1
|
Fernandez ME, Nazar FN, Moine LB, Jaime CE, Kembro JM, Correa SG. Network Analysis of Inflammatory Bowel Disease Research: Towards the Interactome. J Crohns Colitis 2022; 16:1651-1662. [PMID: 35439301 DOI: 10.1093/ecco-jcc/jjac059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Modern views accept that inflammatory bowel diseases [IBD] emerge from complex interactions among the multiple components of a biological network known as the 'IBD interactome'. These diverse components belong to different functional levels including cells, molecules, genes and biological processes. This diversity can make it difficult to integrate available empirical information from human patients into a collective view of aetiopathogenesis, a necessary step to understand the interactome. Herein, we quantitatively analyse how the representativeness of components involved in human IBD and their relationships ha ve changed over time. METHODS A bibliographic search in PubMed retrieved 25 971 abstracts of experimental studies on IBD in humans, published between 1990 and 2020. Abstracts were scanned automatically for 1218 IBD interactome components proposed in recent reviews. The resulting databases are freely available and were visualized as networks indicating the frequency at which different components are referenced together within each abstract. RESULTS As expected, over time there was an increase in components added to the IBD network and heightened connectivity within and across functional levels. However, certain components were consistently studied together, forming preserved motifs in the networks. These overrepresented and highly linked components reflect main 'hypotheses' in IBD research in humans. Interestingly, 82% of the components cited in reviews were absent or showed low frequency, suggesting that many aspects of the proposed IBD interactome still have weak experimental support in humans. CONCLUSIONS A reductionist and fragmented approach to the study of IBD has prevailed in previous decades, highlighting the importance of transitioning towards a more integrated interactome framework.
Collapse
Affiliation(s)
- M Emilia Fernandez
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - F Nicolas Nazar
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), Córdoba, Argentina.,Universidad Nacional de Córdoba, Facultad de Ciencias Exactas, Físicas y Naturales, Instituto de Ciencia y Tecnología de los Alimentos (ICTA), Córdoba, Argentina
| | - Luciana B Moine
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - Cristian E Jaime
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
| | - Jackelyn M Kembro
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), Córdoba, Argentina.,Universidad Nacional de Córdoba, Facultad de Ciencias Exactas, Físicas y Naturales, Instituto de Ciencia y Tecnología de los Alimentos (ICTA), Córdoba, Argentina.,Universidad Nacional de Córdoba, Facultad de Ciencias Exactas, Físicas y Naturales, Cátedra de Química Biológica, Córdoba, Argentina
| | - Silvia G Correa
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina.,Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Inmunología, Córdoba, Argentina
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Galano-Frutos JJ, García-Cebollada H, Sancho J. Molecular dynamics simulations for genetic interpretation in protein coding regions: where we are, where to go and when. Brief Bioinform 2019; 22:3-19. [PMID: 31813950 DOI: 10.1093/bib/bbz146] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/22/2019] [Accepted: 10/25/2019] [Indexed: 12/18/2022] Open
Abstract
The increasing ease with which massive genetic information can be obtained from patients or healthy individuals has stimulated the development of interpretive bioinformatics tools as aids in clinical practice. Most such tools analyze evolutionary information and simple physical-chemical properties to predict whether replacement of one amino acid residue with another will be tolerated or cause disease. Those approaches achieve up to 80-85% accuracy as binary classifiers (neutral/pathogenic). As such accuracy is insufficient for medical decision to be based on, and it does not appear to be increasing, more precise methods, such as full-atom molecular dynamics (MD) simulations in explicit solvent, are also discussed. Then, to describe the goal of interpreting human genetic variations at large scale through MD simulations, we restrictively refer to all possible protein variants carrying single-amino-acid substitutions arising from single-nucleotide variations as the human variome. We calculate its size and develop a simple model that allows calculating the simulation time needed to have a 0.99 probability of observing unfolding events of any unstable variant. The knowledge of that time enables performing a binary classification of the variants (stable-potentially neutral/unstable-pathogenic). Our model indicates that the human variome cannot be simulated with present computing capabilities. However, if they continue to increase as per Moore's law, it could be simulated (at 65°C) spending only 3 years in the task if we started in 2031. The simulation of individual protein variomes is achievable in short times starting at present. International coordination seems appropriate to embark upon massive MD simulations of protein variants.
Collapse
Affiliation(s)
- Juan J Galano-Frutos
- Protein Folding and Molecular Design (ProtMol)' group at BIFI, University of Zaragoza
| | | | - Javier Sancho
- Protein Folding and Molecular Design (ProtMol)' group at BIFI, University of Zaragoza
| |
Collapse
|
5
|
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]
|
6
|
Abstract
Current one drug–one target–one disease approaches in drug discovery have become increasingly inefficient. Network pharmacology defines disease mechanisms as networks best targeted by multiple, synergistic drugs. Using the high unmet medical need indication stroke, we here develop an integrative in silico approach based on a primary target, NADPH oxidase type 4, to identify a mechanistically related cotarget, NO synthase, for network pharmacology. Indeed, we validate both in vivo and in vitro, including humans, that both NOX4 and NOS inhibition is highly synergistic, leading to a significant reduction of infarct volume, direct neuroprotection, and blood–brain-barrier stabilization. This systems medicine approach provides a ground plan to decrease current failure in the field by being implemented in other complex indications. Drug discovery faces an efficacy crisis to which ineffective mainly single-target and symptom-based rather than mechanistic approaches have contributed. We here explore a mechanism-based disease definition for network pharmacology. Beginning with a primary causal target, we extend this to a second using guilt-by-association analysis. We then validate our prediction and explore synergy using both cellular in vitro and mouse in vivo models. As a disease model we chose ischemic stroke, one of the highest unmet medical need indications in medicine, and reactive oxygen species forming NADPH oxidase type 4 (Nox4) as a primary causal therapeutic target. For network analysis, we use classical protein–protein interactions but also metabolite-dependent interactions. Based on this protein–metabolite network, we conduct a gene ontology-based semantic similarity ranking to find suitable synergistic cotargets for network pharmacology. We identify the nitric oxide synthase (Nos1 to 3) gene family as the closest target to Nox4. Indeed, when combining a NOS and a NOX inhibitor at subthreshold concentrations, we observe pharmacological synergy as evidenced by reduced cell death, reduced infarct size, stabilized blood–brain barrier, reduced reoxygenation-induced leakage, and preserved neuromotor function, all in a supraadditive manner. Thus, protein–metabolite network analysis, for example guilt by association, can predict and pair synergistic mechanistic disease targets for systems medicine-driven network pharmacology. Such approaches may in the future reduce the risk of failure in single-target and symptom-based drug discovery and therapy.
Collapse
|
7
|
Peng X, Wang J, Peng W, Wu FX, Pan Y. Protein-protein interactions: detection, reliability assessment and applications. Brief Bioinform 2017; 18:798-819. [PMID: 27444371 DOI: 10.1093/bib/bbw066] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Indexed: 01/06/2023] Open
Abstract
Protein-protein interactions (PPIs) participate in all important biological processes in living organisms, such as catalyzing metabolic reactions, DNA replication, DNA transcription, responding to stimuli and transporting molecules from one location to another. To reveal the function mechanisms in cells, it is important to identify PPIs that take place in the living organism. A large number of PPIs have been discovered by high-throughput experiments and computational methods. However, false-positive PPIs have been introduced too. Therefore, to obtain reliable PPIs, many computational methods have been proposed. Generally, these methods can be classified into two categories. One category includes the methods that are designed to determine new reliable PPIs. The other one is designed to assess the reliability of existing PPIs and filter out the unreliable ones. In this article, we review the two kinds of methods for detecting reliable PPIs, and then focus on evaluating the performance of some of these typical methods. Later on, we also enumerate several PPI network-based applications with taking a reliability assessment of the PPI data into consideration. Finally, we will discuss the challenges for obtaining reliable PPIs and future directions of the construction of reliable PPI networks. Our research will provide readers some guidance for choosing appropriate methods and features for obtaining reliable PPIs.
Collapse
|
8
|
Northey TC, Barešić A, Martin ACR. IntPred: a structure-based predictor of protein-protein interaction sites. Bioinformatics 2017; 34:223-229. [PMID: 28968673 PMCID: PMC5860208 DOI: 10.1093/bioinformatics/btx585] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 08/21/2017] [Accepted: 09/15/2017] [Indexed: 11/17/2022] Open
Abstract
Motivation Protein–protein interactions are vital for protein function with the average protein having between three and ten interacting partners. Knowledge of precise protein–protein interfaces comes from crystal structures deposited in the Protein Data Bank (PDB), but only 50% of structures in the PDB are complexes. There is therefore a need to predict protein–protein interfaces in silico and various methods for this purpose. Here we explore the use of a predictor based on structural features and which exploits random forest machine learning, comparing its performance with a number of popular established methods. Results On an independent test set of obligate and transient complexes, our IntPred predictor performs well (MCC = 0.370, ACC = 0.811, SPEC = 0.916, SENS = 0.411) and compares favourably with other methods. Overall, IntPred ranks second of six methods tested with SPPIDER having slightly better overall performance (MCC = 0.410, ACC = 0.759, SPEC = 0.783, SENS = 0.676), but considerably worse specificity than IntPred. As with SPPIDER, using an independent test set of obligate complexes enhanced performance (MCC = 0.381) while performance is somewhat reduced on a dataset of transient complexes (MCC = 0.303). The trade-off between sensitivity and specificity compared with SPPIDER suggests that the choice of the appropriate tool is application-dependent. Availability and implementation IntPred is implemented in Perl and may be downloaded for local use or run via a web server at www.bioinf.org.uk/intpred/. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Thomas C Northey
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Anja Barešić
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Andrew C R Martin
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| |
Collapse
|
9
|
Nicod C, Banaei-Esfahani A, Collins BC. Elucidation of host-pathogen protein-protein interactions to uncover mechanisms of host cell rewiring. Curr Opin Microbiol 2017; 39:7-15. [PMID: 28806587 DOI: 10.1016/j.mib.2017.07.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/27/2017] [Indexed: 01/08/2023]
Abstract
Infectious diseases are the result of molecular cross-talks between hosts and their pathogens. These cross-talks are in part mediated by host-pathogen protein-protein interactions (HP-PPI). HP-PPI play crucial roles in infections, as they may tilt the balance either in favor of the pathogens' spread or their clearance. The identification of host proteins targeted by viral or bacterial pathogenic proteins necessary for the infection can provide insights into their underlying molecular mechanisms of pathogenicity, and potentially even single out pharmacological intervention targets. Here, we review the available methods to study HP-PPI, with a focus on recent mass spectrometry based methods to decipher bacterial-human infectious diseases and examine their relevance in uncovering host cell rewiring by pathogens.
Collapse
Affiliation(s)
- Charlotte Nicod
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland; PhD Program in Systems Biology, Life Science Zurich Graduate School, University of Zurich and ETH Zurich, CH-8093 Zurich, Switzerland
| | - Amir Banaei-Esfahani
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland; PhD Program in Systems Biology, Life Science Zurich Graduate School, University of Zurich and ETH Zurich, CH-8093 Zurich, Switzerland
| | - Ben C Collins
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
10
|
Alanis-Lobato G, Andrade-Navarro MA, Schaefer MH. HIPPIE v2.0: enhancing meaningfulness and reliability of protein-protein interaction networks. Nucleic Acids Res 2016; 45:D408-D414. [PMID: 27794551 PMCID: PMC5210659 DOI: 10.1093/nar/gkw985] [Citation(s) in RCA: 319] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/28/2016] [Accepted: 10/14/2016] [Indexed: 01/01/2023] Open
Abstract
The increasing number of experimentally detected interactions between proteins makes it difficult for researchers to extract the interactions relevant for specific biological processes or diseases. This makes it necessary to accompany the large-scale detection of protein–protein interactions (PPIs) with strategies and tools to generate meaningful PPI subnetworks. To this end, we generated the Human Integrated Protein–Protein Interaction rEference or HIPPIE (http://cbdm.uni-mainz.de/hippie/). HIPPIE is a one-stop resource for the generation and interpretation of PPI networks relevant to a specific research question. We provide means to generate highly reliable, context-specific PPI networks and to make sense out of them. We just released the second major update of HIPPIE, implementing various new features. HIPPIE grew substantially over the last years and now contains more than 270 000 confidence scored and annotated PPIs. We integrated different types of experimental information for the confidence scoring and the construction of context-specific networks. We implemented basic graph algorithms that highlight important proteins and interactions. HIPPIE's graphical interface implements several ways for wet lab and computational scientists alike to access the PPI data.
Collapse
Affiliation(s)
- Gregorio Alanis-Lobato
- Faculty of Biology, Johannes Gutenberg Universität, Mainz, Germany
- Institute of Molecular Biology, Mainz, Germany
| | - Miguel A Andrade-Navarro
- Faculty of Biology, Johannes Gutenberg Universität, Mainz, Germany
- Institute of Molecular Biology, Mainz, Germany
| | - Martin H Schaefer
- EMBL/CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
11
|
Zhou L, Li Q, Wang J, Huang C, Nice EC. Oncoproteomics: Trials and tribulations. Proteomics Clin Appl 2015; 10:516-31. [PMID: 26518147 DOI: 10.1002/prca.201500081] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/19/2015] [Accepted: 10/27/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu P. R. China
- Department of Neurology; The Affiliated Hospital of Hainan Medical College; Haikou Hainan P. R. China
| | - Qifu Li
- Department of Neurology; The Affiliated Hospital of Hainan Medical College; Haikou Hainan P. R. China
| | - Jiandong Wang
- Department of Biomedical; Chengdu Medical College; Chengdu Sichuan Province P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu P. R. China
| | - Edouard C. Nice
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu P. R. China
- Department of Biochemistry and Molecular Biology; Monash University; Clayton Australia
| |
Collapse
|
12
|
Silva JV, Freitas MJ, Felgueiras J, Fardilha M. The power of the yeast two-hybrid system in the identification of novel drug targets: building and modulating PPP1 interactomes. Expert Rev Proteomics 2015; 12:147-58. [PMID: 25795147 DOI: 10.1586/14789450.2015.1024226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Since the description of the yeast two-hybrid (Y2H) method, it has become more and more evident that it is the most commonly used method to identify protein-protein interactions (PPIs). The improvements in the original Y2H methodology in parallel with the idea that PPIs are promising drug targets, offer an excellent opportunity to apply the principles of this molecular biology technique to the pharmaceutical field. Additionally, the theoretical developments in the networks field make PPI networks very useful frameworks that facilitate many discoveries in biomedicine. This review highlights the relevance of Y2H in the determination of PPIs, specifically phosphoprotein phosphatase 1 interactions, and its possible outcomes in pharmaceutical research.
Collapse
Affiliation(s)
- Joana Vieira Silva
- Signal Transduction Laboratory, Institute for Research in Biomedicine - iBiMED, Health Sciences Program, University of Aveiro, Aveiro, Portugal
| | | | | | | |
Collapse
|
13
|
Jafari M, Mirzaie M, Sadeghi M. Interlog protein network: an evolutionary benchmark of protein interaction networks for the evaluation of clustering algorithms. BMC Bioinformatics 2015; 16:319. [PMID: 26437714 PMCID: PMC4595048 DOI: 10.1186/s12859-015-0755-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 09/29/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the field of network science, exploring principal and crucial modules or communities is critical in the deduction of relationships and organization of complex networks. This approach expands an arena, and thus allows further study of biological functions in the field of network biology. As the clustering algorithms that are currently employed in finding modules have innate uncertainties, external and internal validations are necessary. METHODS Sequence and network structure alignment, has been used to define the Interlog Protein Network (IPN). This network is an evolutionarily conserved network with communal nodes and less false-positive links. In the current study, the IPN is employed as an evolution-based benchmark in the validation of the module finding methods. The clustering results of five algorithms; Markov Clustering (MCL), Restricted Neighborhood Search Clustering (RNSC), Cartographic Representation (CR), Laplacian Dynamics (LD) and Genetic Algorithm; to find communities in Protein-Protein Interaction networks (GAPPI) are assessed by IPN in four distinct Protein-Protein Interaction Networks (PPINs). RESULTS The MCL shows a more accurate algorithm based on this evolutionary benchmarking approach. Also, the biological relevance of proteins in the IPN modules generated by MCL is compatible with biological standard databases such as Gene Ontology, KEGG and Reactome. CONCLUSION In this study, the IPN shows its potential for validation of clustering algorithms due to its biological logic and straightforward implementation.
Collapse
Affiliation(s)
- Mohieddin Jafari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, 69 Pasteur St, PO Box 13164, Tehran, Iran.
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Shahid Lavasani St, PO Box 19395-5746, Tehran, Iran.
| | - Mehdi Mirzaie
- Department of Computational Biology, Faculty of High Technologies, Tarbiat Modares University, Jalal Ale Ahmad Highway, PO Box 14115-111, Tehran, Iran.
| | - Mehdi Sadeghi
- National Institute of Genetic Engineering and Biotechnology (NIGEB), Pajoohesh Blvd, 17 Km Tehran-Karaj Highway, PO Box 161-14965, Tehran, Iran.
| |
Collapse
|
14
|
Schaefer MH, Serrano L, Andrade-Navarro MA. Correcting for the study bias associated with protein-protein interaction measurements reveals differences between protein degree distributions from different cancer types. Front Genet 2015; 6:260. [PMID: 26300911 PMCID: PMC4523822 DOI: 10.3389/fgene.2015.00260] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/21/2015] [Indexed: 01/17/2023] Open
Abstract
Protein-protein interaction (PPI) networks are associated with multiple types of biases partly rooted in technical limitations of the experimental techniques. Another source of bias are the different frequencies with which proteins have been studied for interaction partners. It is generally believed that proteins with a large number of interaction partners tend to be essential, evolutionarily conserved, and involved in disease. It has been repeatedly reported that proteins driving tumor formation have a higher number of PPI partners. However, it has been noticed before that the degree distribution of PPI networks is biased toward disease proteins, which tend to have been studied more often than non-disease proteins. At the same time, for many poorly characterized proteins no interactions have been reported yet. It is unclear to which extent this study bias affects the observation that cancer proteins tend to have more PPI partners. Here, we show that the degree of a protein is a function of the number of times it has been screened for interaction partners. We present a randomization-based method that controls for this bias to decide whether a group of proteins is associated with significantly more PPI partners than the proteomic background. We apply our method to cancer proteins and observe, in contrast to previous studies, no conclusive evidence for a significantly higher degree distribution associated with cancer proteins as compared to non-cancer proteins when we compare them to proteins that have been equally often studied as bait proteins. Comparing proteins from different tumor types, a more complex picture emerges in which proteins of certain cancer classes have significantly more interaction partners while others are associated with a smaller degree. For example, proteins of several hematological cancers tend to be associated with a higher number of interaction partners as expected by chance. Solid tumors, in contrast, are usually associated with a degree distribution similar to those of equally often studied random protein sets. We discuss the biological implications of these findings. Our work shows that accounting for biases in the PPI network is possible and increases the value of PPI data.
Collapse
Affiliation(s)
- Martin H Schaefer
- Systems Biology Research Unit, Centre for Genomic Regulation - European Molecular Biology Laboratory, Barcelona Spain ; Universitat Pompeu Fabra, Barcelona Spain
| | - Luis Serrano
- Systems Biology Research Unit, Centre for Genomic Regulation - European Molecular Biology Laboratory, Barcelona Spain ; Universitat Pompeu Fabra, Barcelona Spain ; Institució Catalana de Recerca i Estudis Avançats, Barcelona Spain
| | - Miguel A Andrade-Navarro
- Faculty of Biology, Johannes Gutenberg University of Mainz Mainz, Germany ; Institute of Molecular Biology, Mainz Germany
| |
Collapse
|
15
|
Tang H, Zhong F, Liu W, He F, Xie H. PathPPI: an integrated dataset of human pathways and protein-protein interactions. SCIENCE CHINA-LIFE SCIENCES 2015; 58:579-89. [PMID: 25591449 DOI: 10.1007/s11427-014-4766-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 07/20/2014] [Indexed: 12/23/2022]
Abstract
Integration of pathway and protein-protein interaction (PPI) data can provide more information that could lead to new biological insights. PPIs are usually represented by a simple binary model, whereas pathways are represented by more complicated models. We developed a series of rules for transforming protein interactions from pathway to binary model, and the protein interactions from seven pathway databases, including PID, BioCarta, Reactome, NetPath, INOH, SPIKE and KEGG, were transformed based on these rules. These pathway-derived binary protein interactions were integrated with PPIs from other five PPI databases including HPRD, IntAct, BioGRID, MINT and DIP, to develop integrated dataset (named PathPPI). More detailed interaction type and modification information on protein interactions can be preserved in PathPPI than other existing datasets. Comparison analysis results indicate that most of the interaction overlaps values (O AB) among these pathway databases were less than 5%, and these databases must be used conjunctively. The PathPPI data was provided at http://proteomeview.hupo.org.cn/PathPPI/PathPPI.html.
Collapse
Affiliation(s)
- HaiLin Tang
- College of Mechanical & Electronic Engineering and Automatization, National University of Defense Technology, Changsha, 410073, China
| | | | | | | | | |
Collapse
|
16
|
Luo X, You Z, Zhou M, Li S, Leung H, Xia Y, Zhu Q. A highly efficient approach to protein interactome mapping based on collaborative filtering framework. Sci Rep 2015; 5:7702. [PMID: 25572661 PMCID: PMC4287731 DOI: 10.1038/srep07702] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/08/2014] [Indexed: 12/17/2022] Open
Abstract
The comprehensive mapping of protein-protein interactions (PPIs) is highly desired for one to gain deep insights into both fundamental cell biology processes and the pathology of diseases. Finely-set small-scale experiments are not only very expensive but also inefficient to identify numerous interactomes despite their high accuracy. High-throughput screening techniques enable efficient identification of PPIs; yet the desire to further extract useful knowledge from these data leads to the problem of binary interactome mapping. Network topology-based approaches prove to be highly efficient in addressing this problem; however, their performance deteriorates significantly on sparse putative PPI networks. Motivated by the success of collaborative filtering (CF)-based approaches to the problem of personalized-recommendation on large, sparse rating matrices, this work aims at implementing a highly efficient CF-based approach to binary interactome mapping. To achieve this, we first propose a CF framework for it. Under this framework, we model the given data into an interactome weight matrix, where the feature-vectors of involved proteins are extracted. With them, we design the rescaled cosine coefficient to model the inter-neighborhood similarity among involved proteins, for taking the mapping process. Experimental results on three large, sparse datasets demonstrate that the proposed approach outperforms several sophisticated topology-based approaches significantly.
Collapse
Affiliation(s)
- Xin Luo
- X. Luo, Y. Xia and Q. Zhu are with the College of Computer Science, Chongqing University, Chongqing, 400044 China
- X. Luo, Z. You, S. Li and H. Leung are with the Department of Computing, Hong Kong Polytechnic University, Hong Kong, HK 999077, China
| | - Zhuhong You
- X. Luo, Z. You, S. Li and H. Leung are with the Department of Computing, Hong Kong Polytechnic University, Hong Kong, HK 999077, China
| | - Mengchu Zhou
- M. Zhou is with the Department of Electrical and Computer Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Shuai Li
- X. Luo, Z. You, S. Li and H. Leung are with the Department of Computing, Hong Kong Polytechnic University, Hong Kong, HK 999077, China
| | - Hareton Leung
- X. Luo, Z. You, S. Li and H. Leung are with the Department of Computing, Hong Kong Polytechnic University, Hong Kong, HK 999077, China
| | - Yunni Xia
- X. Luo, Y. Xia and Q. Zhu are with the College of Computer Science, Chongqing University, Chongqing, 400044 China
| | - Qingsheng Zhu
- X. Luo, Y. Xia and Q. Zhu are with the College of Computer Science, Chongqing University, Chongqing, 400044 China
| |
Collapse
|
17
|
Pinto JP, Machado RSR, Xavier JM, Futschik ME. Targeting molecular networks for drug research. Front Genet 2014; 5:160. [PMID: 24926314 PMCID: PMC4045242 DOI: 10.3389/fgene.2014.00160] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 05/14/2014] [Indexed: 01/18/2023] Open
Abstract
The study of molecular networks has recently moved into the limelight of biomedical research. While it has certainly provided us with plenty of new insights into cellular mechanisms, the challenge now is how to modify or even restructure these networks. This is especially true for human diseases, which can be regarded as manifestations of distorted states of molecular networks. Of the possible interventions for altering networks, the use of drugs is presently the most feasible. In this mini-review, we present and discuss some exemplary approaches of how analysis of molecular interaction networks can contribute to pharmacology (e.g., by identifying new drug targets or prediction of drug side effects), as well as list pointers to relevant resources and software to guide future research. We also outline recent progress in the use of drugs for in vitro reprogramming of cells, which constitutes an example par excellence for altering molecular interaction networks with drugs.
Collapse
Affiliation(s)
- José P Pinto
- SysBioLab, Centre for Molecular and Structural Biomedicine, Universidade do Algarve Faro, Portugal
| | - Rui S R Machado
- SysBioLab, Centre for Molecular and Structural Biomedicine, Universidade do Algarve Faro, Portugal
| | - Joana M Xavier
- SysBioLab, Centre for Molecular and Structural Biomedicine, Universidade do Algarve Faro, Portugal
| | - Matthias E Futschik
- SysBioLab, Centre for Molecular and Structural Biomedicine, Universidade do Algarve Faro, Portugal ; Centre of Marine Sciences, Universidade do Algarve Faro, Portugal
| |
Collapse
|
18
|
Mina M, Guzzi PH. Improving the Robustness of Local Network Alignment: Design and Extensive Assessment of a Markov Clustering-Based Approach. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2014; 11:561-572. [PMID: 26356023 DOI: 10.1109/tcbb.2014.2318707] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The analysis of protein behavior at the network level had been applied to elucidate the mechanisms of protein interaction that are similar in different species. Published network alignment algorithms proved to be able to recapitulate known conserved modules and protein complexes, and infer new conserved interactions confirmed by wet lab experiments. In the meantime, however, a plethora of continuously evolving protein-protein interaction (PPI) data sets have been developed, each featuring different levels of completeness and reliability. For instance, algorithms performance may vary significantly when changing the data set used in their assessment. Moreover, existing papers did not deeply investigate the robustness of alignment algorithms. For instance, some algorithms performances vary significantly when changing the data set used in their assessment. In this work, we design an extensive assessment of current algorithms discussing the robustness of the results on the basis of input networks. We also present AlignMCL, a local network alignment algorithm based on an improved model of alignment graph and Markov Clustering. AlignMCL performs better than other state-of-the-art local alignment algorithms over different updated data sets. In addition, AlignMCL features high levels of robustness, producing similar results regardless the selected data set.
Collapse
|
19
|
Hernandez-Prieto MA, Kalathur RK, Futschik ME. Molecular Networks – Representation and Analysis. SPRINGER HANDBOOK OF BIO-/NEUROINFORMATICS 2014:399-418. [DOI: 10.1007/978-3-642-30574-0_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Ngounou Wetie AG, Sokolowska I, Woods AG, Roy U, Deinhardt K, Darie CC. Protein-protein interactions: switch from classical methods to proteomics and bioinformatics-based approaches. Cell Mol Life Sci 2014; 71:205-28. [PMID: 23579629 PMCID: PMC11113707 DOI: 10.1007/s00018-013-1333-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 03/25/2013] [Accepted: 03/26/2013] [Indexed: 11/28/2022]
Abstract
Following the sequencing of the human genome and many other organisms, research on protein-coding genes and their functions (functional genomics) has intensified. Subsequently, with the observation that proteins are indeed the molecular effectors of most cellular processes, the discipline of proteomics was born. Clearly, proteins do not function as single entities but rather as a dynamic network of team players that have to communicate. Though genetic (yeast two-hybrid Y2H) and biochemical methods (co-immunoprecipitation Co-IP, affinity purification AP) were the methods of choice at the beginning of the study of protein-protein interactions (PPI), in more recent years there has been a shift towards proteomics-based methods and bioinformatics-based approaches. In this review, we first describe in depth PPIs and we make a strong case as to why unraveling the interactome is the next challenge in the field of proteomics. Furthermore, classical methods of investigation of PPIs and structure-based bioinformatics approaches are presented. The greatest emphasis is placed on proteomic methods, especially native techniques that were recently developed and that have been shown to be reliable. Finally, we point out the limitations of these methods and the need to set up a standard for the validation of PPI experiments.
Collapse
Affiliation(s)
- Armand G. Ngounou Wetie
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Izabela Sokolowska
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Alisa G. Woods
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Urmi Roy
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| | - Katrin Deinhardt
- Centre for Biological Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ UK
- Institute for Life Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ UK
| | - Costel C. Darie
- Department of Chemistry and Biomolecular Science, Biochemistry and Proteomics Group, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699-5810 USA
| |
Collapse
|
21
|
Schleker S, Ananthasubramanian S, Klein‐Seetharaman J, Ganapathiraju MK. Prediction of Intra‐ and Interspecies Protein–Protein Interactions Facilitating Systems Biology Studies. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2013:21-53. [DOI: 10.1002/9783527648207.ch2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Tang H, Zhong F, Xie H. A quick guide to biomolecular network studies: construction, analysis, applications, and resources. Biochem Biophys Res Commun 2012; 424:7-11. [PMID: 22732414 DOI: 10.1016/j.bbrc.2012.06.085] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 06/18/2012] [Indexed: 10/28/2022]
Abstract
Over the past decade, a rapid increase in network data including signaling, transcription regulation, metabolic reaction, protein-protein interaction and genetic interaction has been observed. Many biology issues have been investigated by analyzing these diverse networks, providing new insights into biology. Networks also play an important role in disease studies including disease gene screening and clinical diagnosis. Large amounts of databases and software have been developed to facilitate the storage, exchange, integration, and analysis of network data and network analysis is becoming a routine procedure for biologists to infer biological information. In this review, several main aspects of network studies are discussed, including network construction, analysis, application, and resources.
Collapse
Affiliation(s)
- Hailin Tang
- College of Mechanical & Electronic Engineering and Automatization, National University of Defense Technology, Changsha 410073, China
| | | | | |
Collapse
|
23
|
Kirouac DC, Saez-Rodriguez J, Swantek J, Burke JM, Lauffenburger DA, Sorger PK. Creating and analyzing pathway and protein interaction compendia for modelling signal transduction networks. BMC SYSTEMS BIOLOGY 2012; 6:29. [PMID: 22548703 PMCID: PMC3436686 DOI: 10.1186/1752-0509-6-29] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 04/11/2012] [Indexed: 11/11/2022]
Abstract
Background Understanding the information-processing capabilities of signal transduction networks, how those networks are disrupted in disease, and rationally designing therapies to manipulate diseased states require systematic and accurate reconstruction of network topology. Data on networks central to human physiology, such as the inflammatory signalling networks analyzed here, are found in a multiplicity of on-line resources of pathway and interactome databases (Cancer CellMap, GeneGo, KEGG, NCI-Pathway Interactome Database (NCI-PID), PANTHER, Reactome, I2D, and STRING). We sought to determine whether these databases contain overlapping information and whether they can be used to construct high reliability prior knowledge networks for subsequent modeling of experimental data. Results We have assembled an ensemble network from multiple on-line sources representing a significant portion of all machine-readable and reconcilable human knowledge on proteins and protein interactions involved in inflammation. This ensemble network has many features expected of complex signalling networks assembled from high-throughput data: a power law distribution of both node degree and edge annotations, and topological features of a “bow tie” architecture in which diverse pathways converge on a highly conserved set of enzymatic cascades focused around PI3K/AKT, MAPK/ERK, JAK/STAT, NFκB, and apoptotic signaling. Individual pathways exhibit “fuzzy” modularity that is statistically significant but still involving a majority of “cross-talk” interactions. However, we find that the most widely used pathway databases are highly inconsistent with respect to the actual constituents and interactions in this network. Using a set of growth factor signalling networks as examples (epidermal growth factor, transforming growth factor-beta, tumor necrosis factor, and wingless), we find a multiplicity of network topologies in which receptors couple to downstream components through myriad alternate paths. Many of these paths are inconsistent with well-established mechanistic features of signalling networks, such as a requirement for a transmembrane receptor in sensing extracellular ligands. Conclusions Wide inconsistencies among interaction databases, pathway annotations, and the numbers and identities of nodes associated with a given pathway pose a major challenge for deriving causal and mechanistic insight from network graphs. We speculate that these inconsistencies are at least partially attributable to cell, and context-specificity of cellular signal transduction, which is largely unaccounted for in available databases, but the absence of standardized vocabularies is an additional confounding factor. As a result of discrepant annotations, it is very difficult to identify biologically meaningful pathways from interactome networks a priori. However, by incorporating prior knowledge, it is possible to successively build out network complexity with high confidence from a simple linear signal transduction scaffold. Such reduced complexity networks appear suitable for use in mechanistic models while being richer and better justified than the simple linear pathways usually depicted in diagrams of signal transduction.
Collapse
Affiliation(s)
- Daniel C Kirouac
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
24
|
A benchmarked protein microarray-based platform for the identification of novel low-affinity extracellular protein interactions. Anal Biochem 2012; 424:45-53. [PMID: 22342946 PMCID: PMC3325482 DOI: 10.1016/j.ab.2012.01.034] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/04/2011] [Accepted: 01/13/2012] [Indexed: 12/13/2022]
Abstract
Low-affinity extracellular protein interactions are critical for cellular recognition processes, but existing methods to detect them are limited in scale, making genome-wide interaction screens technically challenging. To address this, we report here the miniaturization of the AVEXIS (avidity-based extracellular interaction screen) assay by using protein microarray technology. To achieve this, we have developed protein tags and sample preparation methods that enable the parallel purification of hundreds of recombinant proteins expressed in mammalian cells. We benchmarked the protein microarray-based assay against a set of known quantified receptor–ligand pairs and show that it is sensitive enough to detect even very weak interactions that are typical of this class of interactions. The increase in scale enables interaction screening against a dilution series of immobilized proteins on the microarray enabling the observation of saturation binding behaviors to show interaction specificity and also the estimation of interaction affinities directly from the primary screen. These methodological improvements now permit screening for novel extracellular receptor–ligand interactions on a genome-wide scale.
Collapse
|
25
|
Nazri A, Lio P. Investigating meta-approaches for reconstructing gene networks in a mammalian cellular context. PLoS One 2012; 7:e28713. [PMID: 22253694 PMCID: PMC3253778 DOI: 10.1371/journal.pone.0028713] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 11/14/2011] [Indexed: 11/18/2022] Open
Abstract
The output of state-of-the-art reverse-engineering methods for biological networks is often based on the fitting of a mathematical model to the data. Typically, different datasets do not give single consistent network predictions but rather an ensemble of inconsistent networks inferred under the same reverse-engineering method that are only consistent with the specific experimentally measured data. Here, we focus on an alternative approach for combining the information contained within such an ensemble of inconsistent gene networks called meta-analysis, to make more accurate predictions and to estimate the reliability of these predictions. We review two existing meta-analysis approaches; the Fisher transformation combined coefficient test (FTCCT) and Fisher's inverse combined probability test (FICPT); and compare their performance with five well-known methods, ARACNe, Context Likelihood or Relatedness network (CLR), Maximum Relevance Minimum Redundancy (MRNET), Relevance Network (RN) and Bayesian Network (BN). We conducted in-depth numerical ensemble simulations and demonstrated for biological expression data that the meta-analysis approaches consistently outperformed the best gene regulatory network inference (GRNI) methods in the literature. Furthermore, the meta-analysis approaches have a low computational complexity. We conclude that the meta-analysis approaches are a powerful tool for integrating different datasets to give more accurate and reliable predictions for biological networks.
Collapse
Affiliation(s)
- Azree Nazri
- Department of Computer Science, Faculty of Computer Science & Information Technology, University Putra Malaysia, Malaysia, Selangor, Malaysia.
| | | |
Collapse
|
26
|
Chaurasia G, Futschik M. The integration and annotation of the human interactome in the UniHI Database. Methods Mol Biol 2012; 812:175-188. [PMID: 22218860 DOI: 10.1007/978-1-61779-455-1_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
In recent years, remarkable progress has been made toward the systematic charting of human protein interactions. The utilization of the generated interaction data remained however challenging for biomedical researchers due to lack of integration of currently available resources. To facilitate the direct access and analysis of the human interactome, we have developed the Unified Human Interactome (UniHI) database. It provides researchers with a user-friendly Web-interface and integrates interaction data from 12 major resources in its latest version, establishing one of the largest catalogs for human PPIs worldwide. At present, UniHI houses over 250,000 distinct interactions between 22,300 unique proteins and is publically available at http://www.unihi.org.
Collapse
|
27
|
Hallinan JS, James K, Wipat A. Network approaches to the functional analysis of microbial proteins. Adv Microb Physiol 2011; 59:101-33. [PMID: 22114841 DOI: 10.1016/b978-0-12-387661-4.00005-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Large amounts of detailed biological data have been generated over the past few decades. Much of these data is freely available in over 1000 online databases; an enticing, but frustrating resource for microbiologists interested in a systems-level view of the structure and function of microbial cells. The frustration engendered by the need to trawl manually through hundreds of databases in order to accumulate information about a gene, protein, pathway, or organism of interest can be alleviated by the use of computational data integration to generated network views of the system of interest. Biological networks can be constructed from a single type of data, such as protein-protein binding information, or from data generated by multiple experimental approaches. In an integrated network, nodes usually represent genes or gene products, while edges represent some form of interaction between the nodes. Edges between nodes may be weighted to represent the probability that the edge exists in vivo. Networks may also be enriched with ontological annotations, facilitating both visual browsing and computational analysis via web service interfaces. In this review, we describe the construction, analysis of both single-data source and integrated networks, and their application to the inference of protein function in microbes.
Collapse
Affiliation(s)
- J S Hallinan
- School of Computing Science, Newcastle University, Newcastle, UK
| | | | | |
Collapse
|
28
|
Lindén RO, Eronen VP, Aittokallio T. Quantitative maps of genetic interactions in yeast - comparative evaluation and integrative analysis. BMC SYSTEMS BIOLOGY 2011; 5:45. [PMID: 21435228 PMCID: PMC3079637 DOI: 10.1186/1752-0509-5-45] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 03/24/2011] [Indexed: 01/08/2023]
Abstract
Background High-throughput genetic screening approaches have enabled systematic means to study how interactions among gene mutations contribute to quantitative fitness phenotypes, with the aim of providing insights into the functional wiring diagrams of genetic interaction networks on a global scale. However, it is poorly known how well these quantitative interaction measurements agree across the screening approaches, which hinders their integrated use toward improving the coverage and quality of the genetic interaction maps in yeast and other organisms. Results Using large-scale data matrices from epistatic miniarray profiling (E-MAP), genetic interaction mapping (GIM), and synthetic genetic array (SGA) approaches, we carried out here a systematic comparative evaluation among these quantitative maps of genetic interactions in yeast. The relatively low association between the original interaction measurements or their customized scores could be improved using a matrix-based modelling framework, which enables the use of single- and double-mutant fitness estimates and measurements, respectively, when scoring genetic interactions. Toward an integrative analysis, we show how the detections from the different screening approaches can be combined to suggest novel positive and negative interactions which are complementary to those obtained using any single screening approach alone. The matrix approximation procedure has been made available to support the design and analysis of the future screening studies. Conclusions We have shown here that even if the correlation between the currently available quantitative genetic interaction maps in yeast is relatively low, their comparability can be improved by means of our computational matrix approximation procedure, which will enable integrative analysis and detection of a wider spectrum of genetic interactions using data from the complementary screening approaches.
Collapse
Affiliation(s)
- Rolf O Lindén
- Biomathematics Research Group, Department of Mathematics, University of Turku, Turku, Finland
| | | | | |
Collapse
|
29
|
Dube DH, Li B, Greenblatt EJ, Nimer S, Raymond AK, Kohler JJ. A two-hybrid assay to study protein interactions within the secretory pathway. PLoS One 2010; 5:e15648. [PMID: 21209940 PMCID: PMC3011011 DOI: 10.1371/journal.pone.0015648] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 11/18/2010] [Indexed: 11/18/2022] Open
Abstract
Interactions of transcriptional activators are difficult to study using transcription-based two-hybrid assays due to potent activation resulting in false positives. Here we report the development of the Golgi two-hybrid (G2H), a method that interrogates protein interactions within the Golgi, where transcriptional activators can be assayed with negligible background. The G2H relies on cell surface glycosylation to report extracellularly on protein-protein interactions occurring within the secretory pathway. In the G2H, protein pairs are fused to modular domains of the reporter glycosyltransferase, Och1p, and proper cell wall formation due to Och1p activity is observed only when a pair of proteins interacts. Cells containing interacting protein pairs are identified by selectable phenotypes associated with Och1p activity and proper cell wall formation: cells that have interacting proteins grow under selective conditions and display weak wheat germ agglutinin (WGA) binding by flow cytometry, whereas cells that lack interacting proteins display stunted growth and strong WGA binding. Using this assay, we detected the interaction between transcription factor MyoD and its binding partner Id2. Interfering mutations along the MyoD:Id2 interaction interface ablated signal in the G2H assay. Furthermore, we used the G2H to detect interactions of the activation domain of Gal4p with a variety of binding partners. Finally, selective conditions were used to enrich for cells encoding interacting partners. The G2H detects protein-protein interactions that cannot be identified via traditional two-hybrid methods and should be broadly useful for probing previously inaccessible subsets of the interactome, including transcriptional activators and proteins that traffic through the secretory pathway.
Collapse
Affiliation(s)
- Danielle H. Dube
- Department of Chemistry and Biochemistry, Bowdoin College, Brunswick, Maine, United States of America
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Bin Li
- Division of Translational Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ethan J. Greenblatt
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- Biophysics Program, Stanford University, Stanford, California, United States of America
| | - Sadeieh Nimer
- Division of Translational Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Amanda K. Raymond
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Jennifer J. Kohler
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- Division of Translational Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
30
|
Jaeger S, Sers CT, Leser U. Combining modularity, conservation, and interactions of proteins significantly increases precision and coverage of protein function prediction. BMC Genomics 2010; 11:717. [PMID: 21171995 PMCID: PMC3017542 DOI: 10.1186/1471-2164-11-717] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 12/20/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While the number of newly sequenced genomes and genes is constantly increasing, elucidation of their function still is a laborious and time-consuming task. This has led to the development of a wide range of methods for predicting protein functions in silico. We report on a new method that predicts function based on a combination of information about protein interactions, orthology, and the conservation of protein networks in different species. RESULTS We show that aggregation of these independent sources of evidence leads to a drastic increase in number and quality of predictions when compared to baselines and other methods reported in the literature. For instance, our method generates more than 12,000 novel protein functions for human with an estimated precision of ~76%, among which are 7,500 new functional annotations for 1,973 human proteins that previously had zero or only one function annotated. We also verified our predictions on a set of genes that play an important role in colorectal cancer (MLH1, PMS2, EPHB4 ) and could confirm more than 73% of them based on evidence in the literature. CONCLUSIONS The combination of different methods into a single, comprehensive prediction method infers thousands of protein functions for every species included in the analysis at varying, yet always high levels of precision and very good coverage.
Collapse
Affiliation(s)
- Samira Jaeger
- Knowledge Management in Bioinformatics, Humboldt-Universitat zu Berlin Unter den Linden 6, 10099 Berlin, Germany.
| | | | | |
Collapse
|
31
|
Jesmin J, Rashid MS, Jamil H, Hontecillas R, Bassaganya-Riera J. Gene regulatory network reveals oxidative stress as the underlying molecular mechanism of type 2 diabetes and hypertension. BMC Med Genomics 2010; 3:45. [PMID: 20942928 PMCID: PMC2965702 DOI: 10.1186/1755-8794-3-45] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 10/13/2010] [Indexed: 01/22/2023] Open
Abstract
Background The prevalence of diabetes is increasing worldwide. It has been long known that increased rates of inflammatory diseases, such as obesity (OBS), hypertension (HT) and cardiovascular diseases (CVD) are highly associated with type 2 diabetes (T2D). T2D and/or OBS can develop independently, due to genetic, behavioral or lifestyle-related variables but both lead to oxidative stress generation. The underlying mechanisms by which theses complications arise and manifest together remain poorly understood. Protein-protein interactions regulate nearly every living process. Availability of high-throughput genomic data has enabled unprecedented views of gene and protein co-expression, co-regulations and interactions in cellular systems. Methods The present work, applied a systems biology approach to develop gene interaction network models, comprised of high throughput genomic and PPI data for T2D. The genes differentially regulated through T2D were 'mined' and their 'wirings' were studied to get a more complete understanding of the overall gene network topology and their role in disease progression. Results By analyzing the genes related to T2D, HT and OBS, a highly regulated gene-disease integrated network model has been developed that provides useful functional linkages among groups of genes and thus addressing how different inflammatory diseases are connected and propagated at genetic level. Based on the investigations around the 'hubs' that provided more meaningful insights about the cross-talk within gene-disease networks in terms of disease phenotype association with oxidative stress and inflammation, a hypothetical co-regulation disease mechanism model been proposed. The results from this study revealed that the oxidative stress mediated regulation cascade is the common mechanistic link among the pathogenesis of T2D, HT and other inflammatory diseases such as OBS. Conclusion The findings provide a novel comprehensive approach for understanding the pathogenesis of various co-associated chronic inflammatory diseases by combining the power of pathway analysis with gene regulatory network evaluation.
Collapse
Affiliation(s)
- Jesmin Jesmin
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Bangladesh.
| | | | | | | | | |
Collapse
|
32
|
Marras E, Travaglione A, Chaurasia G, Futschik M, Capobianco E. Inferring modules from human protein interactome classes. BMC SYSTEMS BIOLOGY 2010; 4:102. [PMID: 20653930 PMCID: PMC2923113 DOI: 10.1186/1752-0509-4-102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 07/23/2010] [Indexed: 12/12/2022]
Abstract
Background The integration of protein-protein interaction networks derived from high-throughput screening approaches and complementary sources is a key topic in systems biology. Although integration of protein interaction data is conventionally performed, the effects of this procedure on the result of network analyses has not been examined yet. In particular, in order to optimize the fusion of heterogeneous interaction datasets, it is crucial to consider not only their degree of coverage and accuracy, but also their mutual dependencies and additional salient features. Results We examined this issue based on the analysis of modules detected by network clustering methods applied to both integrated and individual (disaggregated) data sources, which we call interactome classes. Due to class diversity, we deal with variable dependencies of data features arising from structural specificities and biases, but also from possible overlaps. Since highly connected regions of the human interactome may point to potential protein complexes, we have focused on the concept of modularity, and elucidated the detection power of module extraction algorithms by independent validations based on GO, MIPS and KEGG. From the combination of protein interactions with gene expressions, a confidence scoring scheme has been proposed before proceeding via GO with further classification in permanent and transient modules. Conclusions Disaggregated interactomes are shown to be informative for inferring modularity, thus contributing to perform an effective integrative analysis. Validation of the extracted modules by multiple annotation allows for the assessment of confidence measures assigned to the modules in a protein pathway context. Notably, the proposed multilayer confidence scheme can be used for network calibration by enabling a transition from unweighted to weighted interactomes based on biological evidence.
Collapse
Affiliation(s)
- Elisabetta Marras
- CRS4 Bioinformatics Laboratory-Technology Park of Sardinia, Pula (Cagliari), Sardinia, Italy
| | | | | | | | | |
Collapse
|
33
|
Wang J, Zhou X, Zhu J, Zhou C, Guo Z. Revealing and avoiding bias in semantic similarity scores for protein pairs. BMC Bioinformatics 2010; 11:290. [PMID: 20509916 PMCID: PMC2903568 DOI: 10.1186/1471-2105-11-290] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 05/28/2010] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Semantic similarity scores for protein pairs are widely applied in functional genomic researches for finding functional clusters of proteins, predicting protein functions and protein-protein interactions, and for identifying putative disease genes. However, because some proteins, such as those related to diseases, tend to be studied more intensively, annotations are likely to be biased, which may affect applications based on semantic similarity measures. Thus, it is necessary to evaluate the effects of the bias on semantic similarity scores between proteins and then find a method to avoid them. RESULTS First, we evaluated 14 commonly used semantic similarity scores for protein pairs and demonstrated that they significantly correlated with the numbers of annotation terms for the proteins (also known as the protein annotation length). These results suggested that current applications of the semantic similarity scores between proteins might be unreliable. Then, to reduce this annotation bias effect, we proposed normalizing the semantic similarity scores between proteins using the power transformation of the scores. We provide evidence that this improves performance in some applications. CONCLUSIONS Current semantic similarity measures for protein pairs are highly dependent on protein annotation lengths, which are subject to biological research bias. This affects applications that are based on these semantic similarity scores, especially in clustering studies that rely on score magnitudes. The normalized scores proposed in this paper can reduce the effects of this bias to some extent.
Collapse
Affiliation(s)
- Jing Wang
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xianxiao Zhou
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Jing Zhu
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Chenggui Zhou
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Zheng Guo
- Bioinformatics Centre, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 610054, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| |
Collapse
|
34
|
Wright GJ. Signal initiation in biological systems: the properties and detection of transient extracellular protein interactions. MOLECULAR BIOSYSTEMS 2010; 5:1405-12. [PMID: 19593473 PMCID: PMC2898632 DOI: 10.1039/b903580j] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Extracellular glycoprotein interactions are not detected by most high throughput assays creating “blind-spots” in protein interaction maps. This review examines this problem and discusses recent advances that have begun to address it.
Individual cells within biological systems frequently coordinate their functions through signals initiated by specific extracellular protein interactions involving receptors that bridge the cellular membrane. Due to their biochemical nature, these membrane-embedded receptor proteins are difficult to manipulate and their interactions are characterised by very weak binding strengths that cannot be detected using popular high throughput assays. This review will provide a general outline of the biochemical attributes of receptor proteins focussing in particular on the biophysical properties of their transient interactions. Methods that are able to detect these weak extracellular binding events and especially those that can be used for identifying novel interactions will be compared. Finally, I discuss the feasibility of constructing a complete and accurate extracellular protein interaction map, and the methods that are likely to be useful in achieving this goal.
Collapse
Affiliation(s)
- Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.
| |
Collapse
|
35
|
Malik R, Dulla K, Nigg EA, Körner R. From proteome lists to biological impact--tools and strategies for the analysis of large MS data sets. Proteomics 2010; 10:1270-1283. [PMID: 20077408 DOI: 10.1002/pmic.200900365] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 11/16/2009] [Indexed: 01/03/2025]
Abstract
MS has become a method-of-choice for proteome analysis, generating large data sets, which reflect proteome-scale protein-protein interaction and PTM networks. However, while a rapid growth in large-scale proteomics data can be observed, the sound biological interpretation of these results clearly lags behind. Therefore, combined efforts of bioinformaticians and biologists have been made to develop strategies and applications to help experimentalists perform this crucial task. This review presents an overview of currently available analytical strategies and tools to extract biologically relevant information from large protein lists. Moreover, we also present current research publications making use of these tools as examples of how the presented strategies may be incorporated into proteomic workflows. Emphasis is placed on the analysis of Gene Ontology terms, interaction networks, biological pathways and PTMs. In addition, topics including domain analysis and text mining are reviewed in the context of computational analysis of proteomic results. We expect that these types of analyses will significantly contribute to a deeper understanding of the role of individual proteins, protein networks and pathways in complex systems.
Collapse
Affiliation(s)
- Rainer Malik
- Max Planck Institute of Biochemistry, Department of Cell Biology, Martinsried, Germany
| | | | | | | |
Collapse
|
36
|
Qi Y, Dhiman HK, Bhola N, Budyak I, Kar S, Man D, Dutta A, Tirupula K, Carr BI, Grandis J, Bar-Joseph Z, Klein-Seetharaman J. Systematic prediction of human membrane receptor interactions. Proteomics 2010; 9:5243-55. [PMID: 19798668 DOI: 10.1002/pmic.200900259] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Membrane receptor-activated signal transduction pathways are integral to cellular functions and disease mechanisms in humans. Identification of the full set of proteins interacting with membrane receptors by high-throughput experimental means is difficult because methods to directly identify protein interactions are largely not applicable to membrane proteins. Unlike prior approaches that attempted to predict the global human interactome, we used a computational strategy that only focused on discovering the interacting partners of human membrane receptors leading to improved results for these proteins. We predict specific interactions based on statistical integration of biological data containing highly informative direct and indirect evidences together with feedback from experts. The predicted membrane receptor interactome provides a system-wide view, and generates new biological hypotheses regarding interactions between membrane receptors and other proteins. We have experimentally validated a number of these interactions. The results suggest that a framework of systematically integrating computational predictions, global analyses, biological experimentation and expert feedback is a feasible strategy to study the human membrane receptor interactome.
Collapse
Affiliation(s)
- Yanjun Qi
- School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wei P, Pan W. Network-based genomic discovery: application and comparison of Markov random field models. J R Stat Soc Ser C Appl Stat 2010; 59:105-125. [PMID: 21373371 PMCID: PMC3046412 DOI: 10.1111/j.1467-9876.2009.00686.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
As biological knowledge accumulates rapidly, gene networks encoding genome-wide gene-gene interactions have been constructed. As an improvement over the standard mixture model that tests all the genes iid a priori, Wei and Li (2007) and Wei and Pan (2008) proposed modeling a gene network as a Discrete- or Gaussian-Markov random field (DMRF or GMRF) respectively in a mixture model to analyze genomic data. However, how these methods compare in practical applications in not well understood and this is the aim here. We also propose two novel constraints in prior specifications for the GMRF model and a fully Bayesian approach to the DMRF model. We assess the accuracy of estimating the False Discovery Rate (FDR) by posterior probabilities in the context of MRF models. Applications to a ChIP-chip data set and simulated data show that the modified GMRF models has superior performance as compared with other models, while both MRF-based mixture models, with reasonable robustness to misspecified gene networks, outperform the standard mixture model.
Collapse
Affiliation(s)
- Peng Wei
- University of Minnesota, Minneapolis, USA
| | - Wei Pan
- University of Minnesota, Minneapolis, USA
| |
Collapse
|
38
|
Lindfors E, Gopalacharyulu PV, Halperin E, Orešič M. Detection of molecular paths associated with insulitis and type 1 diabetes in non-obese diabetic mouse. PLoS One 2009; 4:e7323. [PMID: 19798418 PMCID: PMC2749452 DOI: 10.1371/journal.pone.0007323] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 09/13/2009] [Indexed: 12/31/2022] Open
Abstract
Recent clinical evidence suggests important role of lipid and amino acid metabolism in early pre-autoimmune stages of type 1 diabetes pathogenesis. We study the molecular paths associated with the incidence of insulitis and type 1 diabetes in the Non-Obese Diabetic (NOD) mouse model using available gene expression data from the pancreatic tissue from young pre-diabetic mice. We apply a graph-theoretic approach by using a modified color coding algorithm to detect optimal molecular paths associated with specific phenotypes in an integrated biological network encompassing heterogeneous interaction data types. In agreement with our recent clinical findings, we identified a path downregulated in early insulitis involving dihydroxyacetone phosphate acyltransferase (DHAPAT), a key regulator of ether phospholipid synthesis. The pathway involving serine/threonine-protein phosphatase (PP2A), an upstream regulator of lipid metabolism and insulin secretion, was found upregulated in early insulitis. Our findings provide further evidence for an important role of lipid metabolism in early stages of type 1 diabetes pathogenesis, as well as suggest that such dysregulation of lipids and related increased oxidative stress can be tracked to beta cells.
Collapse
Affiliation(s)
- Erno Lindfors
- VTT Technical Research Centre of Finland, Espoo, Finland
| | | | - Eran Halperin
- International Computer Science Institute, Berkeley, California, United States of America
| | - Matej Orešič
- VTT Technical Research Centre of Finland, Espoo, Finland
- * E-mail:
| |
Collapse
|
39
|
Çakır T, Hendriks MMWB, Westerhuis JA, Smilde AK. Metabolic network discovery through reverse engineering of metabolome data. Metabolomics 2009; 5:318-329. [PMID: 19718266 PMCID: PMC2731157 DOI: 10.1007/s11306-009-0156-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 01/16/2009] [Indexed: 11/29/2022]
Abstract
Reverse engineering of high-throughput omics data to infer underlying biological networks is one of the challenges in systems biology. However, applications in the field of metabolomics are rather limited. We have focused on a systematic analysis of metabolic network inference from in silico metabolome data based on statistical similarity measures. Three different data types based on biological/environmental variability around steady state were analyzed to compare the relative information content of the data types for inferring the network. Comparing the inference power of different similarity scores indicated the clear superiority of conditioning or pruning based scores as they have the ability to eliminate indirect interactions. We also show that a mathematical measure based on the Fisher information matrix gives clues on the information quality of different data types to better represent the underlying metabolic network topology. Results on several datasets of increasing complexity consistently show that metabolic variations observed at steady state, the simplest experimental analysis, are already informative to reveal the connectivity of the underlying metabolic network with a low false-positive rate when proper similarity-score approaches are employed. For experimental situations this implies that a single organism under slightly varying conditions may already generate more than enough information to rightly infer networks. Detailed examination of the strengths of interactions of the underlying metabolic networks demonstrates that the edges that cannot be captured by similarity scores mainly belong to metabolites connected with weak interaction strength. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s11306-009-0156-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tunahan Çakır
- Biosystems Data Analysis, Swammerdam Institute for Life Sciences, University of Amsterdam, Nieuwe Achtergracht 166, 1018 WV Amsterdam, The Netherlands
- Department of Metabolic and Endocrine Diseases, University Medical Centre Utrecht, Utrecht, The Netherlands
- Netherlands Metabolomics Centre, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Margriet M. W. B. Hendriks
- Department of Metabolic and Endocrine Diseases, University Medical Centre Utrecht, Utrecht, The Netherlands
- Netherlands Metabolomics Centre, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Johan A. Westerhuis
- Biosystems Data Analysis, Swammerdam Institute for Life Sciences, University of Amsterdam, Nieuwe Achtergracht 166, 1018 WV Amsterdam, The Netherlands
- Netherlands Metabolomics Centre, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Age K. Smilde
- Biosystems Data Analysis, Swammerdam Institute for Life Sciences, University of Amsterdam, Nieuwe Achtergracht 166, 1018 WV Amsterdam, The Netherlands
- Netherlands Metabolomics Centre, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
40
|
Sowa ME, Bennett EJ, Gygi SP, Harper JW. Defining the human deubiquitinating enzyme interaction landscape. Cell 2009; 138:389-403. [PMID: 19615732 PMCID: PMC2716422 DOI: 10.1016/j.cell.2009.04.042] [Citation(s) in RCA: 1267] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 02/09/2009] [Accepted: 04/20/2009] [Indexed: 01/11/2023]
Abstract
Deubiquitinating enzymes (Dubs) function to remove covalently attached ubiquitin from proteins, thereby controlling substrate activity and/or abundance. For most Dubs, their functions, targets, and regulation are poorly understood. To systematically investigate Dub function, we initiated a global proteomic analysis of Dubs and their associated protein complexes. This was accomplished through the development of a software platform called CompPASS, which uses unbiased metrics to assign confidence measurements to interactions from parallel nonreciprocal proteomic data sets. We identified 774 candidate interacting proteins associated with 75 Dubs. Using Gene Ontology, interactome topology classification, subcellular localization, and functional studies, we link Dubs to diverse processes, including protein turnover, transcription, RNA processing, DNA damage, and endoplasmic reticulum-associated degradation. This work provides the first glimpse into the Dub interaction landscape, places previously unstudied Dubs within putative biological pathways, and identifies previously unknown interactions and protein complexes involved in this increasingly important arm of the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Mathew E. Sowa
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Eric J. Bennett
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - J. Wade Harper
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
41
|
Chen JY, Mamidipalli S, Huan T. HAPPI: an online database of comprehensive human annotated and predicted protein interactions. BMC Genomics 2009; 10 Suppl 1:S16. [PMID: 19594875 PMCID: PMC2709259 DOI: 10.1186/1471-2164-10-s1-s16] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Human protein-protein interaction (PPIs) data are the foundation for understanding molecular signalling networks and the functional roles of biomolecules. Several human PPI databases have become available; however, comparisons of these datasets have suggested limited data coverage and poor data quality. Ongoing collection and integration of human PPIs from different sources, both experimentally and computationally, can enable disease-specific network biology modelling in translational bioinformatics studies. Results We developed a new web-based resource, the Human Annotated and Predicted Protein Interaction (HAPPI) database, located at . The HAPPI database was created by extracting and integrating publicly available protein interaction databases, including HPRD, BIND, MINT, STRING, and OPHID, using database integration techniques. We designed a unified entity-relationship data model to resolve semantic level differences of diverse concepts involved in PPI data integration. We applied a unified scoring model to give each PPI a measure of its reliability that can place each PPI at one of the five star rank levels from 1 to 5. We assessed the quality of PPIs contained in the new HAPPI database, using evolutionary conserved co-expression pairs called "MetaGene" pairs to measure the extent of MetaGene pair and PPI pair overlaps. While the overall quality of the HAPPI database across all star ranks is comparable to the overall qualities of HPRD or IntNetDB, the subset of the HAPPI database with star ranks between 3 and 5 has a much higher average quality than all other human PPI databases. As of summer 2008, the database contains 142,956 non-redundant, medium to high-confidence level human protein interaction pairs among 10,592 human proteins. The HAPPI database web application also provides …” should be “The HAPPI database web application also provides hyperlinked information of genes, pathways, protein domains, protein structure displays, and sequence feature maps for interactive exploration of PPI data in the database. Conclusion HAPPI is by far the most comprehensive public compilation of human protein interaction information. It enables its users to fully explore PPI data with quality measures and annotated information necessary for emerging network biology studies.
Collapse
Affiliation(s)
- Jake Yue Chen
- School of Informatics, Indiana University - Purdue University, Indianapolis, IN, USA.
| | | | | |
Collapse
|
42
|
Abstract
Over the past few years, the number of known protein-protein interactions has increased substantially. To make this information more readily available, a number of publicly available databases have set out to collect and store protein-protein interaction data. Protein-protein interactions have been retrieved from six major databases, integrated and the results compared. The six databases (the Biological General Repository for Interaction Datasets [BioGRID], the Molecular INTeraction database [MINT], the Biomolecular Interaction Network Database [BIND], the Database of Interacting Proteins [DIP], the IntAct molecular interaction database [IntAct] and the Human Protein Reference Database [HPRD]) differ in scope and content; integration of all datasets is non-trivial owing to differences in data annotation. With respect to human protein-protein interaction data, HPRD seems to be the most comprehensive. To obtain a complete dataset, however, interactions from all six databases have to be combined. To overcome this limitation, meta-databases such as the Agile Protein Interaction Database (APID) offer access to integrated protein-protein interaction datasets, although these also currently have certain restrictions.
Collapse
|
43
|
Banky D, Ordog R, Grolmusz V. NASCENT: an automatic protein interaction network generation tool for non-model organisms. Bioinformation 2009; 3:361-3. [PMID: 19707301 PMCID: PMC2720673 DOI: 10.6026/97320630003361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 04/02/2009] [Accepted: 04/07/2009] [Indexed: 01/30/2023] Open
Abstract
Large quantity of reliable protein interaction data are available for model organisms in public depositories (e.g., MINT, DIP, HPRD, INTERACT). Most data correspond to
experiments with the proteins of Saccharomyces cerevisiae, Drosophila melanogaster, Homo sapiens, Caenorhabditis elegans, Escherichia coli and
Mus musculus. For other important organisms the data availability is poor or non-existent. Here we present NASCENT, a completely automatic web-based tool and also
a downloadable Java program, capable of modeling and generating protein interaction networks even for non-model organisms. The tool performs protein interaction network modeling
through gene-name mapping, and outputs the resulting network in graphical form and also in computer-readable graph-forms, directly applicable by popular network modeling
software.
Collapse
Affiliation(s)
- Daniel Banky
- Protein Information Technology Group, Eotvos University, H-1117 Budapest, Hungary
| | | | | |
Collapse
|
44
|
Bernthaler A, Mühlberger I, Fechete R, Perco P, Lukas A, Mayer B. A dependency graph approach for the analysis of differential gene expression profiles. MOLECULAR BIOSYSTEMS 2009; 5:1720-31. [PMID: 19585005 DOI: 10.1039/b903109j] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Andreas Bernthaler
- Theory and Logics Group, Institute of Computer Languages, Vienna University of Technology, Favoritenstrasse 9-11, A-1040 Vienna, Austria.
| | | | | | | | | | | |
Collapse
|
45
|
Venkatesan K, Rual JF, Vazquez A, Stelzl U, Lemmens I, Hirozane-Kishikawa T, Hao T, Zenkner M, Xin X, Goh KI, Yildirim MA, Simonis N, Heinzmann K, Gebreab F, Sahalie JM, Cevik S, Simon C, de Smet AS, Dann E, Smolyar A, Vinayagam A, Yu H, Szeto D, Borick H, Dricot A, Klitgord N, Murray RR, Lin C, Lalowski M, Timm J, Rau K, Boone C, Braun P, Cusick ME, Roth FP, Hill DE, Tavernier J, Wanker EE, Barabási AL, Vidal M. An empirical framework for binary interactome mapping. Nat Methods 2009; 6:83-90. [PMID: 19060904 PMCID: PMC2872561 DOI: 10.1038/nmeth.1280] [Citation(s) in RCA: 658] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Accepted: 11/10/2008] [Indexed: 01/05/2023]
Abstract
Several attempts have been made to systematically map protein-protein interaction, or 'interactome', networks. However, it remains difficult to assess the quality and coverage of existing data sets. Here we describe a framework that uses an empirically-based approach to rigorously dissect quality parameters of currently available human interactome maps. Our results indicate that high-throughput yeast two-hybrid (HT-Y2H) interactions for human proteins are more precise than literature-curated interactions supported by a single publication, suggesting that HT-Y2H is suitable to map a significant portion of the human interactome. We estimate that the human interactome contains approximately 130,000 binary interactions, most of which remain to be mapped. Similar to estimates of DNA sequence data quality and genome size early in the Human Genome Project, estimates of protein interaction data quality and interactome size are crucial to establish the magnitude of the task of comprehensive human interactome mapping and to elucidate a path toward this goal.
Collapse
Affiliation(s)
- Kavitha Venkatesan
- Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, 1 Jimmy Fund Way, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sanderson CM. The Cartographers toolbox: building bigger and better human protein interaction networks. BRIEFINGS IN FUNCTIONAL GENOMICS AND PROTEOMICS 2008; 8:1-11. [DOI: 10.1093/bfgp/elp003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
47
|
Chaurasia G, Malhotra S, Russ J, Schnoegl S, Hänig C, Wanker EE, Futschik ME. UniHI 4: new tools for query, analysis and visualization of the human protein-protein interactome. Nucleic Acids Res 2008; 37:D657-60. [PMID: 18984619 PMCID: PMC2686569 DOI: 10.1093/nar/gkn841] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Human protein interaction maps have become important tools of biomedical research for the elucidation of molecular mechanisms and the identification of new modulators of disease processes. The Unified Human Interactome database (UniHI, http://www.unihi.org) provides researchers with a comprehensive platform to query and access human protein–protein interaction (PPI) data. Since its first release, UniHI has considerably increased in size. The latest update of UniHI includes over 250 000 interactions between ∼22 300 unique proteins collected from 14 major PPI sources. However, this wealth of data also poses new challenges for researchers due to the complexity of interaction networks retrieved from the database. We therefore developed several new tools to query, analyze and visualize human PPI networks. Most importantly, UniHI allows now the construction of tissue-specific interaction networks and focused querying of canonical pathways. This will enable researchers to target their analysis and to prioritize candidate proteins for follow-up studies.
Collapse
Affiliation(s)
- Gautam Chaurasia
- Institute for Theoretical Biology, Charité, Humboldt-University, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Abstraction of intracellular biomolecular interactions into networks is useful for data integration and graph analysis. Network analysis tools facilitate predictions of novel functions for proteins, prediction of functional interactions and identification of intracellular modules. These efforts are linked with drug and phenotype data to accelerate drug-target and biomarker discovery. This review highlights the currently available varieties of mammalian biomolecular networks, and surveys methods and tools to construct, compare, integrate, visualise and analyse such networks.
Collapse
Affiliation(s)
- A Ma'ayan
- Mount Sinai School of Medicine, Department of Pharmacology and Systems Therapeutics, New York, NY 10029-6574, USA.
| |
Collapse
|
49
|
Furney SJ, Calvo B, Larrañaga P, Lozano JA, Lopez-Bigas N. Prioritization of candidate cancer genes--an aid to oncogenomic studies. Nucleic Acids Res 2008; 36:e115. [PMID: 18710882 PMCID: PMC2566894 DOI: 10.1093/nar/gkn482] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The development of techniques for oncogenomic analyses such as array comparative genomic hybridization, messenger RNA expression arrays and mutational screens have come to the fore in modern cancer research. Studies utilizing these techniques are able to highlight panels of genes that are altered in cancer. However, these candidate cancer genes must then be scrutinized to reveal whether they contribute to oncogenesis or are coincidental and non-causative. We present a computational method for the prioritization of candidate (i) proto-oncogenes and (ii) tumour suppressor genes from oncogenomic experiments. We constructed computational classifiers using different combinations of sequence and functional data including sequence conservation, protein domains and interactions, and regulatory data. We found that these classifiers are able to distinguish between known cancer genes and other human genes. Furthermore, the classifiers also discriminate candidate cancer genes from a recent mutational screen from other human genes. We provide a web-based facility through which cancer biologists may access our results and we propose computational cancer gene classification as a useful method of prioritizing candidate cancer genes identified in oncogenomic studies.
Collapse
Affiliation(s)
- Simon J Furney
- Research Unit on Biomedical Informatics, Experimental and Health Science Department, Universitat Pompeu Fabra, Barcelona 08080, Spain
| | | | | | | | | |
Collapse
|
50
|
Abstract
Interactions are the essence of all biomolecules because they cannot fulfill their roles without interacting with other molecules. Hence, mapping the interactions of biomolecules can be useful for understanding their roles and functions. Furthermore, the development of molecular based systems biology requires an understanding of the biomolecular interactions. In recent years, the mapping of protein-protein interactions in different species has been reported, but few reports have focused on the large-scale mapping of protein-protein interactions in human. Here, we review the developments in protein interaction mapping and we discuss issues and strategies for the mapping of the human protein interactome.
Collapse
|