1
|
Zhang L, Huang L, Zhou Y, Meng J, Zhang L, Zhou Y, Zheng N, Guo T, Zhao S, Wang Z, Huo Y, Zhao Y, Chen XF, Zheng H, Holtzman DM, Zhang YW. Microglial CD2AP deficiency exerts protection in an Alzheimer's disease model of amyloidosis. Mol Neurodegener 2024; 19:95. [PMID: 39695808 DOI: 10.1186/s13024-024-00789-7] [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: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The CD2-associated protein (CD2AP) was initially identified in peripheral immune cells and regulates cytoskeleton and protein trafficking. Single nucleotide polymorphisms (SNPs) in the CD2AP gene have been associated with Alzheimer's disease (AD). However, the functional role of CD2AP, especially its role in microglia during AD onset, remains elusive. METHODS CD2AP protein levels in cultured primary cells and in 5xFAD mice was studied. Microglial CD2AP-deficient mice were crossed with 5xFAD mice and the offspring were subjected to neuropathological assessment, behavioral tests, electrophysiology, RNA-seq, Golgi staining, and biochemistry analysis. Primary microglia were also isolated for assessing their uptake and morphology changes. RESULTS We find that CD2AP is abundantly expressed in microglia and its levels are elevated in the brain of AD patients and the 5xFAD model mice at pathological stages. We demonstrate that CD2AP haploinsufficiency in microglia significantly attenuates cognitive and synaptic deficits, weakens the response of microglia to Aβ and the formation of disease-associated microglia (DAM), and alleviates synapse loss in 5xFAD mice. We show that CD2AP-deficient microglia exhibit compromised uptake ability. In addition, we find that CD2AP expression is positively correlated with the expression of the complement C1q that is important for synapse phagocytosis and the formation of DAM in response to Aβ deposition. Moreover, we reveal that CD2AP interacts with colony stimulating factor 1 receptor (CSF1R) and regulates CSF1R cell surface levels, which may further affect C1q expression. CONCLUSIONS Our results demonstrate that CD2AP regulates microgliosis and identify a protective function of microglial CD2AP deficiency against Aβ deposition, suggesting the importance of detailed investigation of AD-associated genes in different brain cells for thoroughly understanding their exact contribution to AD.
Collapse
Affiliation(s)
- Lingliang Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lingling Huang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yuhang Zhou
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jian Meng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Liang Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunqiang Zhou
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Naizhen Zheng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Tiantian Guo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shanshan Zhao
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zijie Wang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yuanhui Huo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yingjun Zhao
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiao-Fen Chen
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Honghua Zheng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yun-Wu Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
2
|
Tahir ul Qamar M, Noor F, Guo YX, Zhu XT, Chen LL. Deep-HPI-pred: An R-Shiny applet for network-based classification and prediction of Host-Pathogen protein-protein interactions. Comput Struct Biotechnol J 2024; 23:316-329. [PMID: 38192372 PMCID: PMC10772389 DOI: 10.1016/j.csbj.2023.12.010] [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: 10/22/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/10/2024] Open
Abstract
Host-pathogen interactions (HPIs) are vital in numerous biological activities and are intrinsically linked to the onset and progression of infectious diseases. HPIs are pivotal in the entire lifecycle of diseases: from the onset of pathogen introduction, navigating through the mechanisms that bypass host cellular defenses, to its subsequent proliferation inside the host. At the heart of these stages lies the synergy of proteins from both the host and the pathogen. By understanding these interlinking protein dynamics, we can gain crucial insights into how diseases progress and pave the way for stronger plant defenses and the swift formulation of countermeasures. In the framework of current study, we developed a web-based R/Shiny app, Deep-HPI-pred, that uses network-driven feature learning method to predict the yet unmapped interactions between pathogen and host proteins. Leveraging citrus and CLas bacteria training datasets as case study, we spotlight the effectiveness of Deep-HPI-pred in discerning Protein-protein interaction (PPIs) between them. Deep-HPI-pred use Multilayer Perceptron (MLP) models for HPI prediction, which is based on a comprehensive evaluation of topological features and neural network architectures. When subjected to independent validation datasets, the predicted models consistently surpassed a Matthews correlation coefficient (MCC) of 0.80 in host-pathogen interactions. Remarkably, the use of Eigenvector Centrality as the leading topological feature further enhanced this performance. Further, Deep-HPI-pred also offers relevant gene ontology (GO) term information for each pathogen and host protein within the system. This protein annotation data contributes an additional layer to our understanding of the intricate dynamics within host-pathogen interactions. In the additional benchmarking studies, the Deep-HPI-pred model has proven its robustness by consistently delivering reliable results across different host-pathogen systems, including plant-pathogens (accuracy of 98.4% and 97.9%), human-virus (accuracy of 94.3%), and animal-bacteria (accuracy of 96.6%) interactomes. These results not only demonstrate the model's versatility but also pave the way for gaining comprehensive insights into the molecular underpinnings of complex host-pathogen interactions. Taken together, the Deep-HPI-pred applet offers a unified web service for both identifying and illustrating interaction networks. Deep-HPI-pred applet is freely accessible at its homepage: https://cbi.gxu.edu.cn/shiny-apps/Deep-HPI-pred/ and at github: https://github.com/tahirulqamar/Deep-HPI-pred.
Collapse
Affiliation(s)
- Muhammad Tahir ul Qamar
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China
| | - Fatima Noor
- Integrative Omics and Molecular Modeling Laboratory, Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad 38000, Pakistan
| | - Yi-Xiong Guo
- National Key Laboratory of Crop Genetic Improvement, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Xi-Tong Zhu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China
| | - Ling-Ling Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Life Science and Technology, Guangxi University, Nanning 530004, China
| |
Collapse
|
3
|
Utgés JS, Barton GJ. Comparative evaluation of methods for the prediction of protein-ligand binding sites. J Cheminform 2024; 16:126. [PMID: 39529176 PMCID: PMC11552181 DOI: 10.1186/s13321-024-00923-z] [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: 08/02/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The accurate identification of protein-ligand binding sites is of critical importance in understanding and modulating protein function. Accordingly, ligand binding site prediction has remained a research focus for over three decades with over 50 methods developed and a change of paradigm from geometry-based to machine learning. In this work, we collate 13 ligand binding site predictors, spanning 30 years, focusing on the latest machine learning-based methods such as VN-EGNN, IF-SitePred, GrASP, PUResNet, and DeepPocket and compare them to the established P2Rank, PRANK and fpocket and earlier methods like PocketFinder, Ligsite and Surfnet. We benchmark the methods against the human subset of our new curated reference dataset, LIGYSIS. LIGYSIS is a comprehensive protein-ligand complex dataset comprising 30,000 proteins with bound ligands which aggregates biologically relevant unique protein-ligand interfaces across biological units of multiple structures from the same protein. LIGYSIS is an improvement for testing methods over earlier datasets like sc-PDB, PDBbind, binding MOAD, COACH420 and HOLO4K which either include 1:1 protein-ligand complexes or consider asymmetric units. Re-scoring of fpocket predictions by PRANK and DeepPocket display the highest recall (60%) whilst IF-SitePred presents the lowest recall (39%). We demonstrate the detrimental effect that redundant prediction of binding sites has on performance as well as the beneficial impact of stronger pocket scoring schemes, with improvements up to 14% in recall (IF-SitePred) and 30% in precision (Surfnet). Finally, we propose top-N+2 recall as the universal benchmark metric for ligand binding site prediction and urge authors to share not only the source code of their methods, but also of their benchmark.Scientific contributionsThis study conducts the largest benchmark of ligand binding site prediction methods to date, comparing 13 original methods and 15 variants using 10 informative metrics. The LIGYSIS dataset is introduced, which aggregates biologically relevant protein-ligand interfaces across multiple structures of the same protein. The study highlights the detrimental effect of redundant binding site prediction and demonstrates significant improvement in recall and precision through stronger scoring schemes. Finally, top-N+2 recall is proposed as a universal benchmark metric for ligand binding site prediction, with a recommendation for open-source sharing of both methods and benchmarks.
Collapse
Affiliation(s)
- Javier S Utgés
- Division of Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Geoffrey J Barton
- Division of Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK.
| |
Collapse
|
4
|
Kim JH, Koo B, Kim S. PONYTA: prioritization of phenotype-related genes from mouse KO events using PU learning on a biological network. Bioinformatics 2024; 40:btae634. [PMID: 39432684 PMCID: PMC11561041 DOI: 10.1093/bioinformatics/btae634] [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: 07/12/2024] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
MOTIVATION Transcriptome data from gene knock-out (KO) experiments in mice provide crucial insights into the intricate interactions between genotype and phenotype. Differentially expressed gene (DEG) analysis and network propagation (NP) are well-established methods for analysing transcriptome data. To determine genes related to phenotype changes from a KO experiment, we need to choose a cutoff value for the corresponding criterion based on the specific method. Using a rigorous cutoff value for DEG analysis and NP is likely to select mostly positive genes related to the phenotype, but many will be rejected as false negatives. On the other hand, using a loose cutoff value for either method is prone to include a number of genes that are not phenotype-related, which are false positives. Thus, the research problem at hand is how to deal with the trade-off between false negatives and false positives. RESULTS We propose a novel framework called PONYTA for gene prioritization via positive-unlabeled (PU) learning on biological networks. Beginning with the selection of true phenotype-related genes using a rigorous cutoff value for DEG analysis and NP, we address the issue of handling false negatives by rescuing them through PU learning. Evaluations on transcriptome data from multiple studies show that our approach has superior gene prioritization ability compared to benchmark models. Therefore, PONYTA effectively prioritizes genes related to phenotypes derived from gene KO events and guides in vitro and in vivo gene KO experiments for increased efficiency. AVAILABILITY AND IMPLEMENTATION The source code of PONYTA is available at https://github.com/Jun-Hyeong-Kim/PONYTA.
Collapse
Affiliation(s)
- Jun Hyeong Kim
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul 08826, Republic of Korea
| | - Bonil Koo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul 08758, Republic of Korea
| | - Sun Kim
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul 08758, Republic of Korea
- Department of Computer Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
5
|
Volzhenin K, Bittner L, Carbone A. SENSE-PPI reconstructs interactomes within, across, and between species at the genome scale. iScience 2024; 27:110371. [PMID: 39055916 PMCID: PMC11269938 DOI: 10.1016/j.isci.2024.110371] [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: 12/12/2023] [Revised: 05/04/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Ab initio computational reconstructions of protein-protein interaction (PPI) networks will provide invaluable insights into cellular systems, enabling the discovery of novel molecular interactions and elucidating biological mechanisms within and between organisms. Leveraging the latest generation protein language models and recurrent neural networks, we present SENSE-PPI, a sequence-based deep learning model that efficiently reconstructs ab initio PPIs, distinguishing partners among tens of thousands of proteins and identifying specific interactions within functionally similar proteins. SENSE-PPI demonstrates high accuracy, limited training requirements, and versatility in cross-species predictions, even with non-model organisms and human-virus interactions. Its performance decreases for phylogenetically more distant model and non-model organisms, but signal alteration is very slow. In this regard, it demonstrates the important role of parameters in protein language models. SENSE-PPI is very fast and can test 10,000 proteins against themselves in a matter of hours, enabling the reconstruction of genome-wide proteomes.
Collapse
Affiliation(s)
- Konstantin Volzhenin
- Sorbonne Université, CNRS, IBPS, UMR 7238, Laboratoire de Biologie Computationnelle et Quantitative (LCQB), 75005 Paris, France
| | - Lucie Bittner
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Muséum national d’Histoire naturelle, CNRS, Sorbonne Université, EPHE, Université des Antilles, Paris, France
- Institut Universitaire de France, Paris, France
| | - Alessandra Carbone
- Sorbonne Université, CNRS, IBPS, UMR 7238, Laboratoire de Biologie Computationnelle et Quantitative (LCQB), 75005 Paris, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
6
|
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
|
7
|
Zhang HB, Ding XB, Jin J, Guo WP, Yang QL, Chen PC, Yao H, Ruan L, Tao YT, Chen X. Predicted mouse interactome and network-based interpretation of differentially expressed genes. PLoS One 2022; 17:e0264174. [PMID: 35390003 PMCID: PMC8989236 DOI: 10.1371/journal.pone.0264174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 02/04/2022] [Indexed: 11/18/2022] Open
Abstract
The house mouse or Mus musculus has become a premier mammalian model for genetic research due to its genetic and physiological similarities to humans. It brought mechanistic insights into numerous human diseases and has been routinely used to assess drug efficiency and toxicity, as well as to predict patient responses. To facilitate molecular mechanism studies in mouse, we present the Mouse Interactome Database (MID, Version 1), which includes 155,887 putative functional associations between mouse protein-coding genes inferred from functional association evidence integrated from 9 public databases. These putative functional associations are expected to cover 19.32% of all mouse protein interactions, and 26.02% of these function associations may represent protein interactions. On top of MID, we developed a gene set linkage analysis (GSLA) web tool to annotate potential functional impacts from observed differentially expressed genes. Two case studies show that the MID/GSLA system provided precise and informative annotations that other widely used gene set annotation tools, such as PANTHER and DAVID, did not. Both MID and GSLA are accessible through the website http://mouse.biomedtzc.cn.
Collapse
Affiliation(s)
- Hai-Bo Zhang
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Xiao-Bao Ding
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Jie Jin
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Wen-Ping Guo
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Qiao-Lei Yang
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng-Cheng Chen
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Heng Yao
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Ruan
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
| | - Yu-Tian Tao
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
- * E-mail: (YTT); (XC)
| | - Xin Chen
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics & Information Engineering, Taizhou University, Taizhou, China
- Institute of Pharmaceutical Biotechnology, School of Medicine, Zhejiang University, Hangzhou, China
- Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou, China
- * E-mail: (YTT); (XC)
| |
Collapse
|
8
|
Abstract
Since the large-scale experimental characterization of protein–protein interactions (PPIs) is not possible for all species, several computational PPI prediction methods have been developed that harness existing data from other species. While PPI network prediction has been extensively used in eukaryotes, microbial network inference has lagged behind. However, bacterial interactomes can be built using the same principles and techniques; in fact, several methods are better suited to bacterial genomes. These predicted networks allow systems-level analyses in species that lack experimental interaction data. This review describes the current network inference and analysis techniques and summarizes the use of computationally-predicted microbial interactomes to date.
Collapse
|
9
|
OUP accepted manuscript. Brief Funct Genomics 2022; 21:243-269. [DOI: 10.1093/bfgp/elac007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/14/2022] Open
|
10
|
Omranian S, Angeleska A, Nikoloski Z. Efficient and accurate identification of protein complexes from protein-protein interaction networks based on the clustering coefficient. Comput Struct Biotechnol J 2021; 19:5255-5263. [PMID: 34630943 PMCID: PMC8479235 DOI: 10.1016/j.csbj.2021.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 12/23/2022] Open
Abstract
Provided a family of efficient network algorithms for protein complex identification. The parameter-free family outperforms existing approaches on different networks. It exactly recovered ~ 35% of protein complexes in a pan-plant PPI network. We examined of network perturbations on predicted protein complexes.
Identification of protein complexes from protein-protein interaction (PPI) networks is a key problem in PPI mining, solved by parameter-dependent approaches that suffer from small recall rates. Here we introduce GCC-v, a family of efficient, parameter-free algorithms to accurately predict protein complexes using the (weighted) clustering coefficient of proteins in PPI networks. Through comparative analyses with gold standards and PPI networks from Escherichia coli, Saccharomyces cerevisiae, and Homo sapiens, we demonstrate that GCC-v outperforms twelve state-of-the-art approaches for identification of protein complexes with respect to twelve performance measures in at least 85.71% of scenarios. We also show that GCC-v results in the exact recovery of ∼35% of protein complexes in a pan-plant PPI network and discover 144 new protein complexes in Arabidopsis thaliana, with high support from GO semantic similarity. Our results indicate that findings from GCC-v are robust to network perturbations, which has direct implications to assess the impact of the PPI network quality on the predicted protein complexes.
Collapse
Affiliation(s)
- Sara Omranian
- Bioinformatics, Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany.,Systems Biology and Mathematical Modeling, Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam, Germany
| | | | - Zoran Nikoloski
- Bioinformatics, Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany.,Systems Biology and Mathematical Modeling, Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam, Germany
| |
Collapse
|
11
|
Wang J, Liu R, Mo H, Xiao X, Xu Q, Zhao W. Deubiquitinase PSMD7 promotes the proliferation, invasion, and cisplatin resistance of gastric cancer cells by stabilizing RAD23B. Int J Biol Sci 2021; 17:3331-3342. [PMID: 34512150 PMCID: PMC8416741 DOI: 10.7150/ijbs.61128] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/20/2021] [Indexed: 11/05/2022] Open
Abstract
Ubiquitination, a crucial post-translational modification, controls substrate degradation and can be reversed by deubiquitinases (DUBs). An increasing number of studies are showing that DUBs regulate the malignant behavior and chemotherapy resistance of gastric cancer (GC) by stabilizing various proteins. However, the expression level and biological function of the DUB, proteasome 26S subunit, non-ATPase 7 (PSMD7), in GC remains unknown. Herein, we report for the first time that PSMD7 is frequently overexpressed in GC tissues. Elevated levels of PSMD7 were also detected in GC cell lines. Notably, the upregulation of PSMD7 closely correlated with malignant clinical parameters and reduced the survival of GC patients. Functionally, we found that PSMD7 knockdown consistently suppressed the proliferation, migration, and invasion of AGS and SGC-7901 cells. Ectopic expression of PSMD7 facilitated GC cell proliferation and mobility. Based on protein-protein interaction prediction, RAD23 homolog B (RAD23B) protein was identified as a candidate substrate of PSMD7. PSMD7 positively regulated the abundance of RAD23B and xeroderma pigmentosum, complementation group C (XPC) protein in GC cells. The interaction between PSMD7 and RAD23B was confirmed using protein immunoprecipitation. PSMD7 knockdown enhanced the ubiquitination and degradation of RAD23B protein in GC cells. PSMD7 promoted cell viability, apoptosis resistance, and DNA damage repair in GC cells upon cisplatin (DDP) treatment. Moreover, PSMD7 silencing inhibited tumor growth and enhanced the sensitivity of GC cells to DDP treatment in mice. In summary, PSMD7 was highly expressed in GC and contributed to the malignant behavior and DDP resistance of tumor cells by stabilizing RAD23B.
Collapse
Affiliation(s)
- Jianjiang Wang
- Department of Hepatobiliary Surgery, The First People's Hospital of Hangzhou Lin'an District, Affiliated Lin'an People's Hospital, Hangzhou Medical College, Hangzhou 311399, China
| | - Runkun Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Huanye Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuelian Xiao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
12
|
Kuleshov MV, Xie Z, London ABK, Yang J, Evangelista J, Lachmann A, Shu I, Torre D, Ma’ayan A. KEA3: improved kinase enrichment analysis via data integration. Nucleic Acids Res 2021; 49:W304-W316. [PMID: 34019655 PMCID: PMC8265130 DOI: 10.1093/nar/gkab359] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 12/27/2022] Open
Abstract
Phosphoproteomics and proteomics experiments capture a global snapshot of the cellular signaling network, but these methods do not directly measure kinase state. Kinase Enrichment Analysis 3 (KEA3) is a webserver application that infers overrepresentation of upstream kinases whose putative substrates are in a user-inputted list of proteins. KEA3 can be applied to analyze data from phosphoproteomics and proteomics studies to predict the upstream kinases responsible for observed differential phosphorylations. The KEA3 background database contains measured and predicted kinase-substrate interactions (KSI), kinase-protein interactions (KPI), and interactions supported by co-expression and co-occurrence data. To benchmark the performance of KEA3, we examined whether KEA3 can predict the perturbed kinase from single-kinase perturbation followed by gene expression experiments, and phosphoproteomics data collected from kinase-targeting small molecules. We show that integrating KSIs and KPIs across data sources to produce a composite ranking improves the recovery of the expected kinase. The KEA3 webserver is available at https://maayanlab.cloud/kea3.
Collapse
Affiliation(s)
- Maxim V Kuleshov
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Zhuorui Xie
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Alexandra B K London
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Janice Yang
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - John Erol Evangelista
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Alexander Lachmann
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Ingrid Shu
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Denis Torre
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Avi Ma’ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| |
Collapse
|
13
|
Raimondi D, Simm J, Arany A, Moreau Y. A novel method for data fusion over Entity-Relation graphs and its application to protein-protein interaction prediction. Bioinformatics 2021; 37:2275-2281. [PMID: 33560405 DOI: 10.1093/bioinformatics/btab092] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/14/2021] [Accepted: 02/04/2021] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Modern Bioinformatics is facing increasingly complex problems to solve, and we are indeed rapidly approaching an era in which the ability to seamlessly integrate heterogeneous sources of information will be crucial for the scientific progress. Here we present a novel non-linear data fusion framework that generalizes the conventional Matrix Factorization paradigm allowing inference over arbitrary Entity-Relation graphs, and we applied it to the prediction of Protein-Protein Interactions (PPIs). Improving our knowledge of Protein Protein Interaction (PPI) networks at the proteome scale is indeed crucial to understand protein function, physiological and disease states and cell life in general. RESULTS We devised three data-fusion based models for the proteome-level prediction of PPIs, and we show that our method outperforms state of the art approaches on common benchmarks. Moreover, we investigate its predictions on newly published PPIs, showing that this new data has a clear shift in its underlying distributions and we thus train and test our models on this extended dataset. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Jaak Simm
- ESAT-STADIUS, KU Leuven, 3001 Leuven, Belgium
| | - Adam Arany
- ESAT-STADIUS, KU Leuven, 3001 Leuven, Belgium
| | - Yves Moreau
- ESAT-STADIUS, KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
14
|
Slater O, Miller B, Kontoyianni M. Decoding Protein-protein Interactions: An Overview. Curr Top Med Chem 2021; 20:855-882. [PMID: 32101126 DOI: 10.2174/1568026620666200226105312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022]
Abstract
Drug discovery has focused on the paradigm "one drug, one target" for a long time. However, small molecules can act at multiple macromolecular targets, which serves as the basis for drug repurposing. In an effort to expand the target space, and given advances in X-ray crystallography, protein-protein interactions have become an emerging focus area of drug discovery enterprises. Proteins interact with other biomolecules and it is this intricate network of interactions that determines the behavior of the system and its biological processes. In this review, we briefly discuss networks in disease, followed by computational methods for protein-protein complex prediction. Computational methodologies and techniques employed towards objectives such as protein-protein docking, protein-protein interactions, and interface predictions are described extensively. Docking aims at producing a complex between proteins, while interface predictions identify a subset of residues on one protein that could interact with a partner, and protein-protein interaction sites address whether two proteins interact. In addition, approaches to predict hot spots and binding sites are presented along with a representative example of our internal project on the chemokine CXC receptor 3 B-isoform and predictive modeling with IP10 and PF4.
Collapse
Affiliation(s)
- Olivia Slater
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Bethany Miller
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Maria Kontoyianni
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| |
Collapse
|
15
|
Wang JH, Chen YH. Interaction screening by Kendall's partial correlation for ultrahigh-dimensional data with survival trait. Bioinformatics 2020; 36:2763-2769. [PMID: 31926011 DOI: 10.1093/bioinformatics/btaa017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/06/2019] [Accepted: 01/07/2020] [Indexed: 12/14/2022] Open
Abstract
MOTIVATION In gene expression and genome-wide association studies, the identification of interaction effects is an important and challenging issue owing to its ultrahigh-dimensional nature. In particular, contaminated data and right-censored survival outcome make the associated feature screening even challenging. RESULTS In this article, we propose an inverse probability-of-censoring weighted Kendall's tau statistic to measure association of a survival trait with biomarkers, as well as a Kendall's partial correlation statistic to measure the relationship of a survival trait with an interaction variable conditional on the main effects. The Kendall's partial correlation is then used to conduct interaction screening. Simulation studies under various scenarios are performed to compare the performance of our proposal with some commonly available methods. In the real data application, we utilize our proposed method to identify epistasis associated with the clinical survival outcomes of non-small-cell lung cancer, diffuse large B-cell lymphoma and lung adenocarcinoma patients. Both simulation and real data studies demonstrate that our method performs well and outperforms existing methods in identifying main and interaction biomarkers. AVAILABILITY AND IMPLEMENTATION R-package 'IPCWK' is available to implement this method, together with a reference manual describing how to perform the 'IPCWK' package. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jie-Huei Wang
- Department of Statistics, Feng Chia University, Taichung 40724, Taiwan
| | - Yi-Hau Chen
- Institute of Statistical Science, Academia Sinica, Nankang, Taipei 11529, Taiwan
| |
Collapse
|
16
|
Poverennaya EV, Kiseleva OI, Ivanov AS, Ponomarenko EA. Methods of Computational Interactomics for Investigating Interactions of Human Proteoforms. BIOCHEMISTRY (MOSCOW) 2020; 85:68-79. [PMID: 32079518 DOI: 10.1134/s000629792001006x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human genome contains ca. 20,000 protein-coding genes that could be translated into millions of unique protein species (proteoforms). Proteoforms coded by a single gene often have different functions, which implies different protein partners. By interacting with each other, proteoforms create a network reflecting the dynamics of cellular processes in an organism. Perturbations of protein-protein interactions change the network topology, which often triggers pathological processes. Studying proteoforms is a relatively new research area in proteomics, and this is why there are comparatively few experimental studies on the interaction of proteoforms. Bioinformatics tools can facilitate such studies by providing valuable complementary information to the experimental data and, in particular, expanding the possibilities of the studies of proteoform interactions.
Collapse
Affiliation(s)
| | - O I Kiseleva
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | - A S Ivanov
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | | |
Collapse
|
17
|
Yang X, Yang S, Li Q, Wuchty S, Zhang Z. Prediction of human-virus protein-protein interactions through a sequence embedding-based machine learning method. Comput Struct Biotechnol J 2019; 18:153-161. [PMID: 31969974 PMCID: PMC6961065 DOI: 10.1016/j.csbj.2019.12.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
The identification of human-virus protein-protein interactions (PPIs) is an essential and challenging research topic, potentially providing a mechanistic understanding of viral infection. Given that the experimental determination of human-virus PPIs is time-consuming and labor-intensive, computational methods are playing an important role in providing testable hypotheses, complementing the determination of large-scale interactome between species. In this work, we applied an unsupervised sequence embedding technique (doc2vec) to represent protein sequences as rich feature vectors of low dimensionality. Training a Random Forest (RF) classifier through a training dataset that covers known PPIs between human and all viruses, we obtained excellent predictive accuracy outperforming various combinations of machine learning algorithms and commonly-used sequence encoding schemes. Rigorous comparison with three existing human-virus PPI prediction methods, our proposed computational framework further provided very competitive and promising performance, suggesting that the doc2vec encoding scheme effectively captures context information of protein sequences, pertaining to corresponding protein-protein interactions. Our approach is freely accessible through our web server as part of our host-pathogen PPI prediction platform (http://zzdlab.com/InterSPPI/). Taken together, we hope the current work not only contributes a useful predictor to accelerate the exploration of human-virus PPIs, but also provides some meaningful insights into human-virus relationships.
Collapse
Key Words
- AC, Auto Covariance
- ACC, Accuracy
- AUC, area under the ROC curve
- AUPRC, area under the PR curve
- Adaboost, Adaptive Boosting
- CT, Conjoint Triad
- Doc2vec
- Embedding
- Human-virus interaction
- LD, Local Descriptor
- MCC, Matthews correlation coefficient
- ML, machine learning
- MLP, Multiple Layer Perceptron
- MS, mass spectroscopy
- Machine learning
- PPIs, protein-protein interactions
- PR, Precision-Recall
- Prediction
- Protein-protein interaction
- RBF, radial basis function
- RF, Random Forest
- ROC, Receiver Operating Characteristic
- SGD, stochastic gradient descent
- SVM, Support Vector Machine
- Y2H, yeast two-hybrid
Collapse
Affiliation(s)
- Xiaodi Yang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shiping Yang
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qinmengge Li
- National Demonstration Center for Experimental Biological Sciences Education, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Stefan Wuchty
- Dept. of Computer Science, University of Miami, Miami, FL 33146, USA
- Dept. of Biology, University of Miami, Miami, FL 33146, USA
- Center of Computational Science, University of Miami, Miami, FL 33146, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Ziding Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
18
|
Dimitrakopoulos C, Hindupur SK, Häfliger L, Behr J, Montazeri H, Hall MN, Beerenwinkel N. Network-based integration of multi-omics data for prioritizing cancer genes. Bioinformatics 2019; 34:2441-2448. [PMID: 29547932 PMCID: PMC6041755 DOI: 10.1093/bioinformatics/bty148] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
Motivation Several molecular events are known to be cancer-related, including genomic aberrations, hypermethylation of gene promoter regions and differential expression of microRNAs. These aberration events are very heterogeneous across tumors and it is poorly understood how they affect the molecular makeup of the cell, including the transcriptome and proteome. Protein interaction networks can help decode the functional relationship between aberration events and changes in gene and protein expression. Results We developed NetICS (Network-based Integration of Multi-omics Data), a new graph diffusion-based method for prioritizing cancer genes by integrating diverse molecular data types on a directed functional interaction network. NetICS prioritizes genes by their mediator effect, defined as the proximity of the gene to upstream aberration events and to downstream differentially expressed genes and proteins in an interaction network. Genes are prioritized for individual samples separately and integrated using a robust rank aggregation technique. NetICS provides a comprehensive computational framework that can aid in explaining the heterogeneity of aberration events by their functional convergence to common differentially expressed genes and proteins. We demonstrate NetICS’ competitive performance in predicting known cancer genes and in generating robust gene lists using TCGA data from five cancer types. Availability and implementation NetICS is available at https://github.com/cbg-ethz/netics. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Christos Dimitrakopoulos
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | | | - Luca Häfliger
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jonas Behr
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Hesam Montazeri
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | | | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
19
|
Sarkar D, Saha S. Machine-learning techniques for the prediction of protein-protein interactions. J Biosci 2019; 44:104. [PMID: 31502581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Protein-protein interactions (PPIs) are important for the study of protein functions and pathways involved in different biological processes, as well as for understanding the cause and progression of diseases. Several high-throughput experimental techniques have been employed for the identification of PPIs in a few model organisms, but still, there is a huge gap in identifying all possible binary PPIs in an organism. Therefore, PPI prediction using machine-learning algorithms has been used in conjunction with experimental methods for discovery of novel protein interactions. The two most popular supervised machine-learning techniques used in the prediction of PPIs are support vector machines and random forest classifiers. Bayesian-probabilistic inference has also been used but mainly for the scoring of high-throughput PPI dataset confidence measures. Recently, deep-learning algorithms have been used for sequence-based prediction of PPIs. Several clustering methods such as hierarchical and k-means are useful as unsupervised machine-learning algorithms for the prediction of interacting protein pairs without explicit data labelling. In summary, machine-learning techniques have been widely used for the prediction of PPIs thus allowing experimental researchers to study cellular PPI networks.
Collapse
|
20
|
Di Nanni N, Gnocchi M, Moscatelli M, Milanesi L, Mosca E. Gene relevance based on multiple evidences in complex networks. Bioinformatics 2019; 36:865-871. [PMID: 31504182 PMCID: PMC9883679 DOI: 10.1093/bioinformatics/btz652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/17/2019] [Accepted: 08/19/2019] [Indexed: 02/02/2023] Open
Abstract
MOTIVATION Multi-omics approaches offer the opportunity to reconstruct a more complete picture of the molecular events associated with human diseases, but pose challenges in data analysis. Network-based methods for the analysis of multi-omics leverage the complex web of macromolecular interactions occurring within cells to extract significant patterns of molecular alterations. Existing network-based approaches typically address specific combinations of omics and are limited in terms of the number of layers that can be jointly analysed. In this study, we investigate the application of network diffusion to quantify gene relevance on the basis of multiple evidences (layers). RESULTS We introduce a gene score (mND) that quantifies the relevance of a gene in a biological process taking into account the network proximity of the gene and its first neighbours to other altered genes. We show that mND has a better performance over existing methods in finding altered genes in network proximity in one or more layers. We also report good performances in recovering known cancer genes. The pipeline described in this article is broadly applicable, because it can handle different types of inputs: in addition to multi-omics datasets, datasets that are stratified in many classes (e.g., cell clusters emerging from single cell analyses) or a combination of the two scenarios. AVAILABILITY AND IMPLEMENTATION The R package 'mND' is available at URL: https://www.itb.cnr.it/mnd. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Noemi Di Nanni
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy,Department of Industrial and Information Engineering, University of Pavia, Italy
| | - Matteo Gnocchi
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | - Marco Moscatelli
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | - Luciano Milanesi
- Department of Biomedical Sciences, Institute of Biomedical Technologies, National Research Council, 20090 Segrate (MI), Italy
| | | |
Collapse
|
21
|
Sarkar D, Saha S. Machine-learning techniques for the prediction of protein–protein interactions. J Biosci 2019. [DOI: 10.1007/s12038-019-9909-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Sumonja N, Gemovic B, Veljkovic N, Perovic V. Automated feature engineering improves prediction of protein-protein interactions. Amino Acids 2019; 51:1187-1200. [PMID: 31278492 DOI: 10.1007/s00726-019-02756-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 06/26/2019] [Indexed: 10/26/2022]
Abstract
Over the last decade, various machine learning (ML) and statistical approaches for protein-protein interaction (PPI) predictions have been developed to help annotating functional interactions among proteins, essential for our system-level understanding of life. Efficient ML approaches require informative and non-redundant features. In this paper, we introduce novel types of expert-crafted sequence, evolutionary and graph features and apply automatic feature engineering to further expand feature space to improve predictive modeling. The two-step automatic feature-engineering process encompasses the hybrid method for feature generation and unsupervised feature selection, followed by supervised feature selection through a genetic algorithm (GA). The optimization of both steps allows the feature-engineering procedure to operate on a large transformed feature space with no considerable computational cost and to efficiently provide newly engineered features. Based on GA and correlation filtering, we developed a stacking algorithm GA-STACK for automatic ensembling of different ML algorithms to improve prediction performance. We introduced a unified method, HP-GAS, for the prediction of human PPIs, which incorporates GA-STACK and rests on both expert-crafted and 40% of newly engineered features. The extensive cross validation and comparison with the state-of-the-art methods showed that HP-GAS represents currently the most efficient method for proteome-wide forecasting of protein interactions, with prediction efficacy of 0.93 AUC and 0.85 accuracy. We implemented the HP-GAS method as a free standalone application which is a time-efficient and easy-to-use tool. HP-GAS software with supplementary data can be downloaded from: http://www.vinca.rs/180/tools/HP-GAS.php .
Collapse
Affiliation(s)
- Neven Sumonja
- Laboratory for Bioinformatics and Computational Chemistry, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, Vinca, Belgrade, 11351, Serbia
| | - Branislava Gemovic
- Laboratory for Bioinformatics and Computational Chemistry, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, Vinca, Belgrade, 11351, Serbia
| | - Nevena Veljkovic
- Laboratory for Bioinformatics and Computational Chemistry, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, Vinca, Belgrade, 11351, Serbia
| | - Vladimir Perovic
- Laboratory for Bioinformatics and Computational Chemistry, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, Vinca, Belgrade, 11351, Serbia.
| |
Collapse
|
23
|
Hamel-Côté G, Lapointe F, Véronneau S, Mayhue M, Rola-Pleszczynski M, Stankova J. Regulation of platelet-activating factor-mediated interleukin-6 promoter activation by the 48 kDa but not the 45 kDa isoform of protein tyrosine phosphatase non-receptor type 2. Cell Biosci 2019; 9:51. [PMID: 31289638 PMCID: PMC6593612 DOI: 10.1186/s13578-019-0316-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background An underlying state of inflammation is thought to be an important cause of cardiovascular disease. Among cells involved in the early steps of atherosclerosis, monocyte-derived dendritic cells (Mo-DCs) respond to inflammatory stimuli, including platelet-activating factor (PAF), by the induction of various cytokines, such as interleukin 6 (IL-6). PAF is a potent phospholipid mediator involved in both the onset and progression of atherosclerosis. It mediates its effects by binding to its cognate G-protein coupled receptor, PAFR. Activation of PAFR-induced signaling pathways is tightly coordinated to ensure specific cell responses. Results Here, we report that PAF stimulated the phosphatase activity of both the 45 and 48 kDa isoforms of the protein tyrosine phosphatase non-receptor type 2 (PTPN2). However, we found that only the 48 kDa PTPN2 isoform has a role in PAFR-induced signal transduction, leading to activation of the IL-6 promoter. In luciferase reporter assays, expression of the 48 kDa, but not the 45 kDa, PTPN2 isoform increased human IL-6 (hIL-6) promoter activity by 40% after PAF stimulation of HEK-293 cells, stably transfected with PAFR (HEK-PAFR). Our results suggest that the differential localization of the PTPN2 isoforms and the differences in PAF-induced phosphatase activation may contribute to the divergent modulation of PAF-induced IL-6 promoter activation. The involvement of PTPN2 in PAF-induced IL-6 expression was confirmed in immature Mo-DCs (iMo-DCs), using siRNAs targeting the two isoforms of PTPN2, where siRNAs against the 48 kDa PTPN2 significantly inhibited PAF-stimulated IL-6 mRNA expression. Pharmacological inhibition of several signaling pathways suggested a role for PTPN2 in early signaling events. Results obtained by Western blot confirmed that PTPN2 increased the activation of the PI3K/Akt pathway via the modulation of protein kinase D (PKD) activity. WT PKD expression counteracted the effect of PTPN2 on PAF-induced IL-6 promoter transactivation and phosphorylation of Akt. Using siRNAs targeting the individual isoforms of PTPN2, we confirmed that these pathways were also active in iMo-DCs. Conclusion Taken together, our data suggest that PTPN2, in an isoform-specific manner, could be involved in the positive regulation of PI3K/Akt activation, via the modulation of PKD activity, allowing for the maximal induction of PAF-stimulated IL-6 mRNA expression.
Collapse
Affiliation(s)
- Geneviève Hamel-Côté
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Fanny Lapointe
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Steeve Véronneau
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Marian Mayhue
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Marek Rola-Pleszczynski
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| | - Jana Stankova
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC Canada
| |
Collapse
|
24
|
Rasti S, Vogiatzis C. A survey of computational methods in protein–protein interaction networks. ANNALS OF OPERATIONS RESEARCH 2019; 276:35-87. [DOI: 10.1007/s10479-018-2956-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
25
|
Kotlyar M, Pastrello C, Rossos AE, Jurisica I. Protein–Protein Interaction Databases. ENCYCLOPEDIA OF BIOINFORMATICS AND COMPUTATIONAL BIOLOGY 2019:988-996. [DOI: 10.1016/b978-0-12-809633-8.20495-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Ding Z, Kihara D. Computational Methods for Predicting Protein-Protein Interactions Using Various Protein Features. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2018; 93:e62. [PMID: 29927082 PMCID: PMC6097941 DOI: 10.1002/cpps.62] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Understanding protein-protein interactions (PPIs) in a cell is essential for learning protein functions, pathways, and mechanism of diseases. PPIs are also important targets for developing drugs. Experimental methods, both small-scale and large-scale, have identified PPIs in several model organisms. However, results cover only a part of PPIs of organisms; moreover, there are many organisms whose PPIs have not yet been investigated. To complement experimental methods, many computational methods have been developed that predict PPIs from various characteristics of proteins. Here we provide an overview of literature reports to classify computational PPI prediction methods that consider different features of proteins, including protein sequence, genomes, protein structure, function, PPI network topology, and those which integrate multiple methods. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Ziyun Ding
- Department of Biological Science, Purdue University, West Lafayette, IN, 47907 USA
| | - Daisuke Kihara
- Department of Biological Science, Purdue University, West Lafayette, IN, 47907 USA
- Department of Computer Science, Purdue University, West Lafayette, IN, 47907 USA
- Corresponding author: DK; , Phone: 1-765-496-2284 (DK)
| |
Collapse
|
27
|
Roth A, Subramanian S, Ganapathiraju MK. Towards Extracting Supporting Information About Predicted Protein-Protein Interactions. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:1239-1246. [PMID: 26672046 DOI: 10.1109/tcbb.2015.2505278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
One of the goals of relation extraction is to identify protein-protein interactions (PPIs) in biomedical literature. Current systems are capturing binary relations and also the direction and type of an interaction. Besides assisting in the curation PPIs into databases, there has been little real-world application of these algorithms. We describe UPSITE, a text mining tool for extracting evidence in support of a hypothesized interaction. Given a predicted PPI, UPSITE uses a binary relation detector to check whether a PPI is found in abstracts in PubMed. If it is not found, UPSITE retrieves documents relevant to each of the two proteins separately, and extracts contextual information about biological events surrounding each protein, and calculates semantic similarity of the two proteins to provide evidential support for the predicted PPI. In evaluations, relation extraction achieved an Fscore of 0.88 on the HPRD50 corpus, and semantic similarity measured with angular distance was found to be statistically significant. With the development of PPI prediction algorithms, the burden of interpreting the validity and relevance of novel PPIs is on biologists. We suggest that presenting annotations of the two proteins in a PPI side-by-side and a score that quantifies their similarity lessens this burden to some extent.
Collapse
|
28
|
Tong R, Yang B, Xiao H, Peng C, Hu W, Weng X, Cheng S, Du C, Lv Z, Ding C, Zhou L, Xie H, Wu J, Zheng S. KCTD11 inhibits growth and metastasis of hepatocellular carcinoma through activating Hippo signaling. Oncotarget 2018; 8:37717-37729. [PMID: 28465479 PMCID: PMC5514943 DOI: 10.18632/oncotarget.17145] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 03/29/2017] [Indexed: 02/05/2023] Open
Abstract
A lack of effective prognostic biomarkers and molecular targets is a serious problem in hepatocellular carcinoma. KCTD11, reported as a tumor suppressor, are still not well understood. In this study, KCTD11 was found low-expressed in HCC tissues and cell lines. The HCC patients with low expression of KCTD11 suggested shorter overall survival. We found KCTD11 inhibiting cell proliferation in vitro and tumor growth in vivo, by activating p21 and repressing cycle related proteins. KCTD11 also inhibited cell adhesion by decreasing CTGF and CLDN1. We found CTGF binding COL3A1 in HCCLM3, which might lead to reduction of COL3A1 expression. KCTD11 also inhibited cell migration and invasion in HCC, by repressing MMPs and EMT. We found the tumor suppression function of KCTD11 was at least partly through activating Hippo pathway in HCC. Base on the enhanced Hippo pathway, KCTD11 could activate p21 by stabilizing p53 or promoting the MST1/ GSK3β/p21 signaling in HCC. Overall, these results suggest that KCTD11 works as a tumor suppressor and owns prognostic and therapeutic potentials in HCC.
Collapse
Affiliation(s)
- Rongliang Tong
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China
| | - Beng Yang
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou 310000, China
| | - Heng Xiao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Chuanhui Peng
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China
| | - Wendi Hu
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China
| | - Xiaoyu Weng
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China
| | - Shaobing Cheng
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China
| | - Chengli Du
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China
| | - Zhen Lv
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Chaofeng Ding
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China
| | - Lin Zhou
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310000, China
| | - Haiyang Xie
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310000, China
| | - Jian Wu
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310000, China
| | - Shusen Zheng
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, China.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou 310000, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310000, China
| |
Collapse
|
29
|
Kotlyar M, Rossos AEM, Jurisica I. Prediction of Protein-Protein Interactions. ACTA ACUST UNITED AC 2017; 60:8.2.1-8.2.14. [PMID: 29220074 DOI: 10.1002/cpbi.38] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The authors provide an overview of physical protein-protein interaction prediction, covering the main strategies for predicting interactions, approaches for assessing predictions, and online resources for accessing predictions. This unit focuses on the main advancements in each of these areas over the last decade. The methods and resources that are presented here are not an exhaustive set, but characterize the current state of the field-highlighting key challenges and achievements. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Max Kotlyar
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Andrea E M Rossos
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Igor Jurisica
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Departments of Medical Biophysics and Computer Science, University of Toronto, Ontario, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
30
|
Ur Rehman H, Bari I, Ali A, Mahmood H. A Bayesian approach for estimating protein-protein interactions by integrating structural and non-structural biological data. MOLECULAR BIOSYSTEMS 2017; 13:2592-2602. [PMID: 29028065 DOI: 10.1039/c7mb00484b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Accurate elucidation of genome wide protein-protein interactions is crucial for understanding the regulatory processes of the cell. High-throughput techniques, such as the yeast-2-hybrid (Y2H) assay, co-immunoprecipitation (co-IP), mass spectrometric (MS) protein complex identification, affinity purification (AP) etc., are generally relied upon to determine protein interactions. Unfortunately, each type of method is inherently subject to different types of noise and results in false positive interactions. On the other hand, precise understanding of proteins, especially knowledge of their functional associations is necessary for understanding how complex molecular machines function. To solve this problem, computational techniques are generally relied upon to precisely predict protein interactions. In this work, we present a novel method that combines structural and non-structural biological data to precisely predict protein interactions. The conceptual novelty of our approach lies in identifying and precisely associating biological information that provides substantial interaction clues. Our model combines structural and non-structural information using Bayesian statistics to calculate the likelihood of each interaction. The proposed model is tested on Saccharomyces cerevisiae's interactions extracted from the DIP and IntAct databases and provides substantial improvements in terms of accuracy, precision, recall and F1 score, as compared with the most widely used related state-of-the-art techniques.
Collapse
Affiliation(s)
- Hafeez Ur Rehman
- Department of Computer Science, FAST National University of Computer & Emerging Sciences, Peshawar, Pakistan.
| | | | | | | |
Collapse
|
31
|
Calderón-González KG, Hernández-Monge J, Herrera-Aguirre ME, Luna-Arias JP. Bioinformatics Tools for Proteomics Data Interpretation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 919:281-341. [PMID: 27975225 DOI: 10.1007/978-3-319-41448-5_16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biological systems function via intricate cellular processes and networks in which RNAs, metabolites, proteins and other cellular compounds have a precise role and are exquisitely regulated (Kumar and Mann, FEBS Lett 583(11):1703-1712, 2009). The development of high-throughput technologies, such as the Next Generation DNA Sequencing (NGS) and DNA microarrays for sequencing genomes or metagenomes, have triggered a dramatic increase in the last few years in the amount of information stored in the GenBank and UniProt Knowledgebase (UniProtKB). GenBank release 210, reported in October 2015, contains 202,237,081,559 nucleotides corresponding to 188,372,017 sequences, whilst there are only 1,222,635,267,498 nucleotides corresponding to 309,198,943 sequences from Whole Genome Shotgun (WGS) projects. In the case of UniProKB/Swiss-Prot, release 2015_12 (December 9, 2015) contains 196,219,159 amino acids that correspond to 550,116 entries. Meanwhile, UniProtKB/TrEMBL (release 2015_12 of December 9 2015) contains 1,838,851,8871 amino acids corresponding to 555,270,679 entries. Proteomics has also improved our knowledge of proteins that are being expressed in cells at a certain time of the cell cycle. It has also allowed the identification of molecules forming part of multiprotein complexes and an increasing number of posttranslational modifications (PTMs) that are present in proteins, as well as the variants of proteins expressed.
Collapse
Affiliation(s)
- Karla Grisel Calderón-González
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360, Ciudad de México, Mexico
| | - Jesús Hernández-Monge
- Instituto de Física, Universidad Autónoma de San Luis Potosí, Av. Manuel Nava 6, Zona Universitaria, C.P. 78290, San Luis Potosí, S.L.P., Mexico
| | - María Esther Herrera-Aguirre
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360, Ciudad de México, Mexico
| | - Juan Pedro Luna-Arias
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360, Ciudad de México, Mexico.
| |
Collapse
|
32
|
Bioinformatics Resources for Interpreting Proteomics Mass Spectrometry Data. Methods Mol Biol 2017. [PMID: 28809010 DOI: 10.1007/978-1-4939-7201-2_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Developments in mass spectrometry (MS) instrumentation have supported the advance of a variety of proteomic technologies that have enabled scientists to assess differences between healthy and diseased states. In particular, the ability to identify altered biological processes in a cell has led to the identification of novel drug targets, the development of more effective therapeutic drugs, and the growth of new diagnostic approaches and tools for personalized medicine applications. Nevertheless, large-scale proteomic data generated by modern mass spectrometers are extremely complex and necessitate equally complex bioinformatics tools and computational algorithms for their interpretation. A vast number of commercial and public resources have been developed for this purpose, often leaving the researcher perplexed at the overwhelming list of choices that exist. To address this challenge, the aim of this chapter is to provide a roadmap to the basic steps that are involved in mass spectrometry data acquisition and processing, and to describe the most common tools that are available for placing the results in biological context.
Collapse
|
33
|
Zhou J. Effect of Protein Repetitiveness on Protein-Protein Interaction Prediction Results Using Support Vector Machines. J Comput Biol 2016; 24:183-192. [PMID: 27529135 DOI: 10.1089/cmb.2015.0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND There are many computational approaches to predict the protein-protein interactions using support vector machines (SVMs) with high performance. In fact, performance of currently reported methods are significantly over-estimated and affected by the object repetitiveness in the datasets used. OBJECTIVE To study the effect of object repetitiveness of datasets on predicting results. METHOD We present novel methods to construct different positive datasets with or without repeating proteins using graph maximum matching in the protein-protein interaction datasets and corresponding series of negative datasets with different proteins repetitiveness are constructed using graph adjacency matrix. The relationship between the SVM prediction results and the repeated proteins (repeat numbers and repeat rates) and the distributions of repeated proteins in the datasets are analyzed. RESULTS Protein repetitiveness of positive and negative datasets can affect the prediction result: high protein repetitiveness of positive or negative datasets yield high performance prediction result. CONCLUSION This indicate that dealing with object repetitiveness of datasets is a key issue in protein-protein interactions prediction using SVMs since real world data contain certain degrees of repeat proteins.
Collapse
Affiliation(s)
- Jie Zhou
- Guangdong Province Key Laboratory of Computer Network, School of Computer Science and Engineering, South China University of Technology , Guangzhou, China
| |
Collapse
|
34
|
Marei H, Carpy A, Macek B, Malliri A. Proteomic analysis of Rac1 signaling regulation by guanine nucleotide exchange factors. Cell Cycle 2016; 15:1961-74. [PMID: 27152953 PMCID: PMC4968972 DOI: 10.1080/15384101.2016.1183852] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/13/2016] [Accepted: 04/22/2016] [Indexed: 10/30/2022] Open
Abstract
The small GTPase Rac1 is implicated in various cellular processes that are essential for normal cell function. Deregulation of Rac1 signaling has also been linked to a number of diseases, including cancer. The diversity of Rac1 functioning in cells is mainly attributed to its ability to bind to a multitude of downstream effectors following activation by Guanine nucleotide Exchange Factors (GEFs). Despite the identification of a large number of Rac1 binding partners, factors influencing downstream specificity are poorly defined, thus hindering the detailed understanding of both Rac1's normal and pathological functions. In a recent study, we demonstrated a role for 2 Rac-specific GEFs, Tiam1 and P-Rex1, in mediating Rac1 anti- versus pro-migratory effects, respectively. Importantly, via conducting a quantitative proteomic screen, we identified distinct changes in the Rac1 interactome following activation by either GEF, indicating that these opposing effects are mediated through GEF modulation of the Rac1 interactome. Here, we present the full list of identified Rac1 interactors together with functional annotation of the differentially regulated Rac1 binding partners. In light of this data, we also provide additional insights into known and novel signaling cascades that might account for the GEF-mediated Rac1-driven cellular effects.
Collapse
Affiliation(s)
- Hadir Marei
- Cell Signaling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Alejandro Carpy
- Proteome Center Tuebingen, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Boris Macek
- Proteome Center Tuebingen, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Angeliki Malliri
- Cell Signaling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| |
Collapse
|
35
|
Chen H, Shen J, Wang L, Song J. Towards Data Analytics of Pathogen-Host Protein-Protein Interaction: A Survey. 2016 IEEE INTERNATIONAL CONGRESS ON BIG DATA (BIGDATA CONGRESS) 2016:377-388. [DOI: 10.1109/bigdatacongress.2016.60] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Schizophrenia interactome with 504 novel protein-protein interactions. NPJ SCHIZOPHRENIA 2016; 2:16012. [PMID: 27336055 PMCID: PMC4898894 DOI: 10.1038/npjschz.2016.12] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 11/29/2022]
Abstract
Genome-wide association studies of schizophrenia (GWAS) have revealed the role of rare and common genetic variants, but the functional effects of the risk variants remain to be understood. Protein interactome-based studies can facilitate the study of molecular mechanisms by which the risk genes relate to schizophrenia (SZ) genesis, but protein–protein interactions (PPIs) are unknown for many of the liability genes. We developed a computational model to discover PPIs, which is found to be highly accurate according to computational evaluations and experimental validations of selected PPIs. We present here, 365 novel PPIs of liability genes identified by the SZ Working Group of the Psychiatric Genomics Consortium (PGC). Seventeen genes that had no previously known interactions have 57 novel interactions by our method. Among the new interactors are 19 drug targets that are targeted by 130 drugs. In addition, we computed 147 novel PPIs of 25 candidate genes investigated in the pre-GWAS era. While there is little overlap between the GWAS genes and the pre-GWAS genes, the interactomes reveal that they largely belong to the same pathways, thus reconciling the apparent disparities between the GWAS and prior gene association studies. The interactome including 504 novel PPIs overall, could motivate other systems biology studies and trials with repurposed drugs. The PPIs are made available on a webserver, called Schizo-Pi at http://severus.dbmi.pitt.edu/schizo-pi with advanced search capabilities.
Collapse
|
37
|
Marei H, Carpy A, Woroniuk A, Vennin C, White G, Timpson P, Macek B, Malliri A. Differential Rac1 signalling by guanine nucleotide exchange factors implicates FLII in regulating Rac1-driven cell migration. Nat Commun 2016; 7:10664. [PMID: 26887924 PMCID: PMC4759627 DOI: 10.1038/ncomms10664] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 01/08/2016] [Indexed: 01/22/2023] Open
Abstract
The small GTPase Rac1 has been implicated in the formation and dissemination of tumours. Upon activation by guanine nucleotide exchange factors (GEFs), Rac1 associates with a variety of proteins in the cell thereby regulating various functions, including cell migration. However, activation of Rac1 can lead to opposing migratory phenotypes raising the possibility of exacerbating tumour progression when targeting Rac1 in a clinical setting. This calls for the identification of factors that influence Rac1-driven cell motility. Here we show that Tiam1 and P-Rex1, two Rac GEFs, promote Rac1 anti- and pro-migratory signalling cascades, respectively, through regulating the Rac1 interactome. In particular, we demonstrate that P-Rex1 stimulates migration through enhancing the interaction between Rac1 and the actin-remodelling protein flightless-1 homologue, to modulate cell contraction in a RhoA-ROCK-independent manner.
Collapse
Affiliation(s)
- Hadir Marei
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M204BX, UK
| | - Alejandro Carpy
- Proteome Center Tuebingen, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen 72026, Germany
| | - Anna Woroniuk
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M204BX, UK
| | - Claire Vennin
- Invasion and Metastasis Group, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales 2010, Australia
| | - Gavin White
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M204BX, UK
| | - Paul Timpson
- Invasion and Metastasis Group, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Darlinghurst, New South Wales 2010, Australia
| | - Boris Macek
- Proteome Center Tuebingen, Interfaculty Institute for Cell Biology, University of Tuebingen, Tuebingen 72026, Germany
| | - Angeliki Malliri
- Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M204BX, UK
| |
Collapse
|
38
|
Gianazza E, Parravicini C, Primi R, Miller I, Eberini I. In silico prediction and characterization of protein post-translational modifications. J Proteomics 2015; 134:65-75. [PMID: 26436211 DOI: 10.1016/j.jprot.2015.09.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/17/2015] [Accepted: 09/23/2015] [Indexed: 01/06/2023]
Abstract
This review outlines the computational approaches and procedures for predicting post translational modification (PTM)-induced changes in protein conformation and their influence on protein function(s), the latter being assessed as differential affinity in interaction with either low (ligands for receptors or transporters, substrates for enzymes) or high molecular mass molecules (proteins or nucleic acids in supramolecular assemblies). The scope for an in silico approach is discussed against a summary of the in vitro evidence on the structural and functional outcome of protein PTM.
Collapse
Affiliation(s)
- Elisabetta Gianazza
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Gruppo di Studio per la Proteomica e la Struttura delle Proteine, Sezione di Scienze Farmacologiche, Via Balzaretti 9, I-20133 Milan, Italy.
| | - Chiara Parravicini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Laboratorio di Biochimica e Biofisica Computazionale, Sezione di Biochimica, Biofisica, Fisiologia ed Immunopatologia, Via Trentacoste, 2, I-20134 Milan, Italy
| | - Roberto Primi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Laboratorio di Biochimica e Biofisica Computazionale, Sezione di Biochimica, Biofisica, Fisiologia ed Immunopatologia, Via Trentacoste, 2, I-20134 Milan, Italy
| | - Ingrid Miller
- Institut für Medizinische Biochemie, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210 Vienna, Austria
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Laboratorio di Biochimica e Biofisica Computazionale, Sezione di Biochimica, Biofisica, Fisiologia ed Immunopatologia, Via Trentacoste, 2, I-20134 Milan, Italy
| |
Collapse
|
39
|
Pushing the annotation of cellular activities to a higher resolution: Predicting functions at the isoform level. Methods 2015; 93:110-8. [PMID: 26238263 DOI: 10.1016/j.ymeth.2015.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/20/2015] [Accepted: 07/29/2015] [Indexed: 12/23/2022] Open
Abstract
In past decades, the experimental determination of protein functions was expensive and time-consuming, so numerous computational methods were developed to speed up and guide the process. However, most of these methods predict protein functions at the gene level and do not consider the fact that protein isoforms (translated from alternatively spliced transcripts), not genes, are the actual function carriers. Now, high-throughput RNA-seq technology is providing unprecedented opportunities to unravel protein functions at the isoform level. In this article, we review recent progress in the high-resolution functional annotations of protein isoforms, focusing on two methods developed by the authors. Both methods can integrate multiple RNA-seq datasets for comprehensively characterizing functions of protein isoforms.
Collapse
|
40
|
Determination of Temporal Order among the Components of an Oscillatory System. PLoS One 2015; 10:e0124842. [PMID: 26151635 PMCID: PMC4495067 DOI: 10.1371/journal.pone.0124842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/17/2015] [Indexed: 11/19/2022] Open
Abstract
Oscillatory systems in biology are tightly regulated process where the individual components (e.g. genes) express in an orderly manner by virtue of their functions. The temporal order among the components of an oscillatory system may potentially be disrupted for various reasons (e.g. environmental factors). As a result some components of the system may go out of order or even cease to participate in the oscillatory process. In this article, we develop a novel framework to evaluate whether the temporal order is unchanged in different populations (or experimental conditions). We also develop methodology to estimate the order among the components with a suitable notion of “confidence.” Using publicly available data on S. pombe, S. cerevisiae and Homo sapiens we discover that the temporal order among the genes cdc18; mik1; hhf1; hta2; fkh2 and klp5 is evolutionarily conserved from yeast to humans.
Collapse
|
41
|
Surfing the Protein-Protein Interaction Surface Using Docking Methods: Application to the Design of PPI Inhibitors. Molecules 2015; 20:11569-603. [PMID: 26111183 PMCID: PMC6272567 DOI: 10.3390/molecules200611569] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/02/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Blocking protein-protein interactions (PPI) using small molecules or peptides modulates biochemical pathways and has therapeutic significance. PPI inhibition for designing drug-like molecules is a new area that has been explored extensively during the last decade. Considering the number of available PPI inhibitor databases and the limited number of 3D structures available for proteins, docking and scoring methods play a major role in designing PPI inhibitors as well as stabilizers. Docking methods are used in the design of PPI inhibitors at several stages of finding a lead compound, including modeling the protein complex, screening for hot spots on the protein-protein interaction interface and screening small molecules or peptides that bind to the PPI interface. There are three major challenges to the use of docking on the relatively flat surfaces of PPI. In this review we will provide some examples of the use of docking in PPI inhibitor design as well as its limitations. The combination of experimental and docking methods with improved scoring function has thus far resulted in few success stories of PPI inhibitors for therapeutic purposes. Docking algorithms used for PPI are in the early stages, however, and as more data are available docking will become a highly promising area in the design of PPI inhibitors or stabilizers.
Collapse
|
42
|
Laraia L, McKenzie G, Spring DR, Venkitaraman AR, Huggins DJ. Overcoming Chemical, Biological, and Computational Challenges in the Development of Inhibitors Targeting Protein-Protein Interactions. CHEMISTRY & BIOLOGY 2015; 22:689-703. [PMID: 26091166 PMCID: PMC4518475 DOI: 10.1016/j.chembiol.2015.04.019] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/01/2015] [Accepted: 04/08/2015] [Indexed: 01/19/2023]
Abstract
Protein-protein interactions (PPIs) underlie the majority of biological processes, signaling, and disease. Approaches to modulate PPIs with small molecules have therefore attracted increasing interest over the past decade. However, there are a number of challenges inherent in developing small-molecule PPI inhibitors that have prevented these approaches from reaching their full potential. From target validation to small-molecule screening and lead optimization, identifying therapeutically relevant PPIs that can be successfully modulated by small molecules is not a simple task. Following the recent review by Arkin et al., which summarized the lessons learnt from prior successes, we focus in this article on the specific challenges of developing PPI inhibitors and detail the recent advances in chemistry, biology, and computation that facilitate overcoming them. We conclude by providing a perspective on the field and outlining four innovations that we see as key enabling steps for successful development of small-molecule inhibitors targeting PPIs.
Collapse
Affiliation(s)
- Luca Laraia
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - Grahame McKenzie
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Ashok R Venkitaraman
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
| | - David J Huggins
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK; Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK; Theory of Condensed Matter Group, Cavendish Laboratory, University of Cambridge, 19 JJ Thomson Avenue, Cambridge CB3 0HE, UK.
| |
Collapse
|
43
|
Sharma AK, Khandelwal R, Sharma Y, Rajanikanth V. Secretagogin, a hexa EF-hand calcium-binding protein: high level bacterial overexpression, one-step purification and properties. Protein Expr Purif 2015; 109:113-9. [PMID: 25703053 DOI: 10.1016/j.pep.2015.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 12/22/2014] [Accepted: 02/11/2015] [Indexed: 12/20/2022]
Abstract
Secretagogin (SCGN), a hexa EF-hand calcium-binding protein, is highly expressed in the endocrine cells (especially in pancreatic islets) and in restricted neuronal sub-populations, albeit at comparatively low level. Since SCGN is predicted to be a potential neuroendocrine marker in carcinoid tumors of lung and gastrointestinal tract, it is of paramount importance to understand the features of this protein in different environment for assigning its crucial functions in different tissues and under pathophysiological conditions. To score out the limitation of protein for in vitro studies, we report a one-step, high purity and high level bacterial purification of secretagogin by refolding from the inclusion bodies yielding about 40mg protein per litre of bacterial culture. We also report previously undocumented Ca(2+)/Mg(2+) binding and hydrodynamic properties of secretagogin.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India.
| | - Radhika Khandelwal
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | - Yogendra Sharma
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
44
|
Tseng YT, Li W, Chen CH, Zhang S, Chen JJW, Zhou X, Liu CC. IIIDB: a database for isoform-isoform interactions and isoform network modules. BMC Genomics 2015; 16 Suppl 2:S10. [PMID: 25707505 PMCID: PMC4331710 DOI: 10.1186/1471-2164-16-s2-s10] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Protein-protein interactions (PPIs) are key to understanding diverse cellular processes and disease mechanisms. However, current PPI databases only provide low-resolution knowledge of PPIs, in the sense that "proteins" of currently known PPIs generally refer to "genes." It is known that alternative splicing often impacts PPI by either directly affecting protein interacting domains, or by indirectly impacting other domains, which, in turn, impacts the PPI binding. Thus, proteins translated from different isoforms of the same gene can have different interaction partners. RESULTS Due to the limitations of current experimental capacities, little data is available for PPIs at the resolution of isoforms, although such high-resolution data is crucial to map pathways and to understand protein functions. In fact, alternative splicing can often change the internal structure of a pathway by rearranging specific PPIs. To fill the gap, we systematically predicted genome-wide isoform-isoform interactions (IIIs) using RNA-seq datasets, domain-domain interaction and PPIs. Furthermore, we constructed an III database (IIIDB) that is a resource for studying PPIs at isoform resolution. To discover functional modules in the III network, we performed III network clustering, and then obtained 1025 isoform modules. To evaluate the module functionality, we performed the GO/pathway enrichment analysis for each isoform module. CONCLUSIONS The IIIDB provides predictions of human protein-protein interactions at the high resolution of transcript isoforms that can facilitate detailed understanding of protein functions and biological pathways. The web interface allows users to search for IIIs or III network modules. The IIIDB is freely available at http://syslab.nchu.edu.tw/IIIDB.
Collapse
|
45
|
Kotlyar M, Pastrello C, Pivetta F, Lo Sardo A, Cumbaa C, Li H, Naranian T, Niu Y, Ding Z, Vafaee F, Broackes-Carter F, Petschnigg J, Mills GB, Jurisicova A, Stagljar I, Maestro R, Jurisica I. In silico prediction of physical protein interactions and characterization of interactome orphans. Nat Methods 2014; 12:79-84. [PMID: 25402006 DOI: 10.1038/nmeth.3178] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 08/14/2014] [Indexed: 12/12/2022]
Abstract
Protein-protein interactions (PPIs) are useful for understanding signaling cascades, predicting protein function, associating proteins with disease and fathoming drug mechanism of action. Currently, only ∼ 10% of human PPIs may be known, and about one-third of human proteins have no known interactions. We introduce FpClass, a data mining-based method for proteome-wide PPI prediction. At an estimated false discovery rate of 60%, we predicted 250,498 PPIs among 10,531 human proteins; 10,647 PPIs involved 1,089 proteins without known interactions. We experimentally tested 233 high- and medium-confidence predictions and validated 137 interactions, including seven novel putative interactors of the tumor suppressor p53. Compared to previous PPI prediction methods, FpClass achieved better agreement with experimentally detected PPIs. We provide an online database of annotated PPI predictions (http://ophid.utoronto.ca/fpclass/) and the prediction software (http://www.cs.utoronto.ca/~juris/data/fpclass/).
Collapse
Affiliation(s)
- Max Kotlyar
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Chiara Pastrello
- 1] Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. [2] Centro Riferimento Oncologico, Istituto Nazionale Tumori, Aviano, Italy
| | - Flavia Pivetta
- Centro Riferimento Oncologico, Istituto Nazionale Tumori, Aviano, Italy
| | | | - Christian Cumbaa
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Han Li
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Taline Naranian
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Yun Niu
- 1] Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. [2] Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Zhiyong Ding
- Department of Systems Biology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fatemeh Vafaee
- 1] Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. [2] Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Fiona Broackes-Carter
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Julia Petschnigg
- Donnelly Centre, Departments of Molecular Genetics and Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Gordon B Mills
- Department of Systems Biology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrea Jurisicova
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Igor Stagljar
- Donnelly Centre, Departments of Molecular Genetics and Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Roberta Maestro
- Centro Riferimento Oncologico, Istituto Nazionale Tumori, Aviano, Italy
| | - Igor Jurisica
- 1] Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada. [2] Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. [3] Department of Computer Science, University of Toronto, Toronto, Ontario, Canada. [4] TECHNA Institute for the Advancement of Technology for Health, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Protein-protein interaction predictions using text mining methods. Methods 2014; 74:47-53. [PMID: 25448298 DOI: 10.1016/j.ymeth.2014.10.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 09/05/2014] [Accepted: 10/21/2014] [Indexed: 01/10/2023] Open
Abstract
It is beyond any doubt that proteins and their interactions play an essential role in most complex biological processes. The understanding of their function individually, but also in the form of protein complexes is of a great importance. Nowadays, despite the plethora of various high-throughput experimental approaches for detecting protein-protein interactions, many computational methods aiming to predict new interactions have appeared and gained interest. In this review, we focus on text-mining based computational methodologies, aiming to extract information for proteins and their interactions from public repositories such as literature and various biological databases. We discuss their strengths, their weaknesses and how they complement existing experimental techniques by simultaneously commenting on the biological databases which hold such information and the benchmark datasets that can be used for evaluating new tools.
Collapse
|
47
|
Wang J, Yang J, Mao S, Chai X, Hu Y, Hou X, Tang Y, Bi C, Li X. MitProNet: A knowledgebase and analysis platform of proteome, interactome and diseases for mammalian mitochondria. PLoS One 2014; 9:e111187. [PMID: 25347823 PMCID: PMC4210245 DOI: 10.1371/journal.pone.0111187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 09/26/2014] [Indexed: 12/18/2022] Open
Abstract
Mitochondrion plays a central role in diverse biological processes in most eukaryotes, and its dysfunctions are critically involved in a large number of diseases and the aging process. A systematic identification of mitochondrial proteomes and characterization of functional linkages among mitochondrial proteins are fundamental in understanding the mechanisms underlying biological functions and human diseases associated with mitochondria. Here we present a database MitProNet which provides a comprehensive knowledgebase for mitochondrial proteome, interactome and human diseases. First an inventory of mammalian mitochondrial proteins was compiled by widely collecting proteomic datasets, and the proteins were classified by machine learning to achieve a high-confidence list of mitochondrial proteins. The current version of MitProNet covers 1124 high-confidence proteins, and the remainders were further classified as middle- or low-confidence. An organelle-specific network of functional linkages among mitochondrial proteins was then generated by integrating genomic features encoded by a wide range of datasets including genomic context, gene expression profiles, protein-protein interactions, functional similarity and metabolic pathways. The functional-linkage network should be a valuable resource for the study of biological functions of mitochondrial proteins and human mitochondrial diseases. Furthermore, we utilized the network to predict candidate genes for mitochondrial diseases using prioritization algorithms. All proteins, functional linkages and disease candidate genes in MitProNet were annotated according to the information collected from their original sources including GO, GEO, OMIM, KEGG, MIPS, HPRD and so on. MitProNet features a user-friendly graphic visualization interface to present functional analysis of linkage networks. As an up-to-date database and analysis platform, MitProNet should be particularly helpful in comprehensive studies of complicated biological mechanisms underlying mitochondrial functions and human mitochondrial diseases. MitProNet is freely accessible at http://bio.scu.edu.cn:8085/MitProNet.
Collapse
Affiliation(s)
- Jiabin Wang
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Jian Yang
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Song Mao
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Xiaoqiang Chai
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Yuling Hu
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Xugang Hou
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Yiheng Tang
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Cheng Bi
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| | - Xiao Li
- College of Life Sciences, Sichuan University, Ministry of Education Key Laboratory for Bio-resource and Eco-environment, Sichuan Key Laboratory of Molecular Biology and Biotechnology, Chengdu, People’s Republic of China
| |
Collapse
|
48
|
Murali T, Pacifico S, Finley RL. Integrating the interactome and the transcriptome of Drosophila. BMC Bioinformatics 2014; 15:177. [PMID: 24913703 PMCID: PMC4229734 DOI: 10.1186/1471-2105-15-177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 05/28/2014] [Indexed: 12/29/2022] Open
Abstract
Background Networks of interacting genes and gene products mediate most cellular and developmental processes. High throughput screening methods combined with literature curation are identifying many of the protein-protein interactions (PPI) and protein-DNA interactions (PDI) that constitute these networks. Most of the detection methods, however, fail to identify the in vivo spatial or temporal context of the interactions. Thus, the interaction data are a composite of the individual networks that may operate in specific tissues or developmental stages. Genome-wide expression data may be useful for filtering interaction data to identify the subnetworks that operate in specific spatial or temporal contexts. Here we take advantage of the extensive interaction and expression data available for Drosophila to analyze how interaction networks may be unique to specific tissues and developmental stages. Results We ranked genes on a scale from ubiquitously expressed to tissue or stage specific and examined their interaction patterns. Interestingly, ubiquitously expressed genes have many more interactions among themselves than do non-ubiquitously expressed genes both in PPI and PDI networks. While the PDI network is enriched for interactions between tissue-specific transcription factors and their tissue-specific targets, a preponderance of the PDI interactions are between ubiquitous and non-ubiquitously expressed genes and proteins. In contrast to PDI, PPI networks are depleted for interactions among tissue- or stage- specific proteins, which instead interact primarily with widely expressed proteins. In light of these findings, we present an approach to filter interaction data based on gene expression levels normalized across tissues or developmental stages. We show that this filter (the percent maximum or pmax filter) can be used to identify subnetworks that function within individual tissues or developmental stages. Conclusions These observations suggest that protein networks are frequently organized into hubs of widely expressed proteins to which are attached various tissue- or stage-specific proteins. This is consistent with earlier analyses of human PPI data and suggests a similar organization of interaction networks across species. This organization implies that tissue or stage specific networks can be best identified from interactome data by using filters designed to include both ubiquitously expressed and specifically expressed genes and proteins.
Collapse
Affiliation(s)
| | | | - Russell L Finley
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| |
Collapse
|
49
|
Chen SK, Chung CA, Cheng YC, Huang CJ, Chen WY, Ruaan RC, Li C, Tsao CW, Hu WW, Chien CC. Toll-like receptor 6 and connective tissue growth factor are significantly upregulated in mitomycin-C-treated urothelial carcinoma cells under hydrostatic pressure stimulation. Genet Test Mol Biomarkers 2014; 18:410-6. [PMID: 24689870 DOI: 10.1089/gtmb.2013.0443] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Urothelial carcinoma (UC) is the most common histologic subtype of bladder cancer. The administration of mitomycin C (MMC) into the bladder after transurethral resection of the bladder tumor (TURBT) is a common treatment strategy for preventing recurrence after surgery. We previously applied hydrostatic pressure combined with MMC in UC cells and found that hydrostatic pressure synergistically enhanced MMC-induced UC cell apoptosis through the Fas/FasL pathways. To understand the alteration of gene expressions in UC cells caused by hydrostatic pressure and MMC, oligonucleotide microarray was used to explore all the differentially expressed genes. RESULTS After bioinformatics analysis and gene annotation, Toll-like receptor 6 (TLR6) and connective tissue growth factor (CTGF) showed significant upregulation among altered genes, and their gene and protein expressions with each treatment of UC cells were validated by quantitative real-time PCR and immunoblotting. CONCLUSION Under treatment with MMC and hydrostatic pressure, UC cells showed increasing apoptosis using extrinsic pathways through upregulation of TLR6 and CTGF.
Collapse
Affiliation(s)
- Shao-Kuan Chen
- 1 Department of Urology, Sijhih Cathay General Hospital , New Taipei City, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Dimitrakopoulos C, Vlantis AD, Theofilatos K, Likothanassis S, Mavroudi S. A New Framework for Bridging the Gap from Protein-Protein Interactions to Biological Process Interactions. IFIP ADVANCES IN INFORMATION AND COMMUNICATION TECHNOLOGY 2014:196-204. [DOI: 10.1007/978-3-662-44722-2_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|