdue University, West Lafayette, IN, 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
15
|
Vakser IA. Challenges in protein docking. Curr Opin Struct Biol 2020; 64:160-165. [PMID: 32836051 DOI: 10.1016/j.sbi.2020.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/19/2020] [Accepted: 07/11/2020] [Indexed: 11/30/2022]
Abstract
Current developments in protein docking aim at improvement of applicability, accuracy and utility of modeling macromolecular complexes. The challenges include the need for greater emphasis on protein docking to molecules of different types, proper accounting for conformational flexibility upon binding, new promising methodologies based on residue co-evolution and deep learning, affinity prediction, and further development of fully automated docking servers. Importantly, new developments increasingly focus on realistic modeling of protein interactions in vivo, including crowded environment inside a cell, which involves multiple transient encounters, and propagating the system in time. This opinion paper offers the author's perspective on these challenges in structural modeling of protein interactions and the future of protein docking.
Collapse
Affiliation(s)
- Ilya A Vakser
- Computational Biology Program and Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
16
|
Prabhavathi H, Dasegowda KR, Renukananda KH, Lingaraju K, Naika HR. Exploration and evaluation of bioactive phytocompounds against BRCA proteins by in silico approach. J Biomol Struct Dyn 2020; 39:5471-5485. [PMID: 32643536 DOI: 10.1080/07391102.2020.1790424] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The proteins encoded by the two major breast cancer genes (BRCA1 and BRCA2), ensure the stability of DNA and prevent uncontrolled cell growth; mutation of these genes is linked to the development of hereditary breast cancers. Exploration of human breast cancer inhibitors plays a vital role in the drug discovery process. In the current work, in silico studies were performed which involves a computational approach for the identification of active phytocompounds from the diverse set of medicinal plant products against the BRCA receptor. The in silico study through pharmacokinetics and pharmacodynamics properties shown promising outcomes for these phytocompounds data set as breast cancer inhibitors. It was observed that the compounds conformed to the Lipinski's rule of five and had good bioavailability. The drug-likeness model score and ADMET profile of the designed ligands also established their potential as a drug candidate. The docking study provided useful insights on potential target-lead interactions and indicated that the newly designed leads had a good binding affinity for BRCA targets. A pharmacophore model was built to explore the scaffolds for BRCA inhibitory activity. An effort is made to screen an inhibitor against BRCA targets by combining the use of ADMET, docking score, and pharmacophore model.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- H Prabhavathi
- Department of Studies & Research in Biotechnology, Tumkur University, Tumakuru, India
| | - K R Dasegowda
- Department of Biotechnology & Genetics, Ramaiah College of Arts, Science and Commerce, Bangalore, India
| | | | - K Lingaraju
- Department of Studies & Research in Biotechnology, Tumkur University, Tumakuru, India
| | - H Raja Naika
- Department of Studies & Research in Biotechnology, Tumkur University, Tumakuru, India
| |
Collapse
|
17
|
Guven-Maiorov E, Hakouz A, Valjevac S, Keskin O, Tsai CJ, Gursoy A, Nussinov R. HMI-PRED: A Web Server for Structural Prediction of Host-Microbe Interactions Based on Interface Mimicry. J Mol Biol 2020; 432:3395-3403. [PMID: 32061934 PMCID: PMC7261632 DOI: 10.1016/j.jmb.2020.01.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/28/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Microbes, commensals, and pathogens, control the numerous functions in the host cells. They can alter host signaling and modulate immune surveillance by interacting with the host proteins. For shedding light on the contribution of microbes to health and disease, it is vital to discern how microbial proteins rewire host signaling and through which host proteins they do this. Host-Microbe Interaction PREDictor (HMI-PRED) is a user-friendly web server for structural prediction of protein-protein interactions (PPIs) between the host and a microbial species, including bacteria, viruses, fungi, and protozoa. HMI-PRED relies on "interface mimicry" through which the microbial proteins hijack host binding surfaces. Given the structure of a microbial protein of interest, HMI-PRED will return structural models of potential host-microbe interaction (HMI) complexes, the list of host endogenous and exogenous PPIs that can be disrupted, and tissue expression of the microbe-targeted host proteins. The server also allows users to upload homology models of microbial proteins. Broadly, it aims at large-scale, efficient identification of HMIs. The prediction results are stored in a repository for community access. HMI-PRED is free and available at https://interactome.ku.edu.tr/hmi.
Collapse
Affiliation(s)
- Emine Guven-Maiorov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
| | - Asma Hakouz
- Department of Computer Engineering, Koc University, Istanbul, 34450, Turkey.
| | - Sukejna Valjevac
- Department of Computer Engineering, Koc University, Istanbul, 34450, Turkey.
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, Koc University, Istanbul, 34450, Turkey.
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
| | - Attila Gursoy
- Department of Computer Engineering, Koc University, Istanbul, 34450, Turkey.
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA; Sackler Inst. of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|