1
|
Nayar G, Altman RB. Heterogeneous network approaches to protein pathway prediction. Comput Struct Biotechnol J 2024; 23:2727-2739. [PMID: 39035835 PMCID: PMC11260399 DOI: 10.1016/j.csbj.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Understanding protein-protein interactions (PPIs) and the pathways they comprise is essential for comprehending cellular functions and their links to specific phenotypes. Despite the prevalence of molecular data generated by high-throughput sequencing technologies, a significant gap remains in translating this data into functional information regarding the series of interactions that underlie phenotypic differences. In this review, we present an in-depth analysis of heterogeneous network methodologies for modeling protein pathways, highlighting the critical role of integrating multifaceted biological data. It outlines the process of constructing these networks, from data representation to machine learning-driven predictions and evaluations. The work underscores the potential of heterogeneous networks in capturing the complexity of proteomic interactions, thereby offering enhanced accuracy in pathway prediction. This approach not only deepens our understanding of cellular processes but also opens up new possibilities in disease treatment and drug discovery by leveraging the predictive power of comprehensive proteomic data analysis.
Collapse
Affiliation(s)
- Gowri Nayar
- Department of Biomedical Data Science, Stanford University, United States
| | - Russ B. Altman
- Department of Biomedical Data Science, Stanford University, United States
- Department of Genetics, Stanford University, United States
- Department of Medicine, Stanford University, United States
- Department of Bioengineering, Stanford University, United States
| |
Collapse
|
2
|
Bereczki Z, Benczik B, Balogh OM, Marton S, Puhl E, Pétervári M, Váczy-Földi M, Papp ZT, Makkos A, Glass K, Locquet F, Euler G, Schulz R, Ferdinandy P, Ágg B. Mitigating off-target effects of small RNAs: conventional approaches, network theory and artificial intelligence. Br J Pharmacol 2024. [PMID: 39293936 DOI: 10.1111/bph.17302] [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: 11/30/2023] [Revised: 05/07/2024] [Accepted: 06/17/2024] [Indexed: 09/20/2024] Open
Abstract
Three types of highly promising small RNA therapeutics, namely, small interfering RNAs (siRNAs), microRNAs (miRNAs) and the RNA subtype of antisense oligonucleotides (ASOs), offer advantages over small-molecule drugs. These small RNAs can target any gene product, opening up new avenues of effective and safe therapeutic approaches for a wide range of diseases. In preclinical research, synthetic small RNAs play an essential role in the investigation of physiological and pathological pathways as silencers of specific genes, facilitating discovery and validation of drug targets in different conditions. Off-target effects of small RNAs, however, could make it difficult to interpret experimental results in the preclinical phase and may contribute to adverse events of small RNA therapeutics. Out of the two major types of off-target effects we focused on the hybridization-dependent, especially on the miRNA-like off-target effects. Our main aim was to discuss several approaches, including sequence design, chemical modifications and target prediction, to reduce hybridization-dependent off-target effects that should be considered even at the early development phase of small RNA therapy. Because there is no standard way of predicting hybridization-dependent off-target effects, this review provides an overview of all major state-of-the-art computational methods and proposes new approaches, such as the possible inclusion of network theory and artificial intelligence (AI) in the prediction workflows. Case studies and a concise survey of experimental methods for validating in silico predictions are also presented. These methods could contribute to interpret experimental results, to minimize off-target effects and hopefully to avoid off-target-related adverse events of small RNA therapeutics.
Collapse
Affiliation(s)
- Zoltán Bereczki
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bettina Benczik
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Olivér M Balogh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Szandra Marton
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Eszter Puhl
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Mátyás Pétervári
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Sanovigado Kft, Budapest, Hungary
| | - Máté Váczy-Földi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zsolt Tamás Papp
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - András Makkos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Kimberly Glass
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Fabian Locquet
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Gerhild Euler
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
3
|
Zhang C, Gao Q, Li M, Yu T. Implementing link prediction in protein networks via feature fusion models based on graph neural networks. Comput Biol Chem 2024; 108:107980. [PMID: 38000328 DOI: 10.1016/j.compbiolchem.2023.107980] [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: 04/27/2023] [Revised: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023]
Abstract
MOTIVATION Protein-protein interactions serve as the cornerstone for various biochemical processes within biological organisms. Existing research methodologies predominantly employ link prediction techniques to analyze these interaction networks. However, traditional approaches often fall short in delivering satisfactory predictive performance when applied to multi-species datasets. Current computational methods largely focus on analyzing the network topology, resulting in a somewhat monolithic feature set. The integration of diverse features in the model could potentially yield superior performance and broader applicability. To this end, we propose an autoencoder model built on graph neural networks, designed to enhance both predictive performance and generalizability by leveraging the integration of gene ontology. RESULTS In this research, we developed AGraphSAGE, a model specifically designed for analyzing protein-protein interaction network data. By seamlessly integrating gene ontology into the graph structure, we employed a dual-channel graph sampling and aggregation network that capitalizes on topological information to process high-dimensional features. Feature fusion is achieved through the implementation of graph attention mechanisms, and we adopted a link prediction framework as the experimental training model. Performance was evaluated on real-world datasets using key metrics, such as Area Under the Curve (AUC). A hyperparameter search space was established, and a Bayesian optimization strategy was applied to iteratively fine-tune the model, assessing the impact of various parameters on predictive efficacy. The experimental results validate that our proposed model is capable of effectively predicting protein-protein interactions across diverse biological species.
Collapse
Affiliation(s)
- Chi Zhang
- College of Computer and Control Engineering, Qiqihar University, Qiqihar 161006, China
| | - Qian Gao
- College of Computer and Control Engineering, Qiqihar University, Qiqihar 161006, China
| | - Ming Li
- College of Computer and Control Engineering, Qiqihar University, Qiqihar 161006, China.
| | - Tianfei Yu
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar 161006, China.
| |
Collapse
|
4
|
Michalik I, Kuder KJ. Machine Learning Methods in Protein-Protein Docking. Methods Mol Biol 2024; 2780:107-126. [PMID: 38987466 DOI: 10.1007/978-1-0716-3985-6_7] [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] [Indexed: 07/12/2024]
Abstract
An exponential increase in the number of publications that address artificial intelligence (AI) usage in life sciences has been noticed in recent years, while new modeling techniques are constantly being reported. The potential of these methods is vast-from understanding fundamental cellular processes to discovering new drugs and breakthrough therapies. Computational studies of protein-protein interactions, crucial for understanding the operation of biological systems, are no exception in this field. However, despite the rapid development of technology and the progress in developing new approaches, many aspects remain challenging to solve, such as predicting conformational changes in proteins, or more "trivial" issues as high-quality data in huge quantities.Therefore, this chapter focuses on a short introduction to various AI approaches to study protein-protein interactions, followed by a description of the most up-to-date algorithms and programs used for this purpose. Yet, given the considerable pace of development in this hot area of computational science, at the time you read this chapter, the development of the algorithms described, or the emergence of new (and better) ones should come as no surprise.
Collapse
Affiliation(s)
- Ilona Michalik
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| | - Kamil J Kuder
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland.
| |
Collapse
|
5
|
Peng S, Yamamoto A, Ito K. Link prediction on bipartite networks using matrix factorization with negative sample selection. PLoS One 2023; 18:e0289568. [PMID: 37585433 PMCID: PMC10431684 DOI: 10.1371/journal.pone.0289568] [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: 04/03/2023] [Accepted: 07/19/2023] [Indexed: 08/18/2023] Open
Abstract
We propose a new method for bipartite link prediction using matrix factorization with negative sample selection. Bipartite link prediction is a problem that aims to predict the missing links or relations in a bipartite network. One of the most popular solutions to the problem is via matrix factorization (MF), which performs well but requires reliable information on both absent and present network links as training samples. This, however, is sometimes unavailable since there is no ground truth for absent links. To solve the problem, we propose a technique called negative sample selection, which selects reliable negative training samples using formal concept analysis (FCA) of a given bipartite network in advance of the preceding MF process. We conduct experiments on two hypothetical application scenarios to prove that our joint method outperforms the raw MF-based link prediction method as well as all other previously-proposed unsupervised link prediction methods.
Collapse
Affiliation(s)
- Siqi Peng
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Akihiro Yamamoto
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Kimihito Ito
- International Institute for Zoonosis Control, Division of Bioinformatics, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
6
|
Dou B, Zhu Z, Merkurjev E, Ke L, Chen L, Jiang J, Zhu Y, Liu J, Zhang B, Wei GW. Machine Learning Methods for Small Data Challenges in Molecular Science. Chem Rev 2023; 123:8736-8780. [PMID: 37384816 PMCID: PMC10999174 DOI: 10.1021/acs.chemrev.3c00189] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Small data are often used in scientific and engineering research due to the presence of various constraints, such as time, cost, ethics, privacy, security, and technical limitations in data acquisition. However, big data have been the focus for the past decade, small data and their challenges have received little attention, even though they are technically more severe in machine learning (ML) and deep learning (DL) studies. Overall, the small data challenge is often compounded by issues, such as data diversity, imputation, noise, imbalance, and high-dimensionality. Fortunately, the current big data era is characterized by technological breakthroughs in ML, DL, and artificial intelligence (AI), which enable data-driven scientific discovery, and many advanced ML and DL technologies developed for big data have inadvertently provided solutions for small data problems. As a result, significant progress has been made in ML and DL for small data challenges in the past decade. In this review, we summarize and analyze several emerging potential solutions to small data challenges in molecular science, including chemical and biological sciences. We review both basic machine learning algorithms, such as linear regression, logistic regression (LR), k-nearest neighbor (KNN), support vector machine (SVM), kernel learning (KL), random forest (RF), and gradient boosting trees (GBT), and more advanced techniques, including artificial neural network (ANN), convolutional neural network (CNN), U-Net, graph neural network (GNN), Generative Adversarial Network (GAN), long short-term memory (LSTM), autoencoder, transformer, transfer learning, active learning, graph-based semi-supervised learning, combining deep learning with traditional machine learning, and physical model-based data augmentation. We also briefly discuss the latest advances in these methods. Finally, we conclude the survey with a discussion of promising trends in small data challenges in molecular science.
Collapse
Affiliation(s)
- Bozheng Dou
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
| | - Zailiang Zhu
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
| | - Ekaterina Merkurjev
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Lu Ke
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
| | - Long Chen
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
| | - Jian Jiang
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yueying Zhu
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
| | - Jie Liu
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
| | - Bengong Zhang
- Research Center of Nonlinear Science, School of Mathematical and Physical Sciences,Wuhan Textile University, Wuhan 430200, P, R. China
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
7
|
Wang XW, Madeddu L, Spirohn K, Martini L, Fazzone A, Becchetti L, Wytock TP, Kovács IA, Balogh OM, Benczik B, Pétervári M, Ágg B, Ferdinandy P, Vulliard L, Menche J, Colonnese S, Petti M, Scarano G, Cuomo F, Hao T, Laval F, Willems L, Twizere JC, Vidal M, Calderwood MA, Petrillo E, Barabási AL, Silverman EK, Loscalzo J, Velardi P, Liu YY. Assessment of community efforts to advance network-based prediction of protein-protein interactions. Nat Commun 2023; 14:1582. [PMID: 36949045 PMCID: PMC10033937 DOI: 10.1038/s41467-023-37079-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/02/2023] [Indexed: 03/24/2023] Open
Abstract
Comprehensive understanding of the human protein-protein interaction (PPI) network, aka the human interactome, can provide important insights into the molecular mechanisms of complex biological processes and diseases. Despite the remarkable experimental efforts undertaken to date to determine the structure of the human interactome, many PPIs remain unmapped. Computational approaches, especially network-based methods, can facilitate the identification of previously uncharacterized PPIs. Many such methods have been proposed. Yet, a systematic evaluation of existing network-based methods in predicting PPIs is still lacking. Here, we report community efforts initiated by the International Network Medicine Consortium to benchmark the ability of 26 representative network-based methods to predict PPIs across six different interactomes of four different organisms: A. thaliana, C. elegans, S. cerevisiae, and H. sapiens. Through extensive computational and experimental validations, we found that advanced similarity-based methods, which leverage the underlying network characteristics of PPIs, show superior performance over other general link prediction methods in the interactomes we considered.
Collapse
Affiliation(s)
- Xu-Wen Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Lorenzo Madeddu
- Translational and Precision Medicine Department Sapienza University of Rome, Rome, Italy
| | - Kerstin Spirohn
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Leonardo Martini
- Department of Computer, Control, and Management Engineering "Antonio Rubert", Sapienza University of Rome, Rome, Italy
| | | | - Luca Becchetti
- Department of Computer, Control, and Management Engineering "Antonio Rubert", Sapienza University of Rome, Rome, Italy
| | - Thomas P Wytock
- Department of Physics and Astronomy, Northwestern University, Evanston, IL, 60208, USA
| | - István A Kovács
- Department of Physics and Astronomy, Northwestern University, Evanston, IL, 60208, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL, 60208, USA
| | - Olivér M Balogh
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bettina Benczik
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, 6722, Szeged, Hungary
| | - Mátyás Pétervári
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, 6722, Szeged, Hungary
| | - Péter Ferdinandy
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, 6722, Szeged, Hungary
| | - Loan Vulliard
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna, Austria
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, Vienna, Austria
- Faculty of Mathematics, University of Vienna, Vienna, Austria
| | - Stefania Colonnese
- Department of Information Engineering, Electronics, and Telecommunications (DIET), University of Rome "Sapienza", Rome, Italy
| | - Manuela Petti
- Department of Computer, Control, and Management Engineering "Antonio Rubert", Sapienza University of Rome, Rome, Italy
| | - Gaetano Scarano
- Department of Information Engineering, Electronics, and Telecommunications (DIET), University of Rome "Sapienza", Rome, Italy
| | - Francesca Cuomo
- Department of Information Engineering, Electronics, and Telecommunications (DIET), University of Rome "Sapienza", Rome, Italy
| | - Tong Hao
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Florent Laval
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Laboratory of Molecular and Cellular Epigenetic, GIGA Institute, University of Liège, Liège, Belgium
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
- TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - Luc Willems
- Laboratory of Molecular and Cellular Epigenetic, GIGA Institute, University of Liège, Liège, Belgium
- TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - Jean-Claude Twizere
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
- TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael A Calderwood
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Enrico Petrillo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Albert-László Barabási
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Network Science Institute and Department of Physics, Northeastern University, Boston, MA, 02115, USA
- Department of Network and Data Science, Central European University, Budapest, H-1051, Hungary
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Paola Velardi
- Translational and Precision Medicine Department Sapienza University of Rome, Rome, Italy.
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
- Center for Artificial Intelligence and Modeling, The Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, 61801, USA.
| |
Collapse
|
8
|
Temiz M, Bakir-Gungor B, Güner Şahan P, Coskun M. Topological feature generation for link prediction in biological networks. PeerJ 2023; 11:e15313. [PMID: 37187525 PMCID: PMC10178302 DOI: 10.7717/peerj.15313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Graph or network embedding is a powerful method for extracting missing or potential information from interactions between nodes in biological networks. Graph embedding methods learn representations of nodes and interactions in a graph with low-dimensional vectors, which facilitates research to predict potential interactions in networks. However, most graph embedding methods suffer from high computational costs in the form of high computational complexity of the embedding methods and learning times of the classifier, as well as the high dimensionality of complex biological networks. To address these challenges, in this study, we use the Chopper algorithm as an alternative approach to graph embedding, which accelerates the iterative processes and thus reduces the running time of the iterative algorithms for three different (nervous system, blood, heart) undirected protein-protein interaction (PPI) networks. Due to the high dimensionality of the matrix obtained after the embedding process, the data are transformed into a smaller representation by applying feature regularization techniques. We evaluated the performance of the proposed method by comparing it with state-of-the-art methods. Extensive experiments demonstrate that the proposed approach reduces the learning time of the classifier and performs better in link prediction. We have also shown that the proposed embedding method is faster than state-of-the-art methods on three different PPI datasets.
Collapse
Affiliation(s)
- Mustafa Temiz
- Department of Computer Engineering, Abdullah Gul University, Kayseri, Turkey
| | - Burcu Bakir-Gungor
- Department of Computer Engineering, Abdullah Gul University, Kayseri, Turkey
| | - Pınar Güner Şahan
- Department of Computer Engineering, Abdullah Gul University, Kayseri, Turkey
| | - Mustafa Coskun
- Department of Artificial Intelligence and Big Data Engineering, Ankara University, Ankara, Turkey
| |
Collapse
|
9
|
Yang L, Zhang YH, Huang F, Li Z, Huang T, Cai YD. Identification of protein–protein interaction associated functions based on gene ontology and KEGG pathway. Front Genet 2022; 13:1011659. [PMID: 36171880 PMCID: PMC9511048 DOI: 10.3389/fgene.2022.1011659] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Protein–protein interactions (PPIs) are extremely important for gaining mechanistic insights into the functional organization of the proteome. The resolution of PPI functions can help in the identification of novel diagnostic and therapeutic targets with medical utility, thus facilitating the development of new medications. However, the traditional methods for resolving PPI functions are mainly experimental methods, such as co-immunoprecipitation, pull-down assays, cross-linking, label transfer, and far-Western blot analysis, that are not only expensive but also time-consuming. In this study, we constructed an integrated feature selection scheme for the large-scale selection of the relevant functions of PPIs by using the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotations of PPI participants. First, we encoded the proteins in each PPI with their gene ontologies and KEGG pathways. Then, the encoded protein features were refined as features of both positive and negative PPIs. Subsequently, Boruta was used for the initial filtering of features to obtain 5684 features. Three feature ranking algorithms, namely, least absolute shrinkage and selection operator, light gradient boosting machine, and max-relevance and min-redundancy, were applied to evaluate feature importance. Finally, the top-ranked features derived from multiple datasets were comprehensively evaluated, and the intersection of results mined by three feature ranking algorithms was taken to identify the features with high correlation with PPIs. Some functional terms were identified in our study, including cytokine–cytokine receptor interaction (hsa04060), intrinsic component of membrane (GO:0031224), and protein-binding biological process (GO:0005515). Our newly proposed integrated computational approach offers a novel perspective of the large-scale mining of biological functions linked to PPI.
Collapse
Affiliation(s)
- Lili Yang
- Measurement Biotechnique Research Center, School of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Yu-Hang Zhang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - FeiMing Huang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - ZhanDong Li
- Measurement Biotechnique Research Center, School of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Tao Huang, ; Yu-Dong Cai,
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
- *Correspondence: Tao Huang, ; Yu-Dong Cai,
| |
Collapse
|
10
|
Robin V, Bodein A, Scott-Boyer MP, Leclercq M, Périn O, Droit A. Overview of methods for characterization and visualization of a protein-protein interaction network in a multi-omics integration context. Front Mol Biosci 2022; 9:962799. [PMID: 36158572 PMCID: PMC9494275 DOI: 10.3389/fmolb.2022.962799] [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: 06/06/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
At the heart of the cellular machinery through the regulation of cellular functions, protein-protein interactions (PPIs) have a significant role. PPIs can be analyzed with network approaches. Construction of a PPI network requires prediction of the interactions. All PPIs form a network. Different biases such as lack of data, recurrence of information, and false interactions make the network unstable. Integrated strategies allow solving these different challenges. These approaches have shown encouraging results for the understanding of molecular mechanisms, drug action mechanisms, and identification of target genes. In order to give more importance to an interaction, it is evaluated by different confidence scores. These scores allow the filtration of the network and thus facilitate the representation of the network, essential steps to the identification and understanding of molecular mechanisms. In this review, we will discuss the main computational methods for predicting PPI, including ones confirming an interaction as well as the integration of PPIs into a network, and we will discuss visualization of these complex data.
Collapse
Affiliation(s)
- Vivian Robin
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Antoine Bodein
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Marie-Pier Scott-Boyer
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Mickaël Leclercq
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| | - Olivier Périn
- Digital Sciences Department, L'Oréal Advanced Research, Aulnay-sous-bois, France
| | - Arnaud Droit
- Molecular Medicine Department, CHU de Québec Research Center, Université Laval, Québec, QC, Canada
| |
Collapse
|
11
|
Avery C, Patterson J, Grear T, Frater T, Jacobs DJ. Protein Function Analysis through Machine Learning. Biomolecules 2022; 12:1246. [PMID: 36139085 PMCID: PMC9496392 DOI: 10.3390/biom12091246] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/22/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Machine learning (ML) has been an important arsenal in computational biology used to elucidate protein function for decades. With the recent burgeoning of novel ML methods and applications, new ML approaches have been incorporated into many areas of computational biology dealing with protein function. We examine how ML has been integrated into a wide range of computational models to improve prediction accuracy and gain a better understanding of protein function. The applications discussed are protein structure prediction, protein engineering using sequence modifications to achieve stability and druggability characteristics, molecular docking in terms of protein-ligand binding, including allosteric effects, protein-protein interactions and protein-centric drug discovery. To quantify the mechanisms underlying protein function, a holistic approach that takes structure, flexibility, stability, and dynamics into account is required, as these aspects become inseparable through their interdependence. Another key component of protein function is conformational dynamics, which often manifest as protein kinetics. Computational methods that use ML to generate representative conformational ensembles and quantify differences in conformational ensembles important for function are included in this review. Future opportunities are highlighted for each of these topics.
Collapse
Affiliation(s)
- Chris Avery
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - John Patterson
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Tyler Grear
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
- Department of Physics and Optical Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Theodore Frater
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Donald J. Jacobs
- Department of Physics and Optical Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
12
|
Zhang Y, Lu J, Ma Y, Sun L, Wang S, Yue X, Yu J, Xue P. Establishment of fingerprint and mechanism of anti-myocardial ischemic effect of Syringa pinnatifolia. Biomed Chromatogr 2022; 36:e5475. [PMID: 35947036 DOI: 10.1002/bmc.5475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To establish the fingerprint of Syringa pinnatifolia Hemsl. (SP), analyze the blood components of SP, and explore the possible mechanism of SP's anti-myocardial ischemia, so as to provide scientific basis for the follow-up development and research of SP and lay a foundation for its clinical application. METHODS The fingerprint of SP was established by UPLC-QE-MS and GC-MS. A rat Myocardial infarction (MI) was constructed by ligating the left anterior descending branch (LAD) of the rat coronary artery, and SP alcohol extract was administered to evaluate its anti-myocardial ischemic effect. We analyzed the blood components of SP, screened the active compounds, established a database of SP anti-myocardial ischemic targets, and explored the possible mechanism of SP in treating MI by bioinformatics. The rats were examined by echocardiography, serum biomarkers were determined, and pathological changes were observed by histopathological examination. TUNEL staining was performed to detect the apoptotic level of cells, and western blot and qRT-PCR were performed to detect the expression levels of Bcl-2, Bax and caspase-3 in heart tissues. RESULTS In the fingerprint of SP, 24 common peaks were established, and the similarity evaluation results of 10 batches of SP were all > 0.9. UPLC-QE-MS and GC-MS detected a total of 17 active ingredients in the drug-containing serum, including terpenoids, flavonoids, phenols, phenylpropanoids and phenolic acids, the most abundant of which was resveratrol. Enrichment analysis of SP targets against myocardial ischemia revealed that key candidate targets of SP were significantly enriched in multiple pathways associated with apoptosis. Resveratrol was administered to the successfully modeled rats, and the results showed that the resveratrol group significantly reduced LVEDd and LVEDs and significantly increased EF and FS in all groups compared with the model group. Resveratrol significantly reduced the levels of CK-MB and LDH in serum compared to the model group (p < 0.001). Hematoxylin-eosin (HE) staining of rat myocardial tissue showed that all lesions were reduced under microscopic observation in the resveratrol group compared with the model group. RT-PCR and western blot results showed that resveratrol group down-regulated the expression of the pro-apoptotic factor Bax, up-regulated the expression of the anti-apoptotic factor Bcl-2, and decreased the expression of Caspase-3. CONCLUSION The established fingerprints are accurate, reliable and reproducible, and can be used as an effective method for the quality control of the herbs. The anti-myocardial ischemia effect of SP may be that resveratrol can improve cardiac function and inhibit cardiomyocyte apoptosis to protect cardiomyocytes. The present study provides ample evidence for the clinical use of SP, suggesting that this drug has great potential in the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Ye Zhang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Jingkun Lu
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Yuheng Ma
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Lijun Sun
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Suwei Wang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Xin Yue
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Jiuwang Yu
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Peifeng Xue
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| |
Collapse
|