1
|
Dave R, Giordano P, Roy S, Imran H. Identifying novel drug targets with computational precision. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:231-263. [PMID: 40175044 DOI: 10.1016/bs.apha.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Computational precision in drug discovery integrates algorithms and high-performance computing to analyze complex biological data with unprecedented accuracy, revolutionizing the identification of therapeutic targets. This process encompasses diverse computational and experimental approaches that enhance drug discovery's speed and precision. Advanced techniques like next-generation sequencing enable rapid genetic characterization, while proteomics explores protein expression and interactions driving disease progression. In-silico methods, including molecular docking, virtual screening, and pharmacophore modeling, predict interactions between small molecules and biological targets, accelerating early drug candidate identification. Structure-based drug design and molecular dynamics simulations refine drug designs by elucidating target structures and molecular behaviors. Ligand-based methods utilize known chemical properties to anticipate new compound activities. AI and machine learning optimizes data analysis, offering novel insights and improving predictive accuracy. Systems biology and network pharmacology provide a holistic view of biological networks, identifying critical nodes as potential drug targets, which traditional methods might overlook. Computational tools synergize with experimental techniques, enhancing the treatment of complex diseases with personalized medicine by tailoring therapies to individual patients. Ethical and regulatory compliance ensures clinical applicability, bridging computational predictions to effective therapies. This multi-dimensional approach marks a paradigm shift in modern medicine, delivering safer, more effective treatments with precision. By integrating bioinformatics, genomics, and proteomics, computational drug discovery has transformed how therapeutic interventions are developed, ensuring an era of personalized, efficient healthcare.
Collapse
Affiliation(s)
- Riya Dave
- Dentist, Gujarat University, Ahmedabad, Gujarat, India.
| | - Pierpaolo Giordano
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Sakshi Roy
- School of Medicine, Queens University, Belfast, United Kingdom
| | - Hiba Imran
- Karachi Metropolitan University, Karachi, Pakistan
| |
Collapse
|
2
|
Moncla LHM, Mathieu S, Sylla MS, Bossé Y, Thériault S, Arsenault BJ, Mathieu P. Mendelian randomization of circulating proteome identifies actionable targets in heart failure. BMC Genomics 2022; 23:588. [PMID: 35964012 PMCID: PMC9375407 DOI: 10.1186/s12864-022-08811-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/30/2022] [Indexed: 11/21/2022] Open
Abstract
Background Heart failure (HF) is a prevalent cause of mortality and morbidity. The molecular drivers of HF are still largely unknown. Results We aimed to identify circulating proteins causally associated with HF by leveraging genome-wide genetic association data for HF including 47,309 cases and 930,014 controls. We performed two-sample Mendelian randomization (MR) with multiple cis instruments as well as network and enrichment analysis using data from blood protein quantitative trait loci (pQTL) (2,965 blood proteins) measured in 3,301 individuals. Nineteen blood proteins were causally associated with HF, were not subject to reverse causality and were enriched in ligand-receptor and glycosylation molecules. Network pathway analysis of the blood proteins showed enrichment in NF-kappa B, TGF beta, lipid in atherosclerosis and fluid shear stress. Cross-phenotype analysis of HF identified genetic overlap with cardiovascular drugs, myocardial infarction, parental longevity and low-density cholesterol. Multi-trait MR identified causal associations between HF-associated blood proteins and cardiovascular outcomes. Multivariable MR showed that association of BAG3, MIF and APOA5 with HF were mediated by the blood pressure and coronary artery disease. According to the directional effect and biological action, 7 blood proteins are targets of existing drugs or are tractable for the development of novel therapeutics. Among the pathways, sialyl Lewis x and the activin type II receptor are potential druggable candidates. Conclusions Integrative MR analyses of the blood proteins identified causally-associated proteins with HF and revealed pleiotropy of the blood proteome with cardiovascular risk factors. Some of the proteins or pathway related mechanisms could be targeted as novel treatment approach in HF. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08811-2.
Collapse
Affiliation(s)
- Louis-Hippolyte Minvielle Moncla
- Genomic Medecine and Molecular Epidemiology Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec, G1V-4G5, Canada
| | - Samuel Mathieu
- Genomic Medecine and Molecular Epidemiology Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec, G1V-4G5, Canada
| | - Mame Sokhna Sylla
- Genomic Medecine and Molecular Epidemiology Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec, G1V-4G5, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Quebec, Canada
| | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec, Canada
| | - Benoit J Arsenault
- Genomic Medecine and Molecular Epidemiology Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec, G1V-4G5, Canada.,Department of Medicine, Laval University, Quebec, Canada
| | - Patrick Mathieu
- Genomic Medecine and Molecular Epidemiology Laboratory, Quebec Heart and Lung Institute, Laval University, Quebec, G1V-4G5, Canada. .,Department of Surgery, Laval University, Quebec, Canada.
| |
Collapse
|
3
|
Investigation of the mechanism of Shen Qi Wan prescription in the treatment of T2DM via network pharmacology and molecular docking. In Silico Pharmacol 2022; 10:9. [PMID: 35673584 DOI: 10.1007/s40203-022-00124-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 05/16/2022] [Indexed: 10/18/2022] Open
Abstract
Shen Qi Wan (SQW) prescription has been used to treat type 2 diabetes mellitus (T2DM) for thousands of years, but its pharmacological mechanism is still unclear. The network pharmacology method was used to reveal the potential pharmacological mechanism of SQW in the treatment of T2DM in this study. Nine core targets were identified through protein-protein interaction (PPI) network analysis and KEGG pathway enrichment analysis, which were AKT1, INSR, SLC2A1, EGFR, PPARG, PPARA, GCK, NOS3, and PTPN1. Besides, this study found that SQW treated the T2DM through insulin resistance (has04931), insulin signaling pathway (has04910), adipocytokine signaling pathway (has04920), AMPK signaling pathway (has04152) and FoxO signaling pathway (has04068) via ingredient-hub target-pathway network analysis. Finally, molecular docking was used to verify the drug-target interaction network in this research. This study provides a certain explanation for treating T2DM by SQW prescription, and provides a certain angle and method for researchers to study the mechanism of TCM in the treatment of complex diseases. Supplementary information The online version contains supplementary material available at 10.1007/s40203-022-00124-2.
Collapse
|
4
|
In silico Methods for Identification of Potential Therapeutic Targets. Interdiscip Sci 2022; 14:285-310. [PMID: 34826045 PMCID: PMC8616973 DOI: 10.1007/s12539-021-00491-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 11/01/2022]
Abstract
AbstractAt the initial stage of drug discovery, identifying novel targets with maximal efficacy and minimal side effects can improve the success rate and portfolio value of drug discovery projects while simultaneously reducing cycle time and cost. However, harnessing the full potential of big data to narrow the range of plausible targets through existing computational methods remains a key issue in this field. This paper reviews two categories of in silico methods—comparative genomics and network-based methods—for finding potential therapeutic targets among cellular functions based on understanding their related biological processes. In addition to describing the principles, databases, software, and applications, we discuss some recent studies and prospects of the methods. While comparative genomics is mostly applied to infectious diseases, network-based methods can be applied to infectious and non-infectious diseases. Nonetheless, the methods often complement each other in their advantages and disadvantages. The information reported here guides toward improving the application of big data-driven computational methods for therapeutic target discovery.
Graphical abstract
Collapse
|
5
|
Viacava Follis A. Centrality of drug targets in protein networks. BMC Bioinformatics 2021; 22:527. [PMID: 34715787 PMCID: PMC8555226 DOI: 10.1186/s12859-021-04342-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/23/2021] [Indexed: 01/13/2023] Open
Abstract
Background In the pharmaceutical industry, competing for few validated drug targets there is a drive to identify new ways of therapeutic intervention. Here, we attempted to define guidelines to evaluate a target’s ‘fitness’ based on its node characteristics within annotated protein functional networks to complement contingent therapeutic hypotheses. Results We observed that targets of approved, selective small molecule drugs exhibit high node centrality within protein networks relative to a broader set of investigational targets spanning various development stages. Targets of approved drugs also exhibit higher centrality than other proteins within their respective functional class. These findings expand on previous reports of drug targets’ network centrality by suggesting some centrality metrics such as low topological coefficient as inherent characteristics of a ‘good’ target, relative to other exploratory targets and regardless of its functional class. These centrality metrics could thus be indicators of an individual protein’s ‘fitness’ as potential drug target. Correlations between protein nodes’ network centrality and number of associated publications underscored the possibility of knowledge bias as an inherent limitation to such predictions. Conclusions Despite some entanglement with knowledge bias, like structure-oriented ‘druggability’ assessments of new protein targets, centrality metrics could assist early pharmaceutical discovery teams in evaluating potential targets with limited experimental proof of concept and help allocate resources for an effective drug discovery pipeline. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04342-x.
Collapse
Affiliation(s)
- Ariele Viacava Follis
- EMD Serono Research and Development Inc., 45A Middlesex Turnpike, Billerica, MA, 01821, USA.
| |
Collapse
|
6
|
Santos EC, Gomes RB, Fernandes PV, Ferreira MA, Abdelhay ESFW. The protein-protein interaction network of intestinal gastric cancer patients reveals hub proteins with potential prognostic value. Cancer Biomark 2021; 33:83-96. [PMID: 34366321 DOI: 10.3233/cbm-203225] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Gastric cancer (GC) is the third leading cause of cancer worldwide. According to the Lauren classification, gastric adenocarcinoma is divided into two subtypes: diffuse and intestinal. The development of intestinal gastric cancer (IGC) can take years and involves multiple factors. OBJECTIVE To investigate the protein profile of tumor samples from patients with IGC in comparison with adjacent nontumor tissue samples. METHODS We used label-free nano-LC-MS/MS to identify proteins from the tissues samples. The results were analyzed using MetaCore™ software to access functional enrichment information. Protein-protein interactions (PPI) were predicted using STRING analysis. Hub proteins were determined using the Cytoscape plugin, CytoHubba. Survival analysis was performed using KM plotter. We identified 429 differentially expressed proteins whose pathways and processes were related to protein folding, apoptosis, and immune response. RESULTS The PPI network of these proteins showed enrichment modules related to the regulation of cell death, immune system, neutrophil degranulation, metabolism of RNA and chromatin DNA binding. From the PPI network, we identified 20 differentially expressed hub proteins, and assessed the prognostic value of the expression of genes that encode them. Among them, the expression of four hub genes was significantly associated with the overall survival of IGC patients. CONCLUSIONS This study reveals important findings that affect IGC development based on specific biological alterations in IGC patients. Bioinformatics analysis showed that the pathogenesis of IGC patients is complex and involves different interconnected biological processes. These findings may be useful in research on new targets to develop novel therapies to improve the overall survival of patients with IGC.
Collapse
Affiliation(s)
- Everton Cruz Santos
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia Para o Controle do Câncer, Rio de Janeiro, RJ, Brazil
| | - Renata Binato Gomes
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia Para o Controle do Câncer, Rio de Janeiro, RJ, Brazil
| | | | | | - Eliana Saul Furquim Werneck Abdelhay
- Stem Cell Laboratory, Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil.,Instituto Nacional de Ciência e Tecnologia Para o Controle do Câncer, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
7
|
Guo HB, Ghafari M, Dang W, Qin H. Protein interaction potential landscapes for yeast replicative aging. Sci Rep 2021; 11:7143. [PMID: 33785798 PMCID: PMC8010020 DOI: 10.1038/s41598-021-86415-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/15/2021] [Indexed: 11/17/2022] Open
Abstract
We proposed a novel interaction potential landscape approach to map the systems-level profile changes of gene networks during replicative aging in Saccharomyces cerevisiae. This approach enabled us to apply quasi-potentials, the negative logarithm of the probabilities, to calibrate the elevation of the interaction landscapes with young cells as a reference state. Our approach detected opposite landscape changes based on protein abundances from transcript levels, especially for intra-essential gene interactions. We showed that essential proteins play different roles from hub proteins on the age-dependent interaction potential landscapes. We verified that hub proteins tend to avoid other hub proteins, but essential proteins prefer to interact with other essential proteins. Overall, we showed that the interaction potential landscape is promising for inferring network profile change during aging and that the essential hub proteins may play an important role in the uncoupling between protein and transcript levels during replicative aging.
Collapse
Affiliation(s)
- Hao-Bo Guo
- Department of Computer Science and Engineering, The University of Tennessee at Chattanooga, Chattanooga, TN, 37405, USA.
- SimCenter, The University of Tennessee at Chattanooga, Chattanooga, TN, 37405, USA.
- Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson AFB, Dayton, OH, 45433, USA.
| | - Mehran Ghafari
- Department of Computer Science and Engineering, The University of Tennessee at Chattanooga, Chattanooga, TN, 37405, USA
| | - Weiwei Dang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hong Qin
- Department of Computer Science and Engineering, The University of Tennessee at Chattanooga, Chattanooga, TN, 37405, USA.
- SimCenter, The University of Tennessee at Chattanooga, Chattanooga, TN, 37405, USA.
- Department of Biology, Geology and Environmental Science, The University of Tennessee at Chattanooga, Chattanooga, TN, 37405, USA.
| |
Collapse
|
8
|
Active ingredients and mechanisms of Phellinus linteus (grown on Rosa multiflora) for alleviation of Type 2 diabetes mellitus through network pharmacology. Gene 2020; 768:145320. [PMID: 33248199 DOI: 10.1016/j.gene.2020.145320] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/28/2020] [Accepted: 11/14/2020] [Indexed: 12/11/2022]
Abstract
Phellinus linteus (mushroom) grown on Rosa multiflora (PL@RM), exposed beneficial effect and safety on Type 2 diabetes mellitus (T2DM) from Korean folk remedies. However, its active chemical constituents and mechanism(s) against T2DM have not been confirmed. Hence, we deciphered the active compounds and mechanism(s) of PL@RM against T2DM through network pharmacology. GC-MS of PL@RM manifested 54 compounds and drug-likeness properties of these compounds were confirmed by Lipinski's rule. The compound (40) related genes were composed of Similarity Ensemble Approach (SEA) and SwissTargetPrediction (STP). The overlapping genes (61) between the two databases were identified. Besides, the T2DM related genes (4,736) were extracted from DisGeNet and OMIM database. In parallel, a Venn diagram was constructed between the overlapping genes (61) and T2DM related genes (4,736), and finally, 48 genes were picked. The interactive networks between compounds and overlapping genes were plotted and visualized by RStudio. In addition, KEGG Pathway enrichment analysis was evaluated by String. String analysis showed that the mechanisms of PL@RM against T2DM were related to 16 pathways, where inhibition of gluconeogenesis by inactivating metabolic pathways was noted as the hub pathway of PL@RM against T2DM. Besides, bubble chart indicated that activation of the AMPK signaling pathway might enhance the insulin receptor (IR) phosphorylation, which is regarded the key signaling pathway of PL@RM against T2DM. Furthermore, the autodock vina revealed the promising binding affinity energy of the epicholesterol (the most drug-likeness compound) on HMGCR (hub gene). Overall, this work hints at the therapeutic evidence of PL@RM on T2DM, and this data expound the main chemical compounds and mechanisms of PL@RM against T2DM.
Collapse
|
9
|
Md Aksam VK, Chandrasekaran VM, Pandurangan S. Topological alternate centrality measure capturing drug targets in the network of MAPK pathways. IET Syst Biol 2018; 12:226-232. [PMID: 30259868 PMCID: PMC8687289 DOI: 10.1049/iet-syb.2017.0058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 04/04/2018] [Accepted: 04/30/2018] [Indexed: 12/18/2022] Open
Abstract
A new centrality of the nodes in the network is proposed called alternate centrality, which can isolate effective drug targets in the complex signalling network. Alternate centrality metric defined over the network substructure (four nodes - motifs). The nodes involving in alternative activation in the motifs gain in metric values. Targeting high alternative centrality nodes hypothesised to be destructive free to the network due to their alternative activation mechanism. Overlapping and crosstalk among the gene products in the conserved network of MAPK pathways selected for the study. In silico knock-out of high alternate centrality nodes causing rewiring in the network is investigated using MCF-7 breast cancer cell line-based data. Degree of top alternate centrality nodes lies between the degree of bridging and pagerank nodes. Node deletion of high alternate centrality on the centralities such as eccentricity, closeness, betweenness, stress, centroid and radiality causes low perturbation. The authors identified the following alternate centrality nodes ERK1, ERK2, MEKK2, MKK5, MKK4, MLK3, MLK2, MLK1, MEKK4, MEKK1, TAK1, P38alpha, ZAK, DLK, LZK, MLTKa/b and P38beta as efficient drug targets for breast cancer. Alternate centrality identifies effective drug targets and is free from intertwined biological processes and lethality.
Collapse
Affiliation(s)
- V K Md Aksam
- School of Advanced Sciences, VIT University, Vellore 632014, India
| | | | | |
Collapse
|
10
|
Dos Santos Vasconcelos CR, de Lima Campos T, Rezende AM. Building protein-protein interaction networks for Leishmania species through protein structural information. BMC Bioinformatics 2018; 19:85. [PMID: 29510668 PMCID: PMC5840830 DOI: 10.1186/s12859-018-2105-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 03/01/2018] [Indexed: 12/21/2022] Open
Abstract
Background Systematic analysis of a parasite interactome is a key approach to understand different biological processes. It makes possible to elucidate disease mechanisms, to predict protein functions and to select promising targets for drug development. Currently, several approaches for protein interaction prediction for non-model species incorporate only small fractions of the entire proteomes and their interactions. Based on this perspective, this study presents an integration of computational methodologies, protein network predictions and comparative analysis of the protozoan species Leishmania braziliensis and Leishmania infantum. These parasites cause Leishmaniasis, a worldwide distributed and neglected disease, with limited treatment options using currently available drugs. Results The predicted interactions were obtained from a meta-approach, applying rigid body docking tests and template-based docking on protein structures predicted by different comparative modeling techniques. In addition, we trained a machine-learning algorithm (Gradient Boosting) using docking information performed on a curated set of positive and negative protein interaction data. Our final model obtained an AUC = 0.88, with recall = 0.69, specificity = 0.88 and precision = 0.83. Using this approach, it was possible to confidently predict 681 protein structures and 6198 protein interactions for L. braziliensis, and 708 protein structures and 7391 protein interactions for L. infantum. The predicted networks were integrated to protein interaction data already available, analyzed using several topological features and used to classify proteins as essential for network stability. Conclusions The present study allowed to demonstrate the importance of integrating different methodologies of interaction prediction to increase the coverage of the protein interaction of the studied protocols, besides it made available protein structures and interactions not previously reported. Electronic supplementary material The online version of this article (10.1186/s12859-018-2105-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Crhisllane Rafaele Dos Santos Vasconcelos
- Microbiology Department of Instituto Aggeu Magalhães - FIOCRUZ, Recife, PE, Brazil. .,Genetics Department of Universidade Federal de Pernambuco, Recife, PE, Brazil.
| | - Túlio de Lima Campos
- Microbiology Department of Instituto Aggeu Magalhães - FIOCRUZ, Recife, PE, Brazil.,Bioinformatics Plataform of Instituto Aggeu Magalhães - FIOCRUZ, Recife, PE, Brazil
| | - Antonio Mauro Rezende
- Microbiology Department of Instituto Aggeu Magalhães - FIOCRUZ, Recife, PE, Brazil. .,Bioinformatics Plataform of Instituto Aggeu Magalhães - FIOCRUZ, Recife, PE, Brazil. .,Genetics Department of Universidade Federal de Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
11
|
Ludovini V, Bianconi F, Siggillino A, Piobbico D, Vannucci J, Metro G, Chiari R, Bellezza G, Puma F, Della Fazia MA, Servillo G, Crinò L. Gene identification for risk of relapse in stage I lung adenocarcinoma patients: a combined methodology of gene expression profiling and computational gene network analysis. Oncotarget 2017; 7:30561-74. [PMID: 27081700 PMCID: PMC5058701 DOI: 10.18632/oncotarget.8723] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/28/2016] [Indexed: 12/30/2022] Open
Abstract
Risk assessment and treatment choice remains a challenge in early non-small-cell lung cancer (NSCLC). The aim of this study was to identify novel genes involved in the risk of early relapse (ER) compared to no relapse (NR) in resected lung adenocarcinoma (AD) patients using a combination of high throughput technology and computational analysis. We identified 18 patients (n.13 NR and n.5 ER) with stage I AD. Frozen samples of patients in ER, NR and corresponding normal lung (NL) were subjected to Microarray technology and quantitative-PCR (Q-PCR). A gene network computational analysis was performed to select predictive genes. An independent set of 79 ADs stage I samples was used to validate selected genes by Q-PCR.From microarray analysis we selected 50 genes, using the fold change ratio of ER versus NR. They were validated both in pool and individually in patient samples (ER and NR) by Q-PCR. Fourteen increased and 25 decreased genes showed a concordance between two methods. They were used to perform a computational gene network analysis that identified 4 increased (HOXA10, CLCA2, AKR1B10, FABP3) and 6 decreased (SCGB1A1, PGC, TFF1, PSCA, SPRR1B and PRSS1) genes. Moreover, in an independent dataset of ADs samples, we showed that both high FABP3 expression and low SCGB1A1 expression was associated with a worse disease-free survival (DFS).Our results indicate that it is possible to define, through gene expression and computational analysis, a characteristic gene profiling of patients with an increased risk of relapse that may become a tool for patient selection for adjuvant therapy.
Collapse
Affiliation(s)
- Vienna Ludovini
- Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | - Fortunato Bianconi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Danilo Piobbico
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Jacopo Vannucci
- Department of Surgical and Biomedical Science, University of Perugia, Perugia, Italy
| | - Giulio Metro
- Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | - Rita Chiari
- Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | - Guido Bellezza
- Department of Experimental Medicine, Section of Anatomic Pathology and Histology, Perugia, Italy
| | - Francesco Puma
- Department of Surgical and Biomedical Science, University of Perugia, Perugia, Italy
| | | | - Giuseppe Servillo
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Lucio Crinò
- Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
12
|
Korcsmaros T, Schneider MV, Superti-Furga G. Next generation of network medicine: interdisciplinary signaling approaches. Integr Biol (Camb) 2017; 9:97-108. [PMID: 28106223 DOI: 10.1039/c6ib00215c] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In the last decade, network approaches have transformed our understanding of biological systems. Network analyses and visualizations have allowed us to identify essential molecules and modules in biological systems, and improved our understanding of how changes in cellular processes can lead to complex diseases, such as cancer, infectious and neurodegenerative diseases. "Network medicine" involves unbiased large-scale network-based analyses of diverse data describing interactions between genes, diseases, phenotypes, drug targets, drug transport, drug side-effects, disease trajectories and more. In terms of drug discovery, network medicine exploits our understanding of the network connectivity and signaling system dynamics to help identify optimal, often novel, drug targets. Contrary to initial expectations, however, network approaches have not yet delivered a revolution in molecular medicine. In this review, we propose that a key reason for the limited impact, so far, of network medicine is a lack of quantitative multi-disciplinary studies involving scientists from different backgrounds. To support this argument, we present existing approaches from structural biology, 'omics' technologies (e.g., genomics, proteomics, lipidomics) and computational modeling that point towards how multi-disciplinary efforts allow for important new insights. We also highlight some breakthrough studies as examples of the potential of these approaches, and suggest ways to make greater use of the power of interdisciplinarity. This review reflects discussions held at an interdisciplinary signaling workshop which facilitated knowledge exchange from experts from several different fields, including in silico modelers, computational biologists, biochemists, geneticists, molecular and cell biologists as well as cancer biologists and pharmacologists.
Collapse
Affiliation(s)
- Tamas Korcsmaros
- Earlham Institute, Norwich Research Park, Norwich, UK. and Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich, UK
| | | | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria and Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
13
|
Lu W, Wang X, Zhan X, Gazdar A. Meta-analysis approaches to combine multiple gene set enrichment studies. Stat Med 2017; 37:659-672. [PMID: 29052247 DOI: 10.1002/sim.7540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 07/02/2017] [Accepted: 09/29/2017] [Indexed: 11/09/2022]
Abstract
In the field of gene set enrichment analysis (GSEA), meta-analysis has been used to integrate information from multiple studies to present a reliable summarization of the expanding volume of individual biomedical research, as well as improve the power of detecting essential gene sets involved in complex human diseases. However, existing methods, Meta-Analysis for Pathway Enrichment (MAPE), may be subject to power loss because of (1) using gross summary statistics for combining end results from component studies and (2) using enrichment scores whose distributions depend on the set sizes. In this paper, we adapt meta-analysis approaches recently developed for genome-wide association studies, which are based on fixed effect and random effects (RE) models, to integrate multiple GSEA studies. We further develop a mixed strategy via adaptive testing for choosing RE versus FE models to achieve greater statistical efficiency as well as flexibility. In addition, a size-adjusted enrichment score based on a one-sided Kolmogorov-Smirnov statistic is proposed to formally account for varying set sizes when testing multiple gene sets. Our methods tend to have much better performance than the MAPE methods and can be applied to both discrete and continuous phenotypes. Specifically, the performance of the adaptive testing method seems to be the most stable in general situations.
Collapse
Affiliation(s)
- Wentao Lu
- Department of Statistical Science, Southern Methodist University, Dallas, TX 75275, USA
| | - Xinlei Wang
- Department of Statistical Science, Southern Methodist University, Dallas, TX 75275, USA
| | - Xiaowei Zhan
- Quantitative Biomedical Research Center, Center for the Genetics of Host Defense, Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adi Gazdar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
14
|
Kuenzi BM, Remsing Rix LL, Stewart PA, Fang B, Kinose F, Bryant AT, Boyle TA, Koomen JM, Haura EB, Rix U. Polypharmacology-based ceritinib repurposing using integrated functional proteomics. Nat Chem Biol 2017; 13:1222-1231. [PMID: 28991240 DOI: 10.1038/nchembio.2489] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
Targeted drugs are effective when they directly inhibit strong disease drivers, but only a small fraction of diseases feature defined actionable drivers. Alternatively, network-based approaches can uncover new therapeutic opportunities. Applying an integrated phenotypic screening, chemical and phosphoproteomics strategy, here we describe the anaplastic lymphoma kinase (ALK) inhibitor ceritinib as having activity across several ALK-negative lung cancer cell lines and identify new targets and network-wide signaling effects. Combining pharmacological inhibitors and RNA interference revealed a polypharmacology mechanism involving the noncanonical targets IGF1R, FAK1, RSK1 and RSK2. Mutating the downstream signaling hub YB1 protected cells from ceritinib. Consistent with YB1 signaling being known to cause taxol resistance, combination of ceritinib with paclitaxel displayed strong synergy, particularly in cells expressing high FAK autophosphorylation, which we show to be prevalent in lung cancer. Together, we present a systems chemical biology platform for elucidating multikinase inhibitor polypharmacology mechanisms, subsequent design of synergistic drug combinations, and identification of mechanistic biomarker candidates.
Collapse
Affiliation(s)
- Brent M Kuenzi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Cancer Biology PhD Program, University of South Florida, Tampa, Florida, USA
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Paul A Stewart
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Bin Fang
- Proteomics Core Facility, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Annamarie T Bryant
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Theresa A Boyle
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - John M Koomen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
15
|
Kanhaiya K, Czeizler E, Gratie C, Petre I. Controlling Directed Protein Interaction Networks in Cancer. Sci Rep 2017; 7:10327. [PMID: 28871116 PMCID: PMC5583175 DOI: 10.1038/s41598-017-10491-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023] Open
Abstract
Control theory is a well-established approach in network science, with applications in bio-medicine and cancer research. We build on recent results for structural controllability of directed networks, which identifies a set of driver nodes able to control an a-priori defined part of the network. We develop a novel and efficient approach for the (targeted) structural controllability of cancer networks and demonstrate it for the analysis of breast, pancreatic, and ovarian cancer. We build in each case a protein-protein interaction network and focus on the survivability-essential proteins specific to each cancer type. We show that these essential proteins are efficiently controllable from a relatively small computable set of driver nodes. Moreover, we adjust the method to find the driver nodes among FDA-approved drug-target nodes. We find that, while many of the drugs acting on the driver nodes are part of known cancer therapies, some of them are not used for the cancer types analyzed here; some drug-target driver nodes identified by our algorithms are not known to be used in any cancer therapy. Overall we show that a better understanding of the control dynamics of cancer through computational modelling can pave the way for new efficient therapeutic approaches and personalized medicine.
Collapse
Affiliation(s)
- Krishna Kanhaiya
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland
| | - Eugen Czeizler
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland
- National Institute for Research and Development for Biological Sciences, Bucharest, Romania
| | - Cristian Gratie
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland
| | - Ion Petre
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland.
| |
Collapse
|
16
|
Li L, Wang X, Xiao G, Gazdar A. Integrative gene set enrichment analysis utilizing isoform-specific expression. Genet Epidemiol 2017; 41:498-510. [PMID: 28580727 DOI: 10.1002/gepi.22052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 02/12/2017] [Accepted: 03/14/2017] [Indexed: 01/01/2023]
Abstract
Gene set enrichment analysis (GSEA) aims at identifying essential pathways, or more generally, sets of biologically related genes that are involved in complex human diseases. In the past, many studies have shown that GSEA is a very useful bioinformatics tool that plays critical roles in the innovation of disease prevention and intervention strategies. Despite its tremendous success, it is striking that conclusions of GSEA drawn from isolated studies are often sparse, and different studies may lead to inconsistent and sometimes contradictory results. Further, in the wake of next generation sequencing technologies, it has been made possible to measure genome-wide isoform-specific expression levels, calling for innovations that can utilize the unprecedented resolution. Currently, enormous amounts of data have been created from various RNA-seq experiments. All these give rise to a pressing need for developing integrative methods that allow for explicit utilization of isoform-specific expression, to combine multiple enrichment studies, in order to enhance the power, reproducibility, and interpretability of the analysis. We develop and evaluate integrative GSEA methods, based on two-stage procedures, which, for the first time, allow statistically efficient use of isoform-specific expression from multiple RNA-seq experiments. Through simulation and real data analysis, we show that our methods can greatly improve the performance in identifying essential gene sets compared to existing methods that can only use gene-level expression.
Collapse
Affiliation(s)
- Lie Li
- Department of Statistical Science, Southern Methodist University, Dallas, Texas, United States of America
| | - Xinlei Wang
- Department of Statistical Science, Southern Methodist University, Dallas, Texas, United States of America
| | - Guanghua Xiao
- Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Adi Gazdar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
17
|
Wang K, Shan Z, Duan L, Gong T, Liu F, Zhang Y, Wang Z, Shen J, Lei L. iTRAQ-based quantitative proteomic analysis of Yamanaka factors reprogrammed breast cancer cells. Oncotarget 2017; 8:34330-34339. [PMID: 28423718 PMCID: PMC5470971 DOI: 10.18632/oncotarget.16125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/24/2017] [Indexed: 12/17/2022] Open
Abstract
Cancer cells had been developed to be reprogrammed into embryonic stem like cells by induced pluripotent stem cells (iPSCs) technology, however, the tumor differentiation/dedifferentiation mechanisms had not yet been analyzed on a genome-wide scale. Here, we inserted the four stem cell transcription factor genes OCT4, SOX2, C-MYC and KLF4 into MCF cells (MCFs), represented a female breast cancer cell type, and obtained iPSCs (Mcfips) in about 3 weeks. By using the LC MS/MS iTRAQ technology, we analyzed the proteomic changes between MCFs and Mcfips. Of identified 4,616 proteins totally, 247 and 142 differentially expressed (DE) proteins were found in Mcfips compared with human induce pluripotent stem cells (Hips) and MCFs, respectively. 35 co-up and 10 co-down regulated proteins were recognized in DE proteins. Above DE proteins were categorized with GO functional classification annotation and KEGG metabolic pathway analysis into biological processes. In the protein interaction network, we found 37 and 39 hubs interacted with more than one protein in Mcfips comparing to Hips, in addition, 25 and 9 hubs were identified in Mcfips comparing to MCFs. Importantly, the mitochondria, ribosome and tumor suppressor proteins were found to be core regulators of tumor reprogramming, which might contribute to understand the mechanisms in relation to the occurrences and progression of a tumor. Thus, our study provided a valuable data for exploring the possibility to normalize the malignant phenotype.
Collapse
Affiliation(s)
- Kun Wang
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
- Embryo and Stem Cell Engineering Laboratory, Harbin Medical University, Harbin, China
| | - Lian Duan
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Tiantian Gong
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Feng Liu
- Department of Breast Surgery, Cancer Hospital of Harbin Medical University, Harbin, China
| | - Yue Zhang
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Zhendong Wang
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
- Embryo and Stem Cell Engineering Laboratory, Harbin Medical University, Harbin, China
| | - Jingling Shen
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
- Embryo and Stem Cell Engineering Laboratory, Harbin Medical University, Harbin, China
| | - Lei Lei
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
- Embryo and Stem Cell Engineering Laboratory, Harbin Medical University, Harbin, China
| |
Collapse
|
18
|
MD Aksam V, Chandrasekaran V, Pandurangan S. Hub nodes in the network of human Mitogen-Activated Protein Kinase (MAPK) pathways: Characteristics and potential as drug targets. INFORMATICS IN MEDICINE UNLOCKED 2017. [DOI: 10.1016/j.imu.2017.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
19
|
Dissecting the regulation rules of cancer-related miRNAs based on network analysis. Sci Rep 2016; 6:34172. [PMID: 27694936 PMCID: PMC5046108 DOI: 10.1038/srep34172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/06/2016] [Indexed: 01/04/2023] Open
Abstract
miRNAs (microRNAs) are a set of endogenous and small non-coding RNAs which specifically induce degradation of target mRNAs or inhibit protein translation to control gene expression. Obviously, aberrant miRNA expression in human cells will lead to a serious of changes in protein-protein interaction network (PPIN), thus to activate or inactivate some pathways related to various diseases, especially carcinogenesis. In this study, we systematically constructed the miRNA-regulated co-expressed protein-protein interaction network (CePPIN) for 17 cancers firstly. We investigated the topological parameters and functional annotation for the proteins in CePPIN, especially for those miRNA targets. We found that targets regulated by more miRNAs tend to play a more important role in the forming process of cancers. We further elucidated the miRNA regulation rules in PPIN from a more systematical perspective. By GO and KEGG pathway analysis, miRNA targets are involved in various cellular processes mostly related to cell cycle, such as cell proliferation, growth, differentiation, etc. Through the Pfam classification, we found that miRNAs belonging to the same family tend to have targets from the same family which displays the synergistic function of these miRNAs. Finally, the case study on miR-519d and miR-21-regulated sub-network was performed to support our findings.
Collapse
|
20
|
Maron BA, Leopold JA. Systems biology: An emerging strategy for discovering novel pathogenetic mechanisms that promote cardiovascular disease. Glob Cardiol Sci Pract 2016; 2016:e201627. [PMID: 29043273 PMCID: PMC5642838 DOI: 10.21542/gcsp.2016.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Reductionist theory proposes that analyzing complex systems according to their most fundamental components is required for problem resolution, and has served as the cornerstone of scientific methodology for more than four centuries. However, technological gains in the current scientific era now allow for the generation of large datasets that profile the proteomic, genomic, and metabolomic signatures of biological systems across a range of conditions. The accessibility of data on such a vast scale has, in turn, highlighted the limitations of reductionism, which is not conducive to analyses that consider multiple and contemporaneous interactions between intermediates within a pathway or across constructs. Systems biology has emerged as an alternative approach to analyze complex biological systems. This methodology is based on the generation of scale-free networks and, thus, provides a quantitative assessment of relationships between multiple intermediates, such as protein-protein interactions, within and between pathways of interest. In this way, systems biology is well positioned to identify novel targets implicated in the pathogenesis or treatment of diseases. In this review, the historical root and fundamental basis of systems biology, as well as the potential applications of this methodology are discussed with particular emphasis on integration of these concepts to further understanding of cardiovascular disorders such as coronary artery disease and pulmonary hypertension.
Collapse
Affiliation(s)
- Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Cardiology, Boston VA Healthcare System, Boston, MA, USA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Rai S, Bhatnagar S. Hyperlipidemia, Disease Associations, and Top 10 Potential Drug Targets: A Network View. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:152-68. [DOI: 10.1089/omi.2015.0172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sneha Rai
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, India
| |
Collapse
|
22
|
Liu J, Wang Z. Diverse array-designed modes of combination therapies in Fangjiomics. Acta Pharmacol Sin 2015; 36:680-8. [PMID: 25864646 PMCID: PMC4594182 DOI: 10.1038/aps.2014.125] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 10/30/2014] [Indexed: 12/11/2022]
Abstract
In line with the complexity of disease networks, diverse combination therapies have been demonstrated potential in the treatment of different patients with complex diseases in a personal combination profile. However, the identification of rational, compatible and effective drug combinations remains an ongoing challenge. Based on a holistic theory integrated with reductionism, Fangjiomics systematically develops multiple modes of array-designed combination therapies. We define diverse "magic shotgun" vertical, horizontal, focusing, siege and dynamic arrays according to different spatiotemporal distributions of hits on targets, pathways and networks. Through these multiple adaptive modes for treating complex diseases, Fangjiomics may help to identify rational drug combinations with synergistic or additive efficacy but reduced adverse side effects that reverse complex diseases by reconstructing or rewiring multiple targets, pathways and networks. Such a novel paradigm for combination therapies may allow us to achieve more precise treatments by developing phenotype-driven quantitative multi-scale modeling for rational drug combinations.
Collapse
Affiliation(s)
- Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
23
|
Wang RS, Maron BA, Loscalzo J. Systems medicine: evolution of systems biology from bench to bedside. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:141-61. [PMID: 25891169 DOI: 10.1002/wsbm.1297] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 12/11/2022]
Abstract
High-throughput experimental techniques for generating genomes, transcriptomes, proteomes, metabolomes, and interactomes have provided unprecedented opportunities to interrogate biological systems and human diseases on a global level. Systems biology integrates the mass of heterogeneous high-throughput data and predictive computational modeling to understand biological functions as system-level properties. Most human diseases are biological states caused by multiple components of perturbed pathways and regulatory networks rather than individual failing components. Systems biology not only facilitates basic biological research but also provides new avenues through which to understand human diseases, identify diagnostic biomarkers, and develop disease treatments. At the same time, systems biology seeks to assist in drug discovery, drug optimization, drug combinations, and drug repositioning by investigating the molecular mechanisms of action of drugs at a system's level. Indeed, systems biology is evolving to systems medicine as a new discipline that aims to offer new approaches for addressing the diagnosis and treatment of major human diseases uniquely, effectively, and with personalized precision.
Collapse
Affiliation(s)
- Rui-Sheng Wang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Cardiology, Veterans Affairs Boston Healthcare System, West Roxbury, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Bell IR, Schwartz GE. Enhancement of adaptive biological effects by nanotechnology preparation methods in homeopathic medicines. HOMEOPATHY 2015; 104:123-38. [DOI: 10.1016/j.homp.2014.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 11/16/2014] [Indexed: 01/19/2023]
|
25
|
Zhang J, Wang Y, Shang D, Yu F, Liu W, Zhang Y, Feng C, Wang Q, Xu Y, Liu Y, Bai X, Li X, Li C. Characterizing and optimizing human anticancer drug targets based on topological properties in the context of biological pathways. J Biomed Inform 2015; 54:132-40. [PMID: 25724580 DOI: 10.1016/j.jbi.2015.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 01/14/2023]
Abstract
One of the challenging problems in drug discovery is to identify the novel targets for drugs. Most of the traditional methods for drug targets optimization focused on identifying the particular families of "druggable targets", but ignored their topological properties based on the biological pathways. In this study, we characterized the topological properties of human anticancer drug targets (ADTs) in the context of biological pathways. We found that the ADTs tended to present the following seven topological properties: influence the number of the pathways related to cancer, be localized at the start or end of the pathways, interact with cancer related genes, exhibit higher connectivity, vulnerability, betweenness, and closeness than other genes. We first ranked ADTs based on their topological property values respectively, then fused them into one global-rank using the joint cumulative distribution of an N-dimensional order statistic to optimize human ADTs. We applied the optimization method to 13 anticancer drugs, respectively. Results demonstrated that over 70% of known ADTs were ranked in the top 20%. Furthermore, the performance for mercaptopurine was significant: 6 known targets (ADSL, GMPR2, GMPR, HPRT1, AMPD3, AMPD2) were ranked in the top 15 and other four out of the top 15 (MAT2A, CDKN1A, AREG, JUN) have the potentialities to become new targets for cancer therapy.
Collapse
Affiliation(s)
- Jian Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, PR China
| | - Yan Wang
- Majorbio Bio-Pharm Technology Co., Ltd., Shanghai 201203, PR China
| | - Desi Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Fulong Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Wei Liu
- Department of Mathematics, Heilongjiang Institute of Technology, Harbin 150050, PR China
| | - Yan Zhang
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, PR China
| | - Chenchen Feng
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, PR China
| | - Qiuyu Wang
- School of Nursing, Daqing Campus, Harbin Medical University, Daqing 163319, PR China
| | - Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
| | - Yuejuan Liu
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, PR China
| | - Xuefeng Bai
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, PR China
| | - Xuecang Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, PR China
| | - Chunquan Li
- School of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing 163319, PR China.
| |
Collapse
|
26
|
Cui T, He ZG. Improved understanding of pathogenesis from protein interactions inMycobacteriumtuberculosis. Expert Rev Proteomics 2014; 11:745-55. [DOI: 10.1586/14789450.2014.971762] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Katsogiannou M, Andrieu C, Baylot V, Baudot A, Dusetti NJ, Gayet O, Finetti P, Garrido C, Birnbaum D, Bertucci F, Brun C, Rocchi P. The functional landscape of Hsp27 reveals new cellular processes such as DNA repair and alternative splicing and proposes novel anticancer targets. Mol Cell Proteomics 2014; 13:3585-601. [PMID: 25277244 PMCID: PMC4256507 DOI: 10.1074/mcp.m114.041228] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously, we identified the stress-induced chaperone, Hsp27, as highly overexpressed in castration-resistant prostate cancer and developed an Hsp27 inhibitor (OGX-427) currently tested in phase I/II clinical trials as a chemosensitizing agent in different cancers. To better understand the Hsp27 poorly-defined cytoprotective functions in cancers and increase the OGX-427 pharmacological safety, we established the Hsp27-protein interaction network using a yeast two-hybrid approach and identified 226 interaction partners. As an example, we showed that targeting Hsp27 interaction with TCTP, a partner protein identified in our screen increases therapy sensitivity, opening a new promising field of research for therapeutic approaches that could decrease or abolish toxicity for normal cells. Results of an in-depth bioinformatics network analysis allying the Hsp27 interaction map into the human interactome underlined the multifunctional character of this protein. We identified interactions of Hsp27 with proteins involved in eight well known functions previously related to Hsp27 and uncovered 17 potential new ones, such as DNA repair and RNA splicing. Validation of Hsp27 involvement in both processes in human prostate cancer cells supports our system biology-predicted functions and provides new insights into Hsp27 roles in cancer cells.
Collapse
Affiliation(s)
- Maria Katsogiannou
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Claudia Andrieu
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Virginie Baylot
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Anaïs Baudot
- ¶Aix-Marseille Université, F-13284, Marseille, France; **Institut de Mathématiques de Marseille, CNRS UMR7373, Marseille, F-13009, France
| | - Nelson J Dusetti
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Odile Gayet
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Pascal Finetti
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France
| | - Carmen Garrido
- ‡‡Inserm U866, Faculty of Medicine, 21000 Dijon, France; §§CGFL Dijon, France
| | - Daniel Birnbaum
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - François Bertucci
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France
| | - Christine Brun
- ¶Aix-Marseille Université, F-13284, Marseille, France; ¶¶TAGC Inserm U1090, Marseille, F-13009, France; ‖‖CNRS, France
| | - Palma Rocchi
- From the ‡Inserm, UMR1068, CRCM, Marseille, F-13009, France; §Institut Paoli-Calmettes, Marseille, F-13009, France; ¶Aix-Marseille Université, F-13284, Marseille, France; ‖CNRS, UMR7258, CRCM, Marseille, F-13009, France;
| |
Collapse
|
28
|
Anand P, Chandra N. Characterizing the pocketome of Mycobacterium tuberculosis and application in rationalizing polypharmacological target selection. Sci Rep 2014; 4:6356. [PMID: 25220818 PMCID: PMC5376175 DOI: 10.1038/srep06356] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 06/20/2014] [Indexed: 01/13/2023] Open
Abstract
Polypharmacology is beginning to emerge as an important concept in the field of drug discovery. However, there are no established approaches to either select appropriate target sets or design polypharmacological drugs. Here, we propose a structural-proteomics approach that utilizes the structural information of the binding sites at a genome-scale obtained through in-house algorithms to characterize the pocketome, yielding a list of ligands that can participate in various biochemical events in the mycobacterial cell. The pocket-type space is seen to be much larger than the sequence or fold-space, suggesting that variations at the site-level contribute significantly to functional repertoire of the organism. All-pair comparisons of binding sites within Mycobacterium tuberculosis (Mtb), pocket-similarity network construction and clustering result in identification of binding-site sets, each containing a group of similar binding sites, theoretically having a potential to interact with a common set of compounds. A polypharmacology index is formulated to rank targets by incorporating a measure of druggability and similarity to other pockets within the proteome. This study presents a rational approach to identify targets with polypharmacological potential along with possible drugs for repurposing, while simultaneously, obtaining clues on lead compounds for use in new drug-discovery pipelines.
Collapse
Affiliation(s)
- Praveen Anand
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| | - Nagasuma Chandra
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| |
Collapse
|
29
|
Wright B, Tindall MJ, Lovegrove JA, Gibbins JM. Investigating flavonoids as molecular templates for the design of small-molecule inhibitors of cell signaling. J Food Sci 2014; 78:N1921-8. [PMID: 24329957 DOI: 10.1111/1750-3841.12293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/20/2013] [Indexed: 01/03/2023]
Abstract
Epidemiological and clinical trials reveal compelling evidence for the ability of dietary flavonoids to lower cardiovascular disease risk. The mechanisms of action of these polyphenolic compounds are diverse, and of particular interest is their ability to function as protein and lipid kinase inhibitors. We have previously described structure-activity studies that reinforce the possibility for using flavonoid structures as templates for drug design. In the present study, we aim to begin constructing rational screening strategies for exploiting these compounds as templates for the design of clinically relevant, antiplatelet agents. We used the platelet as a model system to dissect the structural influence of flavonoids, stilbenes, anthocyanidins, and phenolic acids on inhibition of cell signaling and function. Functional groups identified as relevant for potent inhibition of platelet function included at least 2 benzene rings, a hydroxylated B ring, a planar C ring, a C ring ketone group, and a C-2 positioned B ring. Hydroxylation of the B ring with either a catechol group or a single C-4' hydroxyl may be required for efficient inhibition of collagen-stimulated tyrosine phosphorylated proteins of 125 to 130 kDa, but may not be necessary for that of phosphotyrosine proteins at approximately 29 kDa. The removal of the C ring C-3 hydroxyl together with a hydroxylated B ring (apigenin) may confer selectivity for 37 to 38 kDa phosphotyrosine proteins. We conclude that this study may form the basis for construction of maps of flavonoid inhibitory activity on kinase targets that may allow a multitargeted therapeutic approach with analogue counterparts and parent compounds.
Collapse
Affiliation(s)
- Bernice Wright
- Inst. for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, Univ. of Reading, Reading, RG6 6UB Berkshire, U.K
| | | | | | | |
Collapse
|
30
|
Chen Y, Wang Z, Wang Y. Spatiotemporal positioning of multipotent modules in diverse biological networks. Cell Mol Life Sci 2014; 71:2605-24. [PMID: 24413666 PMCID: PMC11113103 DOI: 10.1007/s00018-013-1547-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 12/05/2013] [Accepted: 12/19/2013] [Indexed: 02/06/2023]
Abstract
A biological network exhibits a modular organization. The modular structure dependent on functional module is of great significance in understanding the organization and dynamics of network functions. A huge variety of module identification methods as well as approaches to analyze modularity and dynamics of the inter- and intra-module interactions have emerged recently, but they are facing unexpected challenges in further practical applications. Here, we discuss recent progress in understanding how such a modular network can be deconstructed spatiotemporally. We focus particularly on elucidating how various deciphering mechanisms operate to ensure precise module identification and assembly. In this case, a system-level understanding of the entire mechanism of module construction is within reach, with important implications for reasonable perspectives in both constructing a modular analysis framework and deconstructing different modular hierarchical structures.
Collapse
Affiliation(s)
- Yinying Chen
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing, 100700 China
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing, 100700 China
| | - Yongyan Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing, 100700 China
| |
Collapse
|
31
|
Bell IR. Nonlinear effects of nanoparticles: biological variability from hormetic doses, small particle sizes, and dynamic adaptive interactions. Dose Response 2014; 12:202-32. [PMID: 24910581 PMCID: PMC4036395 DOI: 10.2203/dose-response.13-025.bell] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Researchers are increasingly focused on the nanoscale level of organization where biological processes take place in living systems. Nanoparticles (NPs, e.g., 1-100 nm diameter) are small forms of natural or manufactured source material whose properties differ markedly from those of the respective bulk forms of the "same" material. Certain NPs have diagnostic and therapeutic uses; some NPs exhibit low-dose toxicity; other NPs show ability to stimulate low-dose adaptive responses (hormesis). Beyond dose, size, shape, and surface charge variations of NPs evoke nonlinear responses in complex adaptive systems. NPs acquire unique size-dependent biological, chemical, thermal, optical, electromagnetic, and atom-like quantum properties. Nanoparticles exhibit high surface adsorptive capacity for other substances, enhanced bioavailability, and ability to cross otherwise impermeable cell membranes including the blood-brain barrier. With super-potent effects, nano-forms can evoke cellular stress responses or therapeutic effects not only at lower doses than their bulk forms, but also for longer periods of time. Interactions of initial effects and compensatory systemic responses can alter the impact of NPs over time. Taken together, the data suggest the need to downshift the dose-response curve of NPs from that for bulk forms in order to identify the necessarily decreased no-observed-adverse-effect-level and hormetic dose range for nanoparticles.
Collapse
|
32
|
Characterizing and controlling the inflammatory network during influenza A virus infection. Sci Rep 2014; 4:3799. [PMID: 24445954 PMCID: PMC3896911 DOI: 10.1038/srep03799] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 12/31/2013] [Indexed: 12/20/2022] Open
Abstract
To gain insights into the pathogenesis of influenza A virus (IAV) infections, this study focused on characterizing the inflammatory network and identifying key proteins by combining high-throughput data and computational techniques. We constructed the cell-specific normal and inflammatory networks for H5N1 and H1N1 infections through integrating high-throughput data. We demonstrated that better discrimination between normal and inflammatory networks by network entropy than by other topological metrics. Moreover, we identified different dynamical interactions among TLR2, IL-1β, IL10 and NFκB between normal and inflammatory networks using optimization algorithm. In particular, good robustness and multistability of inflammatory sub-networks were discovered. Furthermore, we identified a complex, TNFSF10/HDAC4/HDAC5, which may play important roles in controlling inflammation, and demonstrated that changes in network entropy of this complex negatively correlated to those of three proteins: TNFα, NFκB and COX-2. These findings provide significant hypotheses for further exploring the molecular mechanisms of infectious diseases and developing control strategies.
Collapse
|
33
|
Ma CW, Xiu ZL, Zeng AP. Exploring signal transduction in heteromultimeric protein based on energy dissipation model. J Biomol Struct Dyn 2013; 33:134-46. [PMID: 24279729 DOI: 10.1080/07391102.2013.855145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Dynamic intersubunit interactions are key elements in the regulation of many biological systems. A better understanding of how subunits interact with each other and how their interactions are related to dynamic protein structure is a fundamental task in biology. In this paper, a heteromultimeric allosteric protein, Corynebacterium glutamicum aspartokinase, is used as a model system to explore the signal transduction involved in intersubunit interactions and allosteric communication with an emphasis on the intersubunit signaling process. For this purpose, energy dissipation simulation and network construction are conducted for each subunit and the whole protein. Comparison with experimental results shows that the new approach is able to predict all the mutation sites that have been experimentally proved to desensitize allosteric regulation of the enzyme. Additionally, analysis revealed that the function of the effector threonine is to facilitate the binding of the two subunits without contributing to the allosteric communication. During the allosteric regulation upon the binding of the effector lysine, signals can be transferred from the β-subunit to the catalytic site of the α-subunit through both a direct way of intersubunit signal transduction, and an indirect way: first, to the regulatory region of the α-subunit by intersubunit signal transduction and then to the catalytic region by intramolecular signal transduction. Therefore, the new approach is able to illustrate the diversity of the underlying mechanisms when the strength of feedback inhibition by the effector(s) is modulated, providing useful information that has potential applications in engineering heteromultimeric allosteric regulation.
Collapse
Affiliation(s)
- Cheng-Wei Ma
- a Institute of Bioprocess and Biosystems Engineering , Hamburg University of Technology , Hamburg D-21073 , Germany
| | | | | |
Collapse
|
34
|
Szalay KZ, Csermely P. Perturbation centrality and turbine: a novel centrality measure obtained using a versatile network dynamics tool. PLoS One 2013; 8:e78059. [PMID: 24205090 PMCID: PMC3804472 DOI: 10.1371/journal.pone.0078059] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 09/16/2013] [Indexed: 11/19/2022] Open
Abstract
Analysis of network dynamics became a focal point to understand and predict changes of complex systems. Here we introduce Turbine, a generic framework enabling fast simulation of any algorithmically definable dynamics on very large networks. Using a perturbation transmission model inspired by communicating vessels, we define a novel centrality measure: perturbation centrality. Hubs and inter-modular nodes proved to be highly efficient in perturbation propagation. High perturbation centrality nodes of the Met-tRNA synthetase protein structure network were identified as amino acids involved in intra-protein communication by earlier studies. Changes in perturbation centralities of yeast interactome nodes upon various stresses well recapitulated the functional changes of stressed yeast cells. The novelty and usefulness of perturbation centrality was validated in several other model, biological and social networks. The Turbine software and the perturbation centrality measure may provide a large variety of novel options to assess signaling, drug action, environmental and social interventions.
Collapse
Affiliation(s)
- Kristóf Z. Szalay
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Peter Csermely
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
35
|
Alonso N, Caamaño O, Romero-Duran FJ, Luan F, D. S. Cordeiro MN, Yañez M, González-Díaz H, García-Mera X. Model for high-throughput screening of multitarget drugs in chemical neurosciences: synthesis, assay, and theoretic study of rasagiline carbamates. ACS Chem Neurosci 2013; 4:1393-403. [PMID: 23855599 DOI: 10.1021/cn400111n] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The disappointing results obtained in recent clinical trials renew the interest in experimental/computational techniques for the discovery of neuroprotective drugs. In this context, multitarget or multiplexing QSAR models (mt-QSAR/mx-QSAR) may help to predict neurotoxicity/neuroprotective effects of drugs in multiple assays, on drug targets, and in model organisms. In this work, we study a data set downloaded from CHEMBL; each data point (>8000) contains the values of one out of 37 possible measures of activity, 493 assays, 169 molecular or cellular targets, and 11 different organisms (including human) for a given compound. In this work, we introduce the first mx-QSAR model for neurotoxicity/neuroprotective effects of drugs based on the MARCH-INSIDE (MI) method. First, we used MI to calculate the stochastic spectral moments (structural descriptors) of all compounds. Next, we found a model that classified correctly 2955 out of 3548 total cases in the training and validation series with Accuracy, Sensitivity, and Specificity values>80%. The model also showed excellent results in Computational-Chemistry simulations of High-Throughput Screening (CCHTS) experiments, with accuracy=90.6% for 4671 positive cases. Next, we reported the synthesis, characterization, and experimental assays of new rasagiline derivatives. We carried out three different experimental tests: assay (1) in the absence of neurotoxic agents, assay (2) in the presence of glutamate, and assay (3) in the presence of H2O2. Compounds 11 with 27.4%, 8 with 11.6%, and 9 with 15.4% showed the highest neuroprotective effects in assays (1), (2), and (3), respectively. After that, we used the mx-QSAR model to carry out a CCHTS of the new compounds in >400 unique pharmacological tests not carried out experimentally. Consequently, this model may become a promising auxiliary tool for the discovery of new drugs for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nerea Alonso
- Department of Organic Chemistry,
Faculty of Pharmacy, University of Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
| | - Olga Caamaño
- Department of Organic Chemistry,
Faculty of Pharmacy, University of Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
| | - Francisco J. Romero-Duran
- Department of Organic Chemistry,
Faculty of Pharmacy, University of Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
| | - Feng Luan
- REQUIMTE/Department of Chemistry
and Biochemistry, University of Porto,
4169-007, Porto, Portugal
- Department of Applied Chemistry, Yantai University, Yantai 264005, People’s Republic
of China
| | | | - Matilde Yañez
- Department of
Pharmacology,
Faculty of Pharmacy, USC, 15782, Santiago
de Compostela, Spain
| | - Humberto González-Díaz
- Departament
of Organic Chemistry
II, University of the Basque Country UPV/EHU, 48940, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| | - Xerardo García-Mera
- Department of Organic Chemistry,
Faculty of Pharmacy, University of Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
| |
Collapse
|
36
|
Bellavite P, Olioso D, Marzotto M, Moratti E, Conforti A. A dynamic network model of the similia principle. Complement Ther Med 2013; 21:750-61. [PMID: 24280484 DOI: 10.1016/j.ctim.2013.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 08/29/2013] [Accepted: 09/01/2013] [Indexed: 01/30/2023] Open
Abstract
The use of drugs in high dilutions and the principle of similarity (or "similia") are two basic tenets of homeopathy. However, the plausibility of both is a subject of debate. Although several models have been proposed to explain the similia principle, it can be best understood and appreciated in the framework of complexity science and dynamic systems theory. This work applies a five-node Boolean network to show how self-organization and adaptation are relevant to rationalizing this traditional medical principle. Simulating the trajectories and attractors of the network system in the energy state-space provides a rudimentary and qualitative illustration of how targeted external perturbations can have pathological effects, leading to permanent, self-sustaining alterations. Similarly, changes that conversely enable the system to find its way back to the original state can induce therapeutic effects, by causing specific shifts in attractors when suitable conditions are satisfied. Extrapolating these mechanisms to homeopathy, we can envisage how major changes in the evolution of homeodynamic systems (and, eventually, healing of the entire body) can be achieved through carefully selected remedies that reproduce the whole symptom pattern of the ill state.
Collapse
Affiliation(s)
- Paolo Bellavite
- Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy.
| | | | | | | | | |
Collapse
|
37
|
Nodes having a major influence to break cooperation define a novel centrality measure: game centrality. PLoS One 2013; 8:e67159. [PMID: 23840611 PMCID: PMC3696096 DOI: 10.1371/journal.pone.0067159] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 05/15/2013] [Indexed: 11/25/2022] Open
Abstract
Cooperation played a significant role in the self-organization and evolution of living organisms. Both network topology and the initial position of cooperators heavily affect the cooperation of social dilemma games. We developed a novel simulation program package, called ‘NetworGame’, which is able to simulate any type of social dilemma games on any model, or real world networks with any assignment of initial cooperation or defection strategies to network nodes. The ability of initially defecting single nodes to break overall cooperation was called as ‘game centrality’. The efficiency of this measure was verified on well-known social networks, and was extended to ‘protein games’, i.e. the simulation of cooperation between proteins, or their amino acids. Hubs and in particular, party hubs of yeast protein-protein interaction networks had a large influence to convert the cooperation of other nodes to defection. Simulations on methionyl-tRNA synthetase protein structure network indicated an increased influence of nodes belonging to intra-protein signaling pathways on breaking cooperation. The efficiency of single, initially defecting nodes to convert the cooperation of other nodes to defection in social dilemma games may be an important measure to predict the importance of nodes in the integration and regulation of complex systems. Game centrality may help to design more efficient interventions to cellular networks (in forms of drugs), to ecosystems and social networks.
Collapse
|
38
|
Kubisch J, Türei D, Földvári-Nagy L, Dunai ZA, Zsákai L, Varga M, Vellai T, Csermely P, Korcsmáros T. Complex regulation of autophagy in cancer - integrated approaches to discover the networks that hold a double-edged sword. Semin Cancer Biol 2013; 23:252-61. [PMID: 23810837 DOI: 10.1016/j.semcancer.2013.06.009] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Autophagy, a highly regulated self-degradation process of eukaryotic cells, is a context-dependent tumor-suppressing mechanism that can also promote tumor cell survival upon stress and treatment resistance. Because of this ambiguity, autophagy is considered as a double-edged sword in oncology, making anti-cancer therapeutic approaches highly challenging. In this review, we present how systems-level knowledge on autophagy regulation can help to develop new strategies and efficiently select novel anti-cancer drug targets. We focus on the protein interactors and transcriptional/post-transcriptional regulators of autophagy as the protein and regulatory networks significantly influence the activity of core autophagy proteins during tumor progression. We list several network resources to identify interactors and regulators of autophagy proteins. As in silico analysis of such networks often necessitates experimental validation, we briefly summarize tractable model organisms to examine the role of autophagy in cancer. We also discuss fluorescence techniques for high-throughput monitoring of autophagy in humans. Finally, the challenges of pharmacological modulation of autophagy are reviewed. We suggest network-based concepts to overcome these difficulties. We point out that a context-dependent modulation of autophagy would be favored in anti-cancer therapy, where autophagy is stimulated in normal cells, while inhibited only in stressed cancer cells. To achieve this goal, we introduce the concept of regulo-network drugs targeting specific transcription factors or miRNA families identified with network analysis. The effect of regulo-network drugs propagates indirectly through transcriptional or post-transcriptional regulation of autophagy proteins, and, as a multi-directional intervention tool, they can both activate and inhibit specific proteins in the same time. The future identification and validation of such regulo-network drug targets may serve as novel intervention points, where autophagy can be effectively modulated in cancer therapy.
Collapse
Affiliation(s)
- János Kubisch
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117 Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Csermely P, Korcsmáros T, Kiss HJM, London G, Nussinov R. Structure and dynamics of molecular networks: a novel paradigm of drug discovery: a comprehensive review. Pharmacol Ther 2013; 138:333-408. [PMID: 23384594 PMCID: PMC3647006 DOI: 10.1016/j.pharmthera.2013.01.016] [Citation(s) in RCA: 521] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 01/22/2013] [Indexed: 02/02/2023]
Abstract
Despite considerable progress in genome- and proteome-based high-throughput screening methods and in rational drug design, the increase in approved drugs in the past decade did not match the increase of drug development costs. Network description and analysis not only give a systems-level understanding of drug action and disease complexity, but can also help to improve the efficiency of drug design. We give a comprehensive assessment of the analytical tools of network topology and dynamics. The state-of-the-art use of chemical similarity, protein structure, protein-protein interaction, signaling, genetic interaction and metabolic networks in the discovery of drug targets is summarized. We propose that network targeting follows two basic strategies. The "central hit strategy" selectively targets central nodes/edges of the flexible networks of infectious agents or cancer cells to kill them. The "network influence strategy" works against other diseases, where an efficient reconfiguration of rigid networks needs to be achieved by targeting the neighbors of central nodes/edges. It is shown how network techniques can help in the identification of single-target, edgetic, multi-target and allo-network drug target candidates. We review the recent boom in network methods helping hit identification, lead selection optimizing drug efficacy, as well as minimizing side-effects and drug toxicity. Successful network-based drug development strategies are shown through the examples of infections, cancer, metabolic diseases, neurodegenerative diseases and aging. Summarizing >1200 references we suggest an optimized protocol of network-aided drug development, and provide a list of systems-level hallmarks of drug quality. Finally, we highlight network-related drug development trends helping to achieve these hallmarks by a cohesive, global approach.
Collapse
Affiliation(s)
- Peter Csermely
- Department of Medical Chemistry, Semmelweis University, P.O. Box 260, H-1444 Budapest 8, Hungary.
| | | | | | | | | |
Collapse
|
40
|
Network pharmacology: a new approach for chinese herbal medicine research. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:621423. [PMID: 23762149 PMCID: PMC3671675 DOI: 10.1155/2013/621423] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 03/28/2013] [Accepted: 05/02/2013] [Indexed: 12/29/2022]
Abstract
The dominant paradigm of "one gene, one target, one disease" has influenced many aspects of drug discovery strategy. However, in recent years, it has been appreciated that many effective drugs act on multiple targets rather than a single one. As an integrated multidisciplinary concept, network pharmacology, which is based on system biology and polypharmacology, affords a novel network mode of "multiple targets, multiple effects, complex diseases" and replaces the "magic bullets" by "magic shotguns." Chinese herbal medicine (CHM) has been recognized as one of the most important strategies in complementary and alternative medicine. Though CHM has been practiced for a very long time, its effectiveness and beneficial contribution to public health has not been fully recognized. Also, the knowledge on the mechanisms of CHM formulas is scarce. In the present review, the concept and significance of network pharmacology is briefly introduced. The application and potential role of network pharmacology in the CHM fields is also discussed, such as data collection, target prediction, network visualization, multicomponent interaction, and network toxicology. Furthermore, the developing tendency of network pharmacology is also summarized, and its role in CHM research is discussed.
Collapse
|
41
|
Balogh G, Péter M, Glatz A, Gombos I, Török Z, Horváth I, Harwood JL, Vígh L. Key role of lipids in heat stress management. FEBS Lett 2013; 587:1970-80. [PMID: 23684645 DOI: 10.1016/j.febslet.2013.05.016] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022]
Abstract
Heat stress is a common and, therefore, an important environmental impact on cells and organisms. While much attention has been paid to severe heat stress, moderate temperature elevations are also important. Here we discuss temperature sensing and how responses to heat stress are not necessarily dependent on denatured proteins. Indeed, it is clear that membrane lipids have a pivotal function. Details of membrane lipid changes and the associated production of signalling metabolites are described and suggestions made as to how the interconnected signalling network could be modified for helpful intervention in disease.
Collapse
Affiliation(s)
- Gábor Balogh
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, H-6701 Szeged, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wu M, Yu Q, Li X, Zheng J, Huang JF, Kwoh CK. Benchmarking human protein complexes to investigate drug-related systems and evaluate predicted protein complexes. PLoS One 2013; 8:e53197. [PMID: 23405067 PMCID: PMC3566178 DOI: 10.1371/journal.pone.0053197] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 11/29/2012] [Indexed: 11/18/2022] Open
Abstract
Protein complexes are key entities to perform cellular functions. Human diseases are also revealed to associate with some specific human protein complexes. In fact, human protein complexes are widely used for protein function annotation, inference of human protein interactome, disease gene prediction, and so on. Therefore, it is highly desired to build an up-to-date catalogue of human complexes to support the research in these applications. Protein complexes from different databases are as expected to be highly redundant. In this paper, we designed a set of concise operations to compile these redundant human complexes and built a comprehensive catalogue called CHPC2012 (Catalogue of Human Protein Complexes). CHPC2012 achieves a higher coverage for proteins and protein complexes than those individual databases. It is also verified to be a set of complexes with high quality as its co-complex protein associations have a high overlap with protein-protein interactions (PPI) in various existing PPI databases. We demonstrated two distinct applications of CHPC2012, that is, investigating the relationship between protein complexes and drug-related systems and evaluating the quality of predicted protein complexes. In particular, CHPC2012 provides more insights into drug development. For instance, proteins involved in multiple complexes (the overlapping proteins) are potential drug targets; the drug-complex network is utilized to investigate multi-target drugs and drug-drug interactions; and the disease-specific complex-drug networks will provide new clues for drug repositioning. With this up-to-date reference set of human protein complexes, we believe that the CHPC2012 catalogue is able to enhance the studies for protein interactions, protein functions, human diseases, drugs, and related fields of research. CHPC2012 complexes can be downloaded from http://www1.i2r.a-star.edu.sg/xlli/CHPC2012/CHPC2012.htm.
Collapse
Affiliation(s)
- Min Wu
- School of Computer Engineering, Nanyang Technological University, Singapore, Singapore
| | - Qi Yu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Xiaoli Li
- Data Mining Department, Institute for Infocomm Research, Singapore, Singapore
| | - Jie Zheng
- School of Computer Engineering, Nanyang Technological University, Singapore, Singapore
| | - Jing-Fei Huang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | - Chee-Keong Kwoh
- School of Computer Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
43
|
Bánky D, Iván G, Grolmusz V. Equal opportunity for low-degree network nodes: a PageRank-based method for protein target identification in metabolic graphs. PLoS One 2013; 8:e54204. [PMID: 23382878 PMCID: PMC3558500 DOI: 10.1371/journal.pone.0054204] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 12/11/2012] [Indexed: 11/19/2022] Open
Abstract
Biological network data, such as metabolic-, signaling- or physical interaction graphs of proteins are increasingly available in public repositories for important species. Tools for the quantitative analysis of these networks are being developed today. Protein network-based drug target identification methods usually return protein hubs with large degrees in the networks as potentially important targets. Some known, important protein targets, however, are not hubs at all, and perturbing protein hubs in these networks may have several unwanted physiological effects, due to their interaction with numerous partners. Here, we show a novel method applicable in networks with directed edges (such as metabolic networks) that compensates for the low degree (non-hub) vertices in the network, and identifies important nodes, regardless of their hub properties. Our method computes the PageRank for the nodes of the network, and divides the PageRank by the in-degree (i.e., the number of incoming edges) of the node. This quotient is the same in all nodes in an undirected graph (even for large- and low-degree nodes, that is, for hubs and non-hubs as well), but may differ significantly from node to node in directed graphs. We suggest to assign importance to non-hub nodes with large PageRank/in-degree quotient. Consequently, our method gives high scores to nodes with large PageRank, relative to their degrees: therefore non-hub important nodes can easily be identified in large networks. We demonstrate that these relatively high PageRank scores have biological relevance: the method correctly finds numerous already validated drug targets in distinct organisms (Mycobacterium tuberculosis, Plasmodium falciparum and MRSA Staphylococcus aureus), and consequently, it may suggest new possible protein targets as well. Additionally, our scoring method was not chosen arbitrarily: its value for all nodes of all undirected graphs is constant; therefore its high value captures importance in the directed edge structure of the graph.
Collapse
Affiliation(s)
- Dániel Bánky
- Protein Information Technology Group, Eötvös University, Pázmány Péter stny. 1/C, Budapest, Hungary
- Uratim Ltd., Budapest, Hungary
| | - Gábor Iván
- Protein Information Technology Group, Eötvös University, Pázmány Péter stny. 1/C, Budapest, Hungary
- Uratim Ltd., Budapest, Hungary
| | - Vince Grolmusz
- Protein Information Technology Group, Eötvös University, Pázmány Péter stny. 1/C, Budapest, Hungary
- Uratim Ltd., Budapest, Hungary
- * E-mail:
| |
Collapse
|
44
|
Fazekas D, Koltai M, Türei D, Módos D, Pálfy M, Dúl Z, Zsákai L, Szalay-Bekő M, Lenti K, Farkas IJ, Vellai T, Csermely P, Korcsmáros T. SignaLink 2 - a signaling pathway resource with multi-layered regulatory networks. BMC SYSTEMS BIOLOGY 2013; 7:7. [PMID: 23331499 PMCID: PMC3599410 DOI: 10.1186/1752-0509-7-7] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/16/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Signaling networks in eukaryotes are made up of upstream and downstream subnetworks. The upstream subnetwork contains the intertwined network of signaling pathways, while the downstream regulatory part contains transcription factors and their binding sites on the DNA as well as microRNAs and their mRNA targets. Currently, most signaling and regulatory databases contain only a subsection of this network, making comprehensive analyses highly time-consuming and dependent on specific data handling expertise. The need for detailed mapping of signaling systems is also supported by the fact that several drug development failures were caused by undiscovered cross-talk or regulatory effects of drug targets. We previously created a uniformly curated signaling pathway resource, SignaLink, to facilitate the analysis of pathway cross-talks. Here, we present SignaLink 2, which significantly extends the coverage and applications of its predecessor. DESCRIPTION We developed a novel concept to integrate and utilize different subsections (i.e., layers) of the signaling network. The multi-layered (onion-like) database structure is made up of signaling pathways, their pathway regulators (e.g., scaffold and endocytotic proteins) and modifier enzymes (e.g., phosphatases, ubiquitin ligases), as well as transcriptional and post-transcriptional regulators of all of these components. The user-friendly website allows the interactive exploration of how each signaling protein is regulated. The customizable download page enables the analysis of any user-specified part of the signaling network. Compared to other signaling resources, distinctive features of SignaLink 2 are the following: 1) it involves experimental data not only from humans but from two invertebrate model organisms, C. elegans and D. melanogaster; 2) combines manual curation with large-scale datasets; 3) provides confidence scores for each interaction; 4) operates a customizable download page with multiple file formats (e.g., BioPAX, Cytoscape, SBML). Non-profit users can access SignaLink 2 free of charge at http://SignaLink.org. CONCLUSIONS With SignaLink 2 as a single resource, users can effectively analyze signaling pathways, scaffold proteins, modifier enzymes, transcription factors and miRNAs that are important in the regulation of signaling processes. This integrated resource allows the systems-level examination of how cross-talks and signaling flow are regulated, as well as provide data for cross-species comparisons and drug discovery analyses.
Collapse
Affiliation(s)
- Dávid Fazekas
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
| | - Mihály Koltai
- Statistical and Biological Physics Group of the Hungarian Acad. of Sciences, Pázmány P. s. 1A, H-1117, Budapest, Hungary
- Department of Biological Physics, Eötvös Loránd University, Pázmány P. s. 1A, H-1117, Budapest, Hungary
| | - Dénes Türei
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
- Department of Medical Chemistry, Semmelweis University, PO Box 260, H-1444, Budapest, Hungary
| | - Dezső Módos
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
- Department of Medical Chemistry, Semmelweis University, PO Box 260, H-1444, Budapest, Hungary
- Department of Morphology and Physiology, Semmelweis University, Vas u. 17, H-1088, Budapest, Hungary
| | - Máté Pálfy
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
| | - Zoltán Dúl
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
- Department of Medical Chemistry, Semmelweis University, PO Box 260, H-1444, Budapest, Hungary
| | - Lilian Zsákai
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
- Department of Medical Chemistry, Semmelweis University, PO Box 260, H-1444, Budapest, Hungary
| | - Máté Szalay-Bekő
- Department of Medical Chemistry, Semmelweis University, PO Box 260, H-1444, Budapest, Hungary
| | - Katalin Lenti
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
- Department of Morphology and Physiology, Semmelweis University, Vas u. 17, H-1088, Budapest, Hungary
| | - Illés J Farkas
- Statistical and Biological Physics Group of the Hungarian Acad. of Sciences, Pázmány P. s. 1A, H-1117, Budapest, Hungary
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
| | - Péter Csermely
- Department of Medical Chemistry, Semmelweis University, PO Box 260, H-1444, Budapest, Hungary
| | - Tamás Korcsmáros
- Department of Genetics, Eötvös Loránd University, Pázmány P. s. 1C, H-1117, Budapest, Hungary
- Department of Medical Chemistry, Semmelweis University, PO Box 260, H-1444, Budapest, Hungary
| |
Collapse
|
45
|
Pálfy M, Földvári-Nagy L, Módos D, Lenti K, Korcsmáros T. Reconstruction and Comparison of Cellular Signaling Pathway Resources for the Systems-Level Analysis of Cross-Talks. SYSTEMS BIOLOGY 2013:463-477. [DOI: 10.1007/978-94-007-6803-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
46
|
Ding Y, Chen M, Liu Z, Ding D, Ye Y, Zhang M, Kelly R, Guo L, Su Z, Harris SC, Qian F, Ge W, Fang H, Xu X, Tong W. atBioNet--an integrated network analysis tool for genomics and biomarker discovery. BMC Genomics 2012; 13:325. [PMID: 22817640 PMCID: PMC3443675 DOI: 10.1186/1471-2164-13-325] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 07/09/2012] [Indexed: 11/21/2022] Open
Abstract
Background Large amounts of mammalian protein-protein interaction (PPI) data have been generated and are available for public use. From a systems biology perspective, Proteins/genes interactions encode the key mechanisms distinguishing disease and health, and such mechanisms can be uncovered through network analysis. An effective network analysis tool should integrate different content-specific PPI databases into a comprehensive network format with a user-friendly platform to identify key functional modules/pathways and the underlying mechanisms of disease and toxicity. Results atBioNet integrates seven publicly available PPI databases into a network-specific knowledge base. Knowledge expansion is achieved by expanding a user supplied proteins/genes list with interactions from its integrated PPI network. The statistically significant functional modules are determined by applying a fast network-clustering algorithm (SCAN: a Structural Clustering Algorithm for Networks). The functional modules can be visualized either separately or together in the context of the whole network. Integration of pathway information enables enrichment analysis and assessment of the biological function of modules. Three case studies are presented using publicly available disease gene signatures as a basis to discover new biomarkers for acute leukemia, systemic lupus erythematosus, and breast cancer. The results demonstrated that atBioNet can not only identify functional modules and pathways related to the studied diseases, but this information can also be used to hypothesize novel biomarkers for future analysis. Conclusion atBioNet is a free web-based network analysis tool that provides a systematic insight into proteins/genes interactions through examining significant functional modules. The identified functional modules are useful for determining underlying mechanisms of disease and biomarker discovery. It can be accessed at: http://www.fda.gov/ScienceResearch/BioinformaticsTools/ucm285284.htm.
Collapse
Affiliation(s)
- Yijun Ding
- ICF International at FDA's National Center for Toxicological Research, Jefferson, AR 72079, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang Z, Liu J, Yu Y, Chen Y, Wang Y. Modular pharmacology: the next paradigm in drug discovery. Expert Opin Drug Discov 2012; 7:667-77. [DOI: 10.1517/17460441.2012.692673] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
48
|
Discovery of intramolecular signal transduction network based on a new protein dynamics model of energy dissipation. PLoS One 2012; 7:e31529. [PMID: 22363664 PMCID: PMC3282753 DOI: 10.1371/journal.pone.0031529] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 01/09/2012] [Indexed: 11/19/2022] Open
Abstract
A novel approach to reveal intramolecular signal transduction network is proposed in this work. To this end, a new algorithm of network construction is developed, which is based on a new protein dynamics model of energy dissipation. A key feature of this approach is that direction information is specified after inferring protein residue-residue interaction network involved in the process of signal transduction. This enables fundamental analysis of the regulation hierarchy and identification of regulation hubs of the signaling network. A well-studied allosteric enzyme, E. coli aspartokinase III, is used as a model system to demonstrate the new method. Comparison with experimental results shows that the new approach is able to predict all the sites that have been experimentally proved to desensitize allosteric regulation of the enzyme. In addition, the signal transduction network shows a clear preference for specific structural regions, secondary structural types and residue conservation. Occurrence of super-hubs in the network indicates that allosteric regulation tends to gather residues with high connection ability to collectively facilitate the signaling process. Furthermore, a new parameter of propagation coefficient is defined to determine the propagation capability of residues within a signal transduction network. In conclusion, the new approach is useful for fundamental understanding of the process of intramolecular signal transduction and thus has significant impact on rational design of novel allosteric proteins.
Collapse
|
49
|
MOfinder: a novel algorithm for detecting overlapping modules from protein-protein interaction network. J Biomed Biotechnol 2012; 2012:103702. [PMID: 22500072 PMCID: PMC3303734 DOI: 10.1155/2012/103702] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/19/2011] [Accepted: 10/21/2011] [Indexed: 11/17/2022] Open
Abstract
Since organism development and many critical cell biology processes are organized in modular patterns, many algorithms have been proposed to detect modules. In this study, a new method, MOfinder, was developed to detect overlapping modules in a protein-protein interaction (PPI) network. We demonstrate that our method is more accurate than other 5 methods. Then, we applied MOfinder to yeast and human PPI network and explored the overlapping information. Using the overlapping modules of human PPI network, we constructed the module-module communication network. Functional annotation showed that the immune-related and cancer-related proteins were always together and present in the same modules, which offer some clues for immune therapy for cancer. Our study around overlapping modules suggests a new perspective on the analysis of PPI network and improves our understanding of disease.
Collapse
|
50
|
Nacher JC, Schwartz JM. Modularity in protein complex and drug interactions reveals new polypharmacological properties. PLoS One 2012; 7:e30028. [PMID: 22279562 PMCID: PMC3261189 DOI: 10.1371/journal.pone.0030028] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 12/12/2011] [Indexed: 11/18/2022] Open
Abstract
Recent studies have highlighted the importance of interconnectivity in a large range of molecular and human disease-related systems. Network medicine has emerged as a new paradigm to deal with complex diseases. Connections between protein complexes and key diseases have been suggested for decades. However, it was not until recently that protein complexes were identified and classified in sufficient amounts to carry out a large-scale analysis of the human protein complex system. We here present the first systematic and comprehensive set of relationships between protein complexes and associated drugs and analyzed their topological features. The network structure is characterized by a high modularity, both in the bipartite graph and in its projections, indicating that its topology is highly distinct from a random network and that it contains a rich and heterogeneous internal modular structure. To unravel the relationships between modules of protein complexes, drugs and diseases, we investigated in depth the origins of this modular structure in examples of particular diseases. This analysis unveils new associations between diseases and protein complexes and highlights the potential role of polypharmacological drugs, which target multiple cellular functions to combat complex diseases driven by gain-of-function mutations.
Collapse
Affiliation(s)
- Jose C Nacher
- Department of Complex and Intelligent Systems, Future University Hakodate, Hokkaido, Japan.
| | | |
Collapse
|