151
|
Malaney P, Uversky VN, Davé V. Identification of intrinsically disordered regions in PTEN and delineation of its function via a network approach. Methods 2014; 77-78:69-74. [PMID: 25449897 DOI: 10.1016/j.ymeth.2014.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/01/2014] [Accepted: 10/06/2014] [Indexed: 12/14/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) are proteins that lack stable higher order structures for the entire protein molecule or a significant portion of it. The discovery of IDPs evolved as an antithesis to the conventional structure-function paradigm wherein a higher order structure dictates protein function. Over the last decade, a number of proteins with functionally relevant unstructured regions have been discovered, which includes tumor suppressor PTEN. The protein domains that lack structure provide "hot-spots" for post-translational modifications (PTMs) and protein-protein interactions (PPIs), which facilitate their regulation and participation in multiple cellular processes. Consequently, dysregulation in IDPs contribute to aberrant cellular pathophysiology. Herein, we present PTEN and its translational isoform PTEN-L as a hybrid protein possessing ordered domain and intrinsically disordered C-terminal and an N-terminal tails. We review the role of intrinsic disorder in PTEN function and propose a methodology for the use of intrinsic disorder to study PTEN-regulated higher order protein-networks by associating basic principles of network biology to functional pathway analysis at the systems level.
Collapse
Affiliation(s)
- Prerna Malaney
- Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Institute for Biological Instrumentation, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia; Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Vrushank Davé
- Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States.
| |
Collapse
|
152
|
|
153
|
Muraro D, Lauffenburger DA, Simmons A. Prioritisation and network analysis of Crohn's disease susceptibility genes. PLoS One 2014; 9:e108624. [PMID: 25268122 PMCID: PMC4182533 DOI: 10.1371/journal.pone.0108624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/01/2014] [Indexed: 01/23/2023] Open
Abstract
Recent Genome-Wide Association Studies (GWAS) have revealed numerous Crohn's disease susceptibility genes and a key challenge now is in understanding how risk polymorphisms in associated genes might contribute to development of this disease. For a gene to contribute to disease phenotype, its risk variant will likely adversely communicate with a variety of other gene products to result in dysregulation of common signaling pathways. A vital challenge is to elucidate pathways of potentially greatest influence on pathological behaviour, in a manner recognizing how multiple relevant genes may yield integrative effect. In this work we apply mathematical analysis of networks involving the list of recently described Crohn's susceptibility genes, to prioritise pathways in relation to their potential development of this disease. Prioritisation was performed by applying a text mining and a diffusion based method (GRAIL, GPEC). Prospective biological significance of the resulting prioritised list of proteins is highlighted by changes in their gene expression levels in Crohn's patients intestinal tissue in comparison with healthy donors.
Collapse
Affiliation(s)
- Daniele Muraro
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- * E-mail:
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Alison Simmons
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
154
|
Rai A, Menon AV, Jalan S. Randomness and preserved patterns in cancer network. Sci Rep 2014; 4:6368. [PMID: 25220184 PMCID: PMC5376158 DOI: 10.1038/srep06368] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/26/2014] [Indexed: 01/16/2023] Open
Abstract
Breast cancer has been reported to account for the maximum cases among all female cancers till date. In order to gain a deeper insight into the complexities of the disease, we analyze the breast cancer network and its normal counterpart at the proteomic level. While the short range correlations in the eigenvalues exhibiting universality provide an evidence towards the importance of random connections in the underlying networks, the long range correlations along with the localization properties reveal insightful structural patterns involving functionally important proteins. The analysis provides a benchmark for designing drugs which can target a subgraph instead of individual proteins.
Collapse
Affiliation(s)
- Aparna Rai
- Centre for Bio-Science and Bio-Medical Engineering, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India
| | - A Vipin Menon
- Complex Systems Lab, Discipline of Physics, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India
| | - Sarika Jalan
- 1] Centre for Bio-Science and Bio-Medical Engineering, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India [2] Complex Systems Lab, Discipline of Physics, Indian Institute of Technology Indore, M-Block, IET-DAVV Campus, Khandwa Road, Indore 452017, India
| |
Collapse
|
155
|
Abstract
MOTIVATION Biological network comparison software largely relies on the concept of alignment where close matches between the nodes of two or more networks are sought. These node matches are based on sequence similarity and/or interaction patterns. However, because of the incomplete and error-prone datasets currently available, such methods have had limited success. Moreover, the results of network alignment are in general not amenable for distance-based evolutionary analysis of sets of networks. In this article, we describe Netdis, a topology-based distance measure between networks, which offers the possibility of network phylogeny reconstruction. RESULTS We first demonstrate that Netdis is able to correctly separate different random graph model types independent of network size and density. The biological applicability of the method is then shown by its ability to build the correct phylogenetic tree of species based solely on the topology of current protein interaction networks. Our results provide new evidence that the topology of protein interaction networks contains information about evolutionary processes, despite the lack of conservation of individual interactions. As Netdis is applicable to all networks because of its speed and simplicity, we apply it to a large collection of biological and non-biological networks where it clusters diverse networks by type. AVAILABILITY AND IMPLEMENTATION The source code of the program is freely available at http://www.stats.ox.ac.uk/research/proteins/resources. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Waqar Ali
- Department of Statistics, University of Oxford, Oxford OX1 3TG, UK and Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, CA 90089-2910, USA
| | - Tiago Rito
- Department of Statistics, University of Oxford, Oxford OX1 3TG, UK and Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, CA 90089-2910, USA
| | - Gesine Reinert
- Department of Statistics, University of Oxford, Oxford OX1 3TG, UK and Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, CA 90089-2910, USA
| | - Fengzhu Sun
- Department of Statistics, University of Oxford, Oxford OX1 3TG, UK and Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, CA 90089-2910, USA
| | - Charlotte M Deane
- Department of Statistics, University of Oxford, Oxford OX1 3TG, UK and Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, CA 90089-2910, USA
| |
Collapse
|
156
|
Liu J, Spulber M, Wu D, Talom RM, Palivan CG, Meier W. Poly(N-isopropylacrylamide-co-tris-nitrilotriacetic acid acrylamide) for a Combined Study of Molecular Recognition and Spatial Constraints in Protein Binding and Interactions. J Am Chem Soc 2014; 136:12607-14. [DOI: 10.1021/ja503632w] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Juan Liu
- Department of Chemistry, University of Basel, Klingelbergstrasse
80, Basel 4056, Switzerland
| | - Mariana Spulber
- Department of Chemistry, University of Basel, Klingelbergstrasse
80, Basel 4056, Switzerland
| | - Dalin Wu
- Department of Chemistry, University of Basel, Klingelbergstrasse
80, Basel 4056, Switzerland
| | - Renee M. Talom
- Department of Chemistry, University of Basel, Klingelbergstrasse
80, Basel 4056, Switzerland
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Klingelbergstrasse
80, Basel 4056, Switzerland
| | - Wolfgang Meier
- Department of Chemistry, University of Basel, Klingelbergstrasse
80, Basel 4056, Switzerland
| |
Collapse
|
157
|
Chinenov Y, Coppo M, Gupte R, Sacta MA, Rogatsky I. Glucocorticoid receptor coordinates transcription factor-dominated regulatory network in macrophages. BMC Genomics 2014; 15:656. [PMID: 25099603 PMCID: PMC4133603 DOI: 10.1186/1471-2164-15-656] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/25/2014] [Indexed: 12/11/2022] Open
Abstract
Background Inflammation triggered by infection or injury is tightly controlled by glucocorticoid hormones which signal via a dedicated transcription factor, the Glucocorticoid Receptor (GR), to regulate hundreds of genes. However, the hierarchy of transcriptional responses to GR activation and the molecular basis of their oftentimes non-linear dynamics are not understood. Results We investigated early glucocorticoid-driven transcriptional events in macrophages, a cell type highly responsive to both pro- and anti-inflammatory stimuli. Using whole transcriptome analyses in resting and acutely lipopolysaccharide (LPS)-stimulated macrophages, we show that early GR target genes form dense networks with the majority of control nodes represented by transcription factors. The expression dynamics of several glucocorticoid-responsive genes are consistent with feed forward loops (FFL) and coincide with rapid GR recruitment. Notably, GR binding sites in genes encoding members of the KLF transcription factor family colocalize with KLF binding sites. Moreover, our gene expression, transcription factor binding and computational data are consistent with the existence of the GR-KLF9-KLF2 incoherent FFL. Analysis of LPS-downregulated genes revealed striking enrichment in multimerized Zn-fingers- and KRAB domain-containing proteins known to bind nucleic acids and repress transcription by propagating heterochromatin. This raises an intriguing possibility that an increase in chromatin accessibility in inflammatory macrophages results from broad downregulation of negative chromatin remodelers. Conclusions Pro- and anti-inflammatory stimuli alter the expression of a vast array of transcription factors and chromatin remodelers. By regulating multiple transcription factors, which propagate the initial hormonal signal, GR acts as a coordinating hub in anti-inflammatory responses. As several KLFs promote the anti-inflammatory program in macrophages, we propose that GR and KLFs functionally cooperate to curb inflammation. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-656) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yurii Chinenov
- Hospital for Special Surgery, The David Rosensweig Genomics Center, 535 East 70th Street, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
158
|
Li R, Li X, Ning S, Ye J, Han L, Kang C, Li X. Identification of a core miRNA-pathway regulatory network in glioma by therapeutically targeting miR-181d, miR-21, miR-23b, β-Catenin, CBP, and STAT3. PLoS One 2014; 9:e101903. [PMID: 25007077 PMCID: PMC4090169 DOI: 10.1371/journal.pone.0101903] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 06/12/2014] [Indexed: 11/24/2022] Open
Abstract
The application of microRNAs (miRNAs) in the therapeutics of glioma and other human diseases is an area of intense interest. However, it’s still a great challenge to interpret the functional consequences of using miRNAs in glioma therapy. Here, we examined paired deep sequencing expression profiles of miRNAs and mRNAs from human glioma cell lines after manipulating the levels of miRNAs miR-181d, -21, and -23b, as well as transcriptional regulators β-catenin, CBP, and STAT3. An integrated approach was used to identify functional miRNA-pathway regulatory networks (MPRNs) responding to each manipulation. MiRNAs were identified to regulate glioma related biological pathways collaboratively after manipulating the level of either post-transcriptional or transcriptional regulators, and functional synergy and crosstalk was observed between different MPRNs. MPRNs responsive to multiple interventions were found to occupy central positions in the comprehensive MPRN (cMPRN) generated by integrating all the six MPRNs. Finally, we identified a core module comprising 14 miRNAs and five pathways that could predict the survival of glioma patients and represent potential targets for glioma therapy. Our results provided novel insight into miRNA regulatory mechanisms implicated in therapeutic interventions and could offer more inspiration to miRNA-based glioma therapy.
Collapse
Affiliation(s)
- Ronghong Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xiang Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jingrun Ye
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lei Han
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Laboratory of Neurotrauma, Variation and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, China
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Laboratory of Neurotrauma, Variation and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, China
- * E-mail: (XL); (CK)
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- * E-mail: (XL); (CK)
| |
Collapse
|
159
|
Najafi A, Masoudi-Nejad A, Ghanei M, Nourani MR, Moeini A. Pathway reconstruction of airway remodeling in chronic lung diseases: a systems biology approach. PLoS One 2014; 9:e100094. [PMID: 24978043 PMCID: PMC4076832 DOI: 10.1371/journal.pone.0100094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 05/22/2014] [Indexed: 01/01/2023] Open
Abstract
Airway remodeling is a pathophysiologic process at the clinical, cellular, and molecular level relating to chronic obstructive airway diseases such as chronic obstructive pulmonary disease (COPD), asthma and mustard lung. These diseases are associated with the dysregulation of multiple molecular pathways in the airway cells. Little progress has so far been made in discovering the molecular causes of complex disease in a holistic systems manner. Therefore, pathway and network reconstruction is an essential part of a systems biology approach to solve this challenging problem. In this paper, multiple data sources were used to construct the molecular process of airway remodeling pathway in mustard lung as a model of airway disease. We first compiled a master list of genes that change with airway remodeling in the mustard lung disease and then reconstructed the pathway by generating and merging the protein-protein interaction and the gene regulatory networks. Experimental observations and literature mining were used to identify and validate the master list. The outcome of this paper can provide valuable information about closely related chronic obstructive airway diseases which are of great importance for biologists and their future research. Reconstructing the airway remodeling interactome provides a starting point and reference for the future experimental study of mustard lung, and further analysis and development of these maps will be critical to understanding airway diseases in patients.
Collapse
Affiliation(s)
- Ali Najafi
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- * E-mail:
| | - Mostafa Ghanei
- Genomics Division, Chemical Injury Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohamad-Reza Nourani
- Genomics Division, Chemical Injury Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Moeini
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Department of Algorithms and Computation, College of Engineering, University of Tehran, Tehran, Iran
| |
Collapse
|
160
|
Tran NTL, Mohan S, Xu Z, Huang CH. Current innovations and future challenges of network motif detection. Brief Bioinform 2014; 16:497-525. [PMID: 24966356 DOI: 10.1093/bib/bbu021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 05/25/2014] [Indexed: 01/09/2023] Open
Abstract
Network motif detection is the search for statistically overrepresented subgraphs present in a larger target network. They are thought to represent key structure and control mechanisms. Although the problem is exponential in nature, several algorithms and tools have been developed for efficiently detecting network motifs. This work analyzes 11 network motif detection tools and algorithms. Detailed comparisons and insightful directions for using these tools and algorithms are discussed. Key aspects of network motif detection are investigated. Network motif types and common network motifs as well as their biological functions are discussed. Applications of network motifs are also presented. Finally, the challenges, future improvements and future research directions for network motif detection are also discussed.
Collapse
|
161
|
Wu B, Li C, Du Z, Yao Q, Wu J, Feng L, Zhang P, Li S, Xu L, Li E. Network based analyses of gene expression profile of LCN2 overexpression in esophageal squamous cell carcinoma. Sci Rep 2014; 4:5403. [PMID: 24954627 PMCID: PMC4066265 DOI: 10.1038/srep05403] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/03/2014] [Indexed: 02/05/2023] Open
Abstract
LCN2 (lipocalin 2) is a member of the lipocalin family of proteins that transport small, hydrophobic ligands. LCN2 is elevated in various cancers including esophageal squamous cell carcinoma (ESCC). In this study, LCN2 was overexpressed in the EC109 ESCC cell line and we applied integrated analyses of the gene expression data to identify protein-protein interactions (PPI) network to enhance our understanding of the role of LCN2 in ESCC. Through further mining of PPI sub-networks, hundreds of differentially expressed genes (DEGs) were identified to interact with thousands of other proteins. Subcellular localization analyses found the DEGs and their directly or indirectly interacting proteins distributed in multiple layers, which was applied to analyze the possible paths between two DEGs. Gene Ontology annotation generated a functional annotation map and found hundreds of significant terms, especially those associated with the known and potential roles of LCN2 protein. The algorithm of Random Walk with Restart was applied to prioritize the DEGs and identified several cancer-related DEGs ranked closest to LCN2 protein. These analyses based on PPI network have greatly expanded our understanding of the mRNA expression profile of LCN2 overexpression for future examination of the roles and mechanisms of LCN2.
Collapse
Affiliation(s)
- Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, China
- These authors contributed equally to this work
| | - Chunquan Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, China
- College of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing163319, China
- These authors contributed equally to this work
| | - Zepeng Du
- Department of Pathology, Shantou Central Hospital, Shantou515041, China
| | - Qianlan Yao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin150081, China
| | - Jianyi Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, China
| | - Li Feng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin150081, China
| | - Pixian Zhang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, China
| | - Shang Li
- College of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing163319, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou515041, China
| |
Collapse
|
162
|
Borroto-Escuela DO, Brito I, Romero-Fernandez W, Di Palma M, Oflijan J, Skieterska K, Duchou J, Van Craenenbroeck K, Suárez-Boomgaard D, Rivera A, Guidolin D, Agnati LF, Fuxe K. The G protein-coupled receptor heterodimer network (GPCR-HetNet) and its hub components. Int J Mol Sci 2014; 15:8570-90. [PMID: 24830558 PMCID: PMC4057749 DOI: 10.3390/ijms15058570] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/26/2014] [Accepted: 04/30/2014] [Indexed: 01/09/2023] Open
Abstract
G protein-coupled receptors (GPCRs) oligomerization has emerged as a vital characteristic of receptor structure. Substantial experimental evidence supports the existence of GPCR-GPCR interactions in a coordinated and cooperative manner. However, despite the current development of experimental techniques for large-scale detection of GPCR heteromers, in order to understand their connectivity it is necessary to develop novel tools to study the global heteroreceptor networks. To provide insight into the overall topology of the GPCR heteromers and identify key players, a collective interaction network was constructed. Experimental interaction data for each of the individual human GPCR protomers was obtained manually from the STRING and SCOPUS databases. The interaction data were used to build and analyze the network using Cytoscape software. The network was treated as undirected throughout the study. It is comprised of 156 nodes, 260 edges and has a scale-free topology. Connectivity analysis reveals a significant dominance of intrafamily versus interfamily connections. Most of the receptors within the network are linked to each other by a small number of edges. DRD2, OPRM, ADRB2, AA2AR, AA1R, OPRK, OPRD and GHSR are identified as hubs. In a network representation 10 modules/clusters also appear as a highly interconnected group of nodes. Information on this GPCR network can improve our understanding of molecular integration. GPCR-HetNet has been implemented in Java and is freely available at http://www.iiia.csic.es/~ismel/GPCR-Nets/index.html.
Collapse
Affiliation(s)
| | - Ismel Brito
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden.
| | | | - Michael Di Palma
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden.
| | - Julia Oflijan
- Department of Physiology, Faculty of Medicine, University of Tartu, Tartu 50411, Estonia.
| | - Kamila Skieterska
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University, 9000 Ghent, Belgium.
| | - Jolien Duchou
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University, 9000 Ghent, Belgium.
| | - Kathleen Van Craenenbroeck
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University, 9000 Ghent, Belgium.
| | - Diana Suárez-Boomgaard
- Department of Cell Biology, School of Science, University of Málaga, 29071 Málaga, Spain.
| | - Alicia Rivera
- Department of Cell Biology, School of Science, University of Málaga, 29071 Málaga, Spain.
| | - Diego Guidolin
- Department of Molecular Medicine, University of Padova, Padova 35121, Italy.
| | - Luigi F Agnati
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden.
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden.
| |
Collapse
|
163
|
Differential evolutionary constraints in the evolution of chemoreceptors: a murine and human case study. ScientificWorldJournal 2014; 2014:696485. [PMID: 24587745 PMCID: PMC3920627 DOI: 10.1155/2014/696485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/23/2013] [Indexed: 02/08/2023] Open
Abstract
Chemoreception is among the most important sensory modalities in animals. Organisms use the ability to perceive chemical compounds in all major ecological activities. Recent studies have allowed the characterization of chemoreceptor gene families. These genes present strikingly high variability in copy numbers and pseudogenization degrees among different species, but the mechanisms underlying their evolution are not fully understood. We have analyzed the functional networks of these genes, their orthologs distribution, and performed phylogenetic analyses in order to investigate their evolutionary dynamics. We have modeled the chemosensory networks and compared the evolutionary constraints of their genes in Mus musculus, Homo sapiens, and Rattus norvegicus. We have observed significant differences regarding the constraints on the orthologous groups and network topologies of chemoreceptors and signal transduction machinery. Our findings suggest that chemosensory receptor genes are less constrained than their signal transducing machinery, resulting in greater receptor diversity and conservation of information processing pathways. More importantly, we have observed significant differences among the receptors themselves, suggesting that olfactory and bitter taste receptors are more conserved than vomeronasal receptors.
Collapse
|
164
|
Mining large-scale response networks reveals 'topmost activities' in Mycobacterium tuberculosis infection. Sci Rep 2014; 3:2302. [PMID: 23892477 PMCID: PMC3725478 DOI: 10.1038/srep02302] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/10/2013] [Indexed: 02/02/2023] Open
Abstract
Mycobacterium tuberculosis owes its high pathogenic potential to its ability to evade host immune responses and thrive inside the macrophage. The outcome of infection is largely determined by the cellular response comprising a multitude of molecular events. The complexity and inter-relatedness in the processes makes it essential to adopt systems approaches to study them. In this work, we construct a comprehensive network of infection-related processes in a human macrophage comprising 1888 proteins and 14,016 interactions. We then compute response networks based on available gene expression profiles corresponding to states of health, disease and drug treatment. We use a novel formulation for mining response networks that has led to identifying highest activities in the cell. Highest activity paths provide mechanistic insights into pathogenesis and response to treatment. The approach used here serves as a generic framework for mining dynamic changes in genome-scale protein interaction networks.
Collapse
|
165
|
Netotea S, Sundell D, Street NR, Hvidsten TR. ComPlEx: conservation and divergence of co-expression networks in A. thaliana, Populus and O. sativa. BMC Genomics 2014; 15:106. [PMID: 24498971 PMCID: PMC3925997 DOI: 10.1186/1471-2164-15-106] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 01/29/2014] [Indexed: 01/16/2023] Open
Abstract
Background Divergence in gene regulation has emerged as a key mechanism underlying species differentiation. Comparative analysis of co-expression networks across species can reveal conservation and divergence in the regulation of genes. Results We inferred co-expression networks of A. thaliana, Populus spp. and O. sativa using state-of-the-art methods based on mutual information and context likelihood of relatedness, and conducted a comprehensive comparison of these networks across a range of co-expression thresholds. In addition to quantifying gene-gene link and network neighbourhood conservation, we also applied recent advancements in network analysis to do cross-species comparisons of network properties such as scale free characteristics and gene centrality as well as network motifs. We found that in all species the networks emerged as scale free only above a certain co-expression threshold, and that the high-centrality genes upholding this organization tended to be conserved. Network motifs, in particular the feed-forward loop, were found to be significantly enriched in specific functional subnetworks but where much less conserved across species than gene centrality. Although individual gene-gene co-expression had massively diverged, up to ~80% of the genes still had a significantly conserved network neighbourhood. For genes with multiple predicted orthologs, about half had one ortholog with conserved regulation and another ortholog with diverged or non-conserved regulation. Furthermore, the most sequence similar ortholog was not the one with the most conserved gene regulation in over half of the cases. Conclusions We have provided a comprehensive analysis of gene regulation evolution in plants and built a web tool for Comparative analysis of Plant co-Expression networks (ComPlEx, http://complex.plantgenie.org/). The tool can be particularly useful for identifying the ortholog with the most conserved regulation among several sequence-similar alternatives and can thus be of practical importance in e.g. finding candidate genes for perturbation experiments.
Collapse
Affiliation(s)
| | | | | | - Torgeir R Hvidsten
- Umeå Plant Science Center, Department of Plant Physiology, Umeå University, Umeå, Sweden.
| |
Collapse
|
166
|
Perumal TM, Gunawan R. pathPSA: A Dynamical Pathway-Based Parametric Sensitivity Analysis. Ind Eng Chem Res 2014. [DOI: 10.1021/ie403277d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Thanneer Malai Perumal
- Luxembourg
Centre for Systems Biomedicine, University of Luxembourg, Esch/Alzette 4362, Luxembourg
| | - Rudiyanto Gunawan
- Institute
for Chemical and Bioengineering, ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
167
|
Cardinal-Fernández P, Nin N, Ruíz-Cabello J, Lorente JA. Systems medicine: a new approach to clinical practice. Arch Bronconeumol 2014; 50:444-51. [PMID: 24397963 DOI: 10.1016/j.arbres.2013.10.010] [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: 08/26/2013] [Revised: 10/13/2013] [Accepted: 10/31/2013] [Indexed: 10/25/2022]
Abstract
Most respiratory diseases are considered complex diseases as their susceptibility and outcomes are determined by the interaction between host-dependent factors (genetic factors, comorbidities, etc.) and environmental factors (exposure to microorganisms or allergens, treatments received, etc.) The reductionist approach in the study of diseases has been of fundamental importance for the understanding of the different components of a system. Systems biology or systems medicine is a complementary approach aimed at analyzing the interactions between the different components within one organizational level (genome, transcriptome, proteome), and then between the different levels. Systems medicine is currently used for the interpretation and understanding of the pathogenesis and pathophysiology of different diseases, biomarker discovery, design of innovative therapeutic targets, and the drawing up of computational models for different biological processes. In this review we discuss the most relevant concepts of the theory underlying systems medicine, as well as its applications in the various biological processes in humans.
Collapse
Affiliation(s)
- Pablo Cardinal-Fernández
- Servicio de Medicina Intensiva, Hospital Universitario de Getafe, Madrid, España; CIBER de Enfermedades Respiratorias, Madrid, España
| | - Nicolás Nin
- CIBER de Enfermedades Respiratorias, Madrid, España; Servicio de Medicina Intensiva, Hospital Universitario de Torrejón, Madrid, España
| | - Jesús Ruíz-Cabello
- CIBER de Enfermedades Respiratorias, Madrid, España; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, España; Universidad Complutense de Madrid, Madrid, España
| | - José A Lorente
- Servicio de Medicina Intensiva, Hospital Universitario de Getafe, Madrid, España; CIBER de Enfermedades Respiratorias, Madrid, España; Universidad Europea de Madrid, Madrid, España.
| |
Collapse
|
168
|
Wu BL, Zou HY, Lv GQ, Du ZP, Wu JY, Zhang PX, Xu LY, Li EM. Protein-protein interaction network analyses for elucidating the roles of LOXL2-delta72 in esophageal squamous cell carcinoma. Asian Pac J Cancer Prev 2014; 15:2345-51. [PMID: 24716982 DOI: 10.7314/apjcp.2014.15.5.2345] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Lysyl oxidase-like 2 (LOXL2), a member of the lysyl oxidase (LOX) family, is a copper-dependent enzyme that catalyzes oxidative deamination of lysine residues on protein substrates. LOXL2 was found to be overexpressed in esophageal squamous cell carcinoma (ESCC) in our previous research. We later identified a LOXL2 splicing variant LOXL2-delta72 and we overexpressed LOXL2-delta72 and its wild type counterpart in ESCC cells following microarray analyses. First, the differentially expressed genes (DEGs) of LOXL2 and LOXL2-delta72 compared to empty plasmid were applied to generate protein-protein interaction (PPI) sub-networks. Comparison of these two sub-networks showed hundreds of different proteins. To reveal the potential specific roles of LOXL2- delta72 compared to its wild type, the DEGs of LOXL2-delta72 vs LOXL2 were also applied to construct a PPI sub-network which was annotated by Gene Ontology. The functional annotation map indicated the third PPI sub-network involved hundreds of GO terms, such as "cell cycle arrest", "G1/S transition of mitotic cell cycle", "interphase", "cell-matrix adhesion" and "cell-substrate adhesion", as well as significant "immunity" related terms, such as "innate immune response", "regulation of defense response" and "Toll signaling pathway". These results provide important clues for experimental identification of the specific biological roles and molecular mechanisms of LOXL2-delta72. This study also provided a work flow to test the different roles of a splicing variant with high-throughput data.
Collapse
Affiliation(s)
- Bing-Li Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China E-mail : ,
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Abstract
Tagging items with descriptive annotations or keywords is a very natural way to compress and highlight information about the properties of the given entity. Over the years several methods have been proposed for extracting a hierarchy between the tags for systems with a "flat", egalitarian organization of the tags, which is very common when the tags correspond to free words given by numerous independent people. Here we present a complete framework for automated tag hierarchy extraction based on tag occurrence statistics. Along with proposing new algorithms, we are also introducing different quality measures enabling the detailed comparison of competing approaches from different aspects. Furthermore, we set up a synthetic, computer generated benchmark providing a versatile tool for testing, with a couple of tunable parameters capable of generating a wide range of test beds. Beside the computer generated input we also use real data in our studies, including a biological example with a pre-defined hierarchy between the tags. The encouraging similarity between the pre-defined and reconstructed hierarchy, as well as the seemingly meaningful hierarchies obtained for other real systems indicate that tag hierarchy extraction is a very promising direction for further research with a great potential for practical applications. Tags have become very prevalent nowadays in various online platforms ranging from blogs through scientific publications to protein databases. Furthermore, tagging systems dedicated for voluntary tagging of photos, films, books, etc. with free words are also becoming popular. The emerging large collections of tags associated with different objects are often referred to as folksonomies, highlighting their collaborative origin and the “flat” organization of the tags opposed to traditional hierarchical categorization. Adding a tag hierarchy corresponding to a given folksonomy can very effectively help narrowing or broadening the scope of search. Moreover, recommendation systems could also benefit from a tag hierarchy.
Collapse
Affiliation(s)
- Gergely Tibély
- Dept. of Biological Physics, Eötvös University, Budapest, Hungary
- Eötvös University, Regional Knowledge Centre, Székesfehervár, Hungary
| | - Péter Pollner
- Statistical and Biological Physics Research Group of HAS, Budapest, Hungary
- Eötvös University, Regional Knowledge Centre, Székesfehervár, Hungary
| | - Tamás Vicsek
- Dept. of Biological Physics, Eötvös University, Budapest, Hungary
- Eötvös University, Regional Knowledge Centre, Székesfehervár, Hungary
| | - Gergely Palla
- Statistical and Biological Physics Research Group of HAS, Budapest, Hungary
- Eötvös University, Regional Knowledge Centre, Székesfehervár, Hungary
- * E-mail:
| |
Collapse
|
170
|
Yu D, Kim M, Xiao G, Hwang TH. Review of biological network data and its applications. Genomics Inform 2013; 11:200-10. [PMID: 24465231 PMCID: PMC3897847 DOI: 10.5808/gi.2013.11.4.200] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/20/2013] [Accepted: 11/21/2013] [Indexed: 12/16/2022] Open
Abstract
Studying biological networks, such as protein-protein interactions, is key to understanding complex biological activities. Various types of large-scale biological datasets have been collected and analyzed with high-throughput technologies, including DNA microarray, next-generation sequencing, and the two-hybrid screening system, for this purpose. In this review, we focus on network-based approaches that help in understanding biological systems and identifying biological functions. Accordingly, this paper covers two major topics in network biology: reconstruction of gene regulatory networks and network-based applications, including protein function prediction, disease gene prioritization, and network-based genome-wide association study.
Collapse
Affiliation(s)
- Donghyeon Yu
- Department of Clinical Sciences, Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Minsoo Kim
- Department of Clinical Sciences, Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guanghua Xiao
- Department of Clinical Sciences, Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tae Hyun Hwang
- Department of Clinical Sciences, Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
171
|
Budd WT, Seashols S, Weaver D, Joseph C, Zehner ZE. A networks method for ranking microRNA dysregulation in cancer. BMC SYSTEMS BIOLOGY 2013; 7 Suppl 5:S3. [PMID: 24564923 PMCID: PMC4028974 DOI: 10.1186/1752-0509-7-s5-s3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Despite the lack of agreement on their exact roles, it is known that miRNAs contribute to cancer progression. Many studies utilize methods to detect differential regulation of miRNA expression. It is prohibitively expensive to examine all potentially dysregulated miRNAs and traditionally, researchers have focused their efforts on the most extremely dysregulated miRNAs. These methods may overlook the contribution of less differentially expressed but more functionally relevant miRNAs. The purpose of this study was to outline a method that not only utilizes differential expression but ranks miRNAs based on the functional relevance of their targets. This work uses a networks based approach to determine the sum node degree for all experimentally verified miRNA targets to identify potential regulators of prostate cancer initiation, progression and metastasis. RESULTS Here, we present a method for identifying functionally relevant miRNAs that contribute to prostate cancer development. This paper shows that miRNAs preferentially regulate highly connected, central proteins within a protein-protein interaction network. Known targets of miRNAs differentially regulated during prostate cancer progression are enriched in pathways with known involvement in tumorigenesis. To demonstrate the applicability of our method, we utilized a unique model of prostate cancer progression to identify five miRNAs that may contribute to the oncogenic state of the cell. Three of these miRNAs have been shown by other studies to have a role in cancer but their exact role in prostate cancer remains undefined. CONCLUSION Developing methods to determine which miRNAs to carry forward into biological and biochemical analyses is important as traditional approaches often overlook miRNAs that contribute to oncogenesis. Our method applied to a model of prostate cancer progression was able to identify miRNAs with roles in prostate cancer development.
Collapse
|
172
|
Jung N, Bertrand F, Bahram S, Vallat L, Maumy-Bertrand M. Cascade: a R package to study, predict and simulate the diffusion of a signal through a temporal gene network. ACTA ACUST UNITED AC 2013; 30:571-3. [PMID: 24307703 DOI: 10.1093/bioinformatics/btt705] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SUMMARY Temporal gene interactions, in response to environmental stress, form a complex system that can be efficiently described using gene regulatory networks. They allow highlighting the more influential genes and spotting some targets for biological intervention experiments. Despite that many reverse engineering tools have been designed, the Cascade package is an integrated solution adding several new and original key features such as the ability to predict changes in gene expressions after a biological perturbation in the network and graphical outputs that allow monitoring the spread of a signal through the network. AVAILABILITY AND IMPLEMENTATION The R package Cascade is available online at http://www-math.u-strasbg.fr/genpred/spip.php?rubrique4.
Collapse
Affiliation(s)
- Nicolas Jung
- INSERM UMR S_1109, Labex Transplantex, FMTS, Hôpitaux and Faculté de Médecine, Université de Strasbourg, 67085 Strasbourg Cedex and IRMA, CNRS UMR 7501, Labex IRMIA, Université de Strasbourg, 67084 Strasbourg Cedex, France
| | | | | | | | | |
Collapse
|
173
|
Wodak SJ, Vlasblom J, Turinsky AL, Pu S. Protein–protein interaction networks: the puzzling riches. Curr Opin Struct Biol 2013; 23:941-53. [DOI: 10.1016/j.sbi.2013.08.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 07/14/2013] [Accepted: 08/08/2013] [Indexed: 12/13/2022]
|
174
|
Sun X, Hong P, Kulkarni M, Kwon Y, Perrimon N. PPIRank - an advanced method for ranking protein-protein interations in TAP/MS data. Proteome Sci 2013; 11:S16. [PMID: 24565074 PMCID: PMC3908380 DOI: 10.1186/1477-5956-11-s1-s16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Tandem affinity purification coupled with mass-spectrometry (TAP/MS) analysis is a popular method for the identification of novel endogenous protein-protein interactions (PPIs) in large-scale. Computational analysis of TAP/MS data is a critical step, particularly for high-throughput datasets, yet it remains challenging due to the noisy nature of TAP/MS data. Results We investigated several major TAP/MS data analysis methods for identifying PPIs, and developed an advanced method, which incorporates an improved statistical method to filter out false positives from the negative controls. Our method is named PPIRank that stands for PPI ranking in TAP/MS data. We compared PPIRank with several other existing methods in analyzing two pathway-specific TAP/MS PPI datasets from Drosophila. Conclusion Experimental results show that PPIRank is more capable than other approaches in terms of identifying known interactions collected in the BioGRID PPI database. Specifically, PPIRank is able to capture more true interactions and simultaneously less false positives in both Insulin and Hippo pathways of Drosophila Melanogaster.
Collapse
|
175
|
Xiao Q, Wang J, Peng X, Wu FX. Detecting protein complexes from active protein interaction networks constructed with dynamic gene expression profiles. Proteome Sci 2013; 11:S20. [PMID: 24565281 PMCID: PMC3908890 DOI: 10.1186/1477-5956-11-s1-s20] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Protein interaction networks (PINs) are known to be useful to detect protein complexes. However, most available PINs are static, which cannot reflect the dynamic changes in real networks. At present, some researchers have tried to construct dynamic networks by incorporating time-course (dynamic) gene expression data with PINs. However, the inevitable background noise exists in the gene expression array, which could degrade the quality of dynamic networkds. Therefore, it is needed to filter out contaminated gene expression data before further data integration and analysis. Results Firstly, we adopt a dynamic model-based method to filter noisy data from dynamic expression profiles. Then a new method is proposed for identifying active proteins from dynamic gene expression profiles. An active protein at a time point is defined as the protein the expression level of whose corresponding gene at that time point is higher than a threshold determined by a standard variance involved threshold function. Furthermore, a noise-filtered active protein interaction network (NF-APIN) is constructed. To demonstrate the efficiency of our method, we detect protein complexes from the NF-APIN, compared with those from other dynamic PINs. Conclusion A dynamic model based method can effectively filter out noises in dynamic gene expression data. Our method to compute a threshold for determining the active time points of noise-filtered genes can make the dynamic construction more accuracy and provide a high quality framework for network analysis, such as protein complex prediction.
Collapse
|
176
|
Bernabò N, Barboni B, Maccarrone M. Systems biology analysis of the endocannabinoid system reveals a scale-free network with distinct roles for anandamide and 2-arachidonoylglycerol. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2013; 17:646-54. [PMID: 24117401 DOI: 10.1089/omi.2013.0071] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We represented the endocannabinoid system (ECS) as a biological network, where ECS molecules are the nodes (123) and their interactions the links (189). ECS network follows a scale-free topology, which confers robustness against random damage, easy navigability, and controllability. Network topological parameters, such as clustering coefficient (i.e., how the nodes form clusters) of 0.0009, network diameter (the longest shortest path among all pairs of nodes) of 12, averaged number of neighbors (the mean number of connections per node) of 3.073, and characteristic path length (the expected distance between two connected nodes) of 4.715, suggested that molecular messages are transferred through the ECS network quickly and specifically. Interestingly, ∼75% of nodes are located on, or are active at the level of, the cell membrane. The hubs of ECS network are anandamide (AEA) and 2-arachidonoylglycerol (2-AG), which have also the highest value of betweeness centrality, and their removal causes network collapse into multiple disconnected components. Importantly, AEA is a ubiquitous player while 2-AG plays more restricted actions. Instead, the product of their degradation, arachidonic acid, and their hydrolyzing enzyme, fatty acid amide hydrolase, FAAH, have a marginal impact on ECS network, indeed their removal did not significantly affect its topology.
Collapse
Affiliation(s)
- Nicola Bernabò
- 1 Department of Biomedical Sciences, University of Teramo , Teramo, Italy
| | | | | |
Collapse
|
177
|
Simple topological features reflect dynamics and modularity in protein interaction networks. PLoS Comput Biol 2013; 9:e1003243. [PMID: 24130468 PMCID: PMC3794914 DOI: 10.1371/journal.pcbi.1003243] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/14/2013] [Indexed: 11/30/2022] Open
Abstract
The availability of large-scale protein-protein interaction networks for numerous organisms provides an opportunity to comprehensively analyze whether simple properties of proteins are predictive of the roles they play in the functional organization of the cell. We begin by re-examining an influential but controversial characterization of the dynamic modularity of the S. cerevisiae interactome that incorporated gene expression data into network analysis. We analyse the protein-protein interaction networks of five organisms, S. cerevisiae, H. sapiens, D. melanogaster, A. thaliana, and E. coli, and confirm significant and consistent functional and structural differences between hub proteins that are co-expressed with their interacting partners and those that are not, and support the view that the former tend to be intramodular whereas the latter tend to be intermodular. However, we also demonstrate that in each of these organisms, simple topological measures are significantly correlated with the average co-expression of a hub with its partners, independent of any classification, and therefore also reflect protein intra- and inter- modularity. Further, cross-interactomic analysis demonstrates that these simple topological characteristics of hub proteins tend to be conserved across organisms. Overall, we give evidence that purely topological features of static interaction networks reflect aspects of the dynamics and modularity of interactomes as well as previous measures incorporating expression data, and are a powerful means for understanding the dynamic roles of hubs in interactomes. A better understanding of protein interaction networks would be a great aid in furthering our knowledge of the molecular biology of the cell. Towards this end, large-scale protein-protein physical interaction data have been determined for organisms across the evolutionary spectrum. However, the resulting networks give a static view of interactomes, and our knowledge about protein interactions is rarely time or context specific. A previous prominent but controversial attempt to characterize the dynamic modularity of the interactome was based on integrating physical interaction data with gene activity measurements from transcript expression data. This analysis distinguished between proteins that are co-expressed with their interacting partners and those that are not, and argued that the former are intramodular and the latter are intermodular. By analyzing the interactomes of five organisms, we largely confirm the biological significance of this characterization through a variety of statistical tests and computational experiments. Surprisingly, however, we find that similar results can be obtained using just network information without additionally integrating expression data, suggesting that purely topological characteristics of interaction networks strongly reflect certain aspects of the dynamics and modularity of interactomes.
Collapse
|
178
|
Vey G. Gene coexpression as Hebbian learning in prokaryotic genomes. Bull Math Biol 2013; 75:2431-49. [PMID: 24078338 DOI: 10.1007/s11538-013-9900-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 08/27/2013] [Indexed: 10/26/2022]
Abstract
Biological interaction networks represent a powerful tool for characterizing intracellular functional relationships, such as transcriptional regulation and protein interactions. Although artificial neural networks are routinely employed for a broad range of applications across computational biology, their underlying connectionist basis has not been extensively applied to modeling biological interaction networks. In particular, the Hopfield network offers nonlinear dynamics that represent the minimization of a system energy function through temporally distinct rewiring events. Here, a scaled energy minimization model is presented to test the feasibility of deriving a composite biological interaction network from multiple constituent data sets using the Hebbian learning principle. The performance of the scaled energy minimization model is compared against the standard Hopfield model using simulated data. Several networks are also derived from real data, compared to one another, and then combined to produce an aggregate network. The utility and limitations of the proposed model are discussed, along with possible implications for a genomic learning analogy where the fundamental Hebbian postulate is rendered into its genomic equivalent: Genes that function together junction together.
Collapse
|
179
|
Wanichthanarak K, Cvijovic M, Molt A, Petranovic D. yApoptosis: yeast apoptosis database. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2013; 2013:bat068. [PMID: 24082050 PMCID: PMC3786231 DOI: 10.1093/database/bat068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In the past few years, programmed cell death (PCD) has become a popular research area due to its fundamental aspects and its links to human diseases. Yeast has been used as a model for studying PCD, since the discovery of morphological markers of apoptotic cell death in yeast in 1997. Increasing knowledge in identification of components and molecular pathways created a need for organization of information. To meet the demands from the research community, we have developed a curated yeast apoptosis database, yApoptosis. The database structurally collects an extensively curated set of apoptosis, PCD and related genes, their genomic information, supporting literature and relevant external links. A web interface including necessary functions is provided to access and download the data. In addition, we included several networks where the apoptosis genes or proteins are involved, and present them graphically and interactively to facilitate rapid visualization. We also promote continuous inputs and curation by experts. yApoptosis is a highly specific resource for sharing information online, which supports researches and studies in the field of yeast apoptosis and cell death. Database URL:http://www.ycelldeath.com/yapoptosis/
Collapse
Affiliation(s)
- Kwanjeera Wanichthanarak
- Department of Chemical and Biological Engineering, Chalmers University of Technology, Kemivägen 10, 41296, Gothenburg, Sweden, Department of Mathematics, Chalmers University of Technology, Chalmers tvärgata 3, 41296, Gothenburg, Sweden, Department of Mathematics, University of Gothenburg, Chalmers tvärgata 3, 41296, Gothenburg, Sweden and Fine Chemicals and Biocatalysis Research, BASF SE, GVF/D - A030, 67056 Ludwigshafen, Germany
| | | | | | | |
Collapse
|
180
|
Somvanshi PR, Venkatesh KV. A conceptual review on systems biology in health and diseases: from biological networks to modern therapeutics. SYSTEMS AND SYNTHETIC BIOLOGY 2013; 8:99-116. [PMID: 24592295 DOI: 10.1007/s11693-013-9125-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/10/2013] [Indexed: 12/28/2022]
Abstract
Human physiology is an ensemble of various biological processes spanning from intracellular molecular interactions to the whole body phenotypic response. Systems biology endures to decipher these multi-scale biological networks and bridge the link between genotype to phenotype. The structure and dynamic properties of these networks are responsible for controlling and deciding the phenotypic state of a cell. Several cells and various tissues coordinate together to generate an organ level response which further regulates the ultimate physiological state. The overall network embeds a hierarchical regulatory structure, which when unusually perturbed can lead to undesirable physiological state termed as disease. Here, we treat a disease diagnosis problem analogous to a fault diagnosis problem in engineering systems. Accordingly we review the application of engineering methodologies to address human diseases from systems biological perspective. The review highlights potential networks and modeling approaches used for analyzing human diseases. The application of such analysis is illustrated in the case of cancer and diabetes. We put forth a concept of cell-to-human framework comprising of five modules (data mining, networking, modeling, experimental and validation) for addressing human physiology and diseases based on a paradigm of system level analysis. The review overtly emphasizes on the importance of multi-scale biological networks and subsequent modeling and analysis for drug target identification and designing efficient therapies.
Collapse
Affiliation(s)
- Pramod Rajaram Somvanshi
- Biosystems Engineering, Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076 Maharashtra India
| | - K V Venkatesh
- Biosystems Engineering, Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076 Maharashtra India
| |
Collapse
|
181
|
Skippington E, Ragan MA. Phylogeny rather than ecology or lifestyle biases the construction of Escherichia coli-Shigella genetic exchange communities. Open Biol 2013; 2:120112. [PMID: 23091700 PMCID: PMC3472396 DOI: 10.1098/rsob.120112] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 08/20/2012] [Indexed: 11/12/2022] Open
Abstract
Genetic material can be transmitted not only vertically from parent to offspring, but also laterally (horizontally) from one bacterial lineage to another. Lateral genetic transfer is non-uniform; biases in its nature or frequency construct communities of genetic exchange. These biases have been proposed to arise from phylogenetic relatedness, shared ecology and/or common lifestyle. Here, we test these hypotheses using a graph-based abstraction of inferred genetic-exchange relationships among 27 Escherichia coli and Shigella genomes. We show that although barriers to inter-phylogenetic group lateral transfer are low, E. coli and Shigella are more likely to have exchanged genetic material with close relatives. We find little evidence of bias arising from shared environment or lifestyle. More than one-third of donor-recipient pairs in our analysis show some level of fragmentary gene transfer. Thus, within the E. coli-Shigella clade, intact genes and gene fragments have been disseminated non-uniformly and at appreciable frequency, constructing communities that transgress environmental and lifestyle boundaries.
Collapse
Affiliation(s)
- Elizabeth Skippington
- Institute for Molecular Bioscience and Australian Research Council Centre of Excellence in Bioinformatics, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | |
Collapse
|
182
|
The KUPNetViz: a biological network viewer for multiple -omics datasets in kidney diseases. BMC Bioinformatics 2013; 14:235. [PMID: 23883183 PMCID: PMC3725151 DOI: 10.1186/1471-2105-14-235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 07/21/2013] [Indexed: 02/08/2023] Open
Abstract
Background Constant technological advances have allowed scientists in biology to migrate from conventional single-omics to multi-omics experimental approaches, challenging bioinformatics to bridge this multi-tiered information. Ongoing research in renal biology is no exception. The results of large-scale and/or high throughput experiments, presenting a wealth of information on kidney disease are scattered across the web. To tackle this problem, we recently presented the KUPKB, a multi-omics data repository for renal diseases. Results In this article, we describe KUPNetViz, a biological graph exploration tool allowing the exploration of KUPKB data through the visualization of biomolecule interactions. KUPNetViz enables the integration of multi-layered experimental data over different species, renal locations and renal diseases to protein-protein interaction networks and allows association with biological functions, biochemical pathways and other functional elements such as miRNAs. KUPNetViz focuses on the simplicity of its usage and the clarity of resulting networks by reducing and/or automating advanced functionalities present in other biological network visualization packages. In addition, it allows the extrapolation of biomolecule interactions across different species, leading to the formulations of new plausible hypotheses, adequate experiment design and to the suggestion of novel biological mechanisms. We demonstrate the value of KUPNetViz by two usage examples: the integration of calreticulin as a key player in a larger interaction network in renal graft rejection and the novel observation of the strong association of interleukin-6 with polycystic kidney disease. Conclusions The KUPNetViz is an interactive and flexible biological network visualization and exploration tool. It provides renal biologists with biological network snapshots of the complex integrated data of the KUPKB allowing the formulation of new hypotheses in a user friendly manner.
Collapse
|
183
|
Chung BKS, Dick T, Lee DY. In silico analyses for the discovery of tuberculosis drug targets. J Antimicrob Chemother 2013; 68:2701-9. [DOI: 10.1093/jac/dkt273] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
184
|
Wang J, Peng X, Li M, Pan Y. Construction and application of dynamic protein interaction network based on time course gene expression data. Proteomics 2013; 13:301-12. [PMID: 23225755 DOI: 10.1002/pmic.201200277] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 11/19/2012] [Accepted: 11/22/2012] [Indexed: 01/22/2023]
Abstract
In recent years, researchers have tried to inject dynamic information into static protein interaction networks (PINs). The paper first proposes a three-sigma method to identify active time points of each protein in a cellular cycle, where three-sigma principle is used to compute an active threshold for each gene according to the characteristics of its expression curve. Then a dynamic protein interaction network (DPIN) is constructed, which includes the dynamic changes of protein interactions. To validate the efficiency of DPIN, MCL, CPM, and core attachment algorithms are applied on two different DPINs, the static PIN and the time course PIN (TC-PIN) to detect protein complexes. The performance of each algorithm on DPINs outperforms those on other networks in terms of matching with known complexes, sensitivity, specificity, f-measure, and accuracy. Furthermore, the statistics of three-sigma principle show that 23-45% proteins are active at a time point and most proteins are active in about half of cellular cycle. In addition, we find 94% essential proteins are in the group of proteins that are active at equal or great than 12 timepoints of GSE4987, which indicates the potential existence of feedback mechanisms that can stabilize the expression level of essential proteins and might provide a new insight for predicting essential proteins from dynamic protein networks.
Collapse
Affiliation(s)
- Jianxin Wang
- School of Information Science and Engineering, Central South University, Changsha, China.
| | | | | | | |
Collapse
|
185
|
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: 512] [Impact Index Per Article: 46.5] [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
|
186
|
Demeyer S, Michoel T, Fostier J, Audenaert P, Pickavet M, Demeester P. The index-based subgraph matching algorithm (ISMA): fast subgraph enumeration in large networks using optimized search trees. PLoS One 2013; 8:e61183. [PMID: 23620730 PMCID: PMC3631255 DOI: 10.1371/journal.pone.0061183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 03/05/2013] [Indexed: 11/28/2022] Open
Abstract
Subgraph matching algorithms are designed to find all instances of predefined subgraphs in a large graph or network and play an important role in the discovery and analysis of so-called network motifs, subgraph patterns which occur more often than expected by chance. We present the index-based subgraph matching algorithm (ISMA), a novel tree-based algorithm. ISMA realizes a speedup compared to existing algorithms by carefully selecting the order in which the nodes of a query subgraph are investigated. In order to achieve this, we developed a number of data structures and maximally exploited symmetry characteristics of the subgraph. We compared ISMA to a naive recursive tree-based algorithm and to a number of well-known subgraph matching algorithms. Our algorithm outperforms the other algorithms, especially on large networks and with large query subgraphs. An implementation of ISMA in Java is freely available at http://sourceforge.net/projects/isma/.
Collapse
Affiliation(s)
- Sofie Demeyer
- Department of Information Technology, Ghent University, Ghent, Belgium.
| | | | | | | | | | | |
Collapse
|
187
|
Reddy PS, Murray S, Liu W. Knowledge-Driven, Data-Assisted Integrative Pathway Analytics. Bioinformatics 2013. [DOI: 10.4018/978-1-4666-3604-0.ch009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Target and biomarker selection in drug discovery relies extensively on the use of various genomics platforms. These technologies generate large amounts of data that can be used to gain novel insights in biology. There is a strong need to mine these information-rich datasets in an effective and efficient manner. Pathway and network based approaches have become an increasingly important methodology to mine bioinformatics datasets derived from ‘omics’ technologies. These approaches also find use in exploring the unknown biology of a disease or functional process. This chapter provides an overview of pathway databases and network tools, network architecture, text mining and existing methods used in knowledge-driven data analysis. It shows examples of how these databases and tools can be used integratively to apply existing knowledge and network-based approach in data analytics.
Collapse
Affiliation(s)
| | | | - Wei Liu
- Agios Pharmaceuticals Inc, USA
| |
Collapse
|
188
|
Furlong LI. Human diseases through the lens of network biology. Trends Genet 2013; 29:150-9. [DOI: 10.1016/j.tig.2012.11.004] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 10/24/2012] [Accepted: 11/09/2012] [Indexed: 12/13/2022]
|
189
|
Reyes-Palomares A, Rodríguez-López R, Ranea JAG, Jiménez FS, Medina MA. Global analysis of the human pathophenotypic similarity gene network merges disease module components. PLoS One 2013; 8:e56653. [PMID: 23437198 PMCID: PMC3578923 DOI: 10.1371/journal.pone.0056653] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 01/12/2013] [Indexed: 12/22/2022] Open
Abstract
The molecular complexity of genetic diseases requires novel approaches to break it down into coherent biological modules. For this purpose, many disease network models have been created and analyzed. We highlight two of them, "the human diseases networks" (HDN) and "the orphan disease networks" (ODN). However, in these models, each single node represents one disease or an ambiguous group of diseases. In these cases, the notion of diseases as unique entities reduces the usefulness of network-based methods. We hypothesize that using the clinical features (pathophenotypes) to define pathophenotypic connections between disease-causing genes improve our understanding of the molecular events originated by genetic disturbances. For this, we have built a pathophenotypic similarity gene network (PSGN) and compared it with the unipartite projections (based on gene-to-gene edges) similar to those used in previous network models (HDN and ODN). Unlike these disease network models, the PSGN uses semantic similarities. This pathophenotypic similarity has been calculated by comparing pathophenotypic annotations of genes (human abnormalities of HPO terms) in the "Human Phenotype Ontology". The resulting network contains 1075 genes (nodes) and 26197 significant pathophenotypic similarities (edges). A global analysis of this network reveals: unnoticed pairs of genes showing significant pathophenotypic similarity, a biological meaningful re-arrangement of the pathological relationships between genes, correlations of biochemical interactions with higher similarity scores and functional biases in metabolic and essential genes toward the pathophenotypic specificity and the pleiotropy, respectively. Additionally, pathophenotypic similarities and metabolic interactions of genes associated with maple syrup urine disease (MSUD) have been used to merge into a coherent pathological module.Our results indicate that pathophenotypes contribute to identify underlying co-dependencies among disease-causing genes that are useful to describe disease modularity.
Collapse
Affiliation(s)
- Armando Reyes-Palomares
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Málaga, Spain
| | - Rocío Rodríguez-López
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Málaga, Spain
| | - Juan A. G. Ranea
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Málaga, Spain
| | - Francisca Sánchez Jiménez
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Málaga, Spain
| | - Miguel Angel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Málaga, Spain
| |
Collapse
|
190
|
Ruksha TG, Aksenenko MB, Sergeyeva YY, Fefelova YA. Skin melanoma: from systematic biology to the personalized therapy. VESTNIK DERMATOLOGII I VENEROLOGII 2013. [DOI: 10.25208/vdv555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Systematic biology is a new field of biomedicine based on the integrative approach to molecular mechanisms of the operation of living systems including in case of the development of pathological processes. In this connection, up-to-date therapeutic approaches to skin melanoma treatment can be considered on the basis of key changes in intermolecular interactions taking place during tumor development.
Collapse
|
191
|
Charos AE, Reed BD, Raha D, Szekely AM, Weissman SM, Snyder M. A highly integrated and complex PPARGC1A transcription factor binding network in HepG2 cells. Genome Res 2013; 22:1668-79. [PMID: 22955979 PMCID: PMC3431484 DOI: 10.1101/gr.127761.111] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PPARGC1A is a transcriptional coactivator that binds to and coactivates a variety of transcription factors (TFs) to regulate the expression of target genes. PPARGC1A plays a pivotal role in regulating energy metabolism and has been implicated in several human diseases, most notably type II diabetes. Previous studies have focused on the interplay between PPARGC1A and individual TFs, but little is known about how PPARGC1A combines with all of its partners across the genome to regulate transcriptional dynamics. In this study, we describe a core PPARGC1A transcriptional regulatory network operating in HepG2 cells treated with forskolin. We first mapped the genome-wide binding sites of PPARGC1A using chromatin-IP followed by high-throughput sequencing (ChIP-seq) and uncovered overrepresented DNA sequence motifs corresponding to known and novel PPARGC1A network partners. We then profiled six of these site-specific TF partners using ChIP-seq and examined their network connectivity and combinatorial binding patterns with PPARGC1A. Our analysis revealed extensive overlap of targets including a novel link between PPARGC1A and HSF1, a TF regulating the conserved heat shock response pathway that is misregulated in diabetes. Importantly, we found that different combinations of TFs bound to distinct functional sets of genes, thereby helping to reveal the combinatorial regulatory code for metabolic and other cellular processes. In addition, the different TFs often bound near the promoters and coding regions of each other's genes suggesting an intricate network of interdependent regulation. Overall, our study provides an important framework for understanding the systems-level control of metabolic gene expression in humans.
Collapse
Affiliation(s)
- Alexandra E Charos
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06520, USA
| | | | | | | | | | | |
Collapse
|
192
|
Ghersi D, Singh M. Disentangling function from topology to infer the network properties of disease genes. BMC SYSTEMS BIOLOGY 2013; 7:5. [PMID: 23324116 PMCID: PMC3614482 DOI: 10.1186/1752-0509-7-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/04/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND The topological features of disease genes within interaction networks are the subject of intense study, as they shed light on common mechanisms of pathology and are useful for uncovering additional disease genes. Computational analyses typically try to uncover whether disease genes exhibit distinct network features, as compared to all genes. RESULTS We demonstrate that the functional composition of disease gene sets is an important confounding factor in these types of analyses. We consider five disease sets and show that while they indeed have distinct topological features, they are also enriched in functions that a priori exhibit distinct network properties. To address this, we develop a computational framework to assess the network properties of disease genes based on a sampling algorithm that generates control gene sets that are functionally similar to the disease set. Using our function-constrained sampling approach, we demonstrate that for most of the topological properties studied, disease genes are more similar to sets of genes with similar functional make-up than they are to randomly selected genes; this suggests that these observed differences in topological properties reflect not only the distinguishing network features of disease genes but also their functional composition. Nevertheless, we also highlight many cases where disease genes have distinct topological properties even when accounting for function. CONCLUSIONS Our approach is an important first step in extracting the residual topological differences in disease genes when accounting for function, and leads to new insights into the network properties of disease genes.
Collapse
Affiliation(s)
- Dario Ghersi
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | | |
Collapse
|
193
|
Pérez-Bercoff Å, Hudson CM, Conant GC. A conserved mammalian protein interaction network. PLoS One 2013; 8:e52581. [PMID: 23320073 PMCID: PMC3539715 DOI: 10.1371/journal.pone.0052581] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 11/20/2012] [Indexed: 11/19/2022] Open
Abstract
Physical interactions between proteins mediate a variety of biological functions, including signal transduction, physical structuring of the cell and regulation. While extensive catalogs of such interactions are known from model organisms, their evolutionary histories are difficult to study given the lack of interaction data from phylogenetic outgroups. Using phylogenomic approaches, we infer a upper bound on the time of origin for a large set of human protein-protein interactions, showing that most such interactions appear relatively ancient, dating no later than the radiation of placental mammals. By analyzing paired alignments of orthologous and putatively interacting protein-coding genes from eight mammals, we find evidence for weak but significant co-evolution, as measured by relative selective constraint, between pairs of genes with interacting proteins. However, we find no strong evidence for shared instances of directional selection within an interacting pair. Finally, we use a network approach to show that the distribution of selective constraint across the protein interaction network is non-random, with a clear tendency for interacting proteins to share similar selective constraints. Collectively, the results suggest that, on the whole, protein interactions in mammals are under selective constraint, presumably due to their functional roles.
Collapse
Affiliation(s)
- Åsa Pérez-Bercoff
- Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, Ireland
| | - Corey M. Hudson
- Informatics Institute, University of Missouri, Columbia, Missouri, United States of America
| | - Gavin C. Conant
- Informatics Institute, University of Missouri, Columbia, Missouri, United States of America
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
194
|
Kim KJ, Hwang D, Kim WU. Systems Approach to Rheumatoid Arthritis. JOURNAL OF RHEUMATIC DISEASES 2013. [DOI: 10.4078/jrd.2013.20.6.348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Ki-Jo Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| | - Daehee Hwang
- Center for Systems Biology of Plant Senescence and Life History, Daegu Gyeongbuk Institute of Science & Technology, Daegu, Korea
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea
| |
Collapse
|
195
|
Calabrese G, Bennett BJ, Orozco L, Kang HM, Eskin E, Dombret C, De Backer O, Lusis AJ, Farber CR. Systems genetic analysis of osteoblast-lineage cells. PLoS Genet 2012; 8:e1003150. [PMID: 23300464 PMCID: PMC3531492 DOI: 10.1371/journal.pgen.1003150] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 10/23/2012] [Indexed: 12/20/2022] Open
Abstract
The osteoblast-lineage consists of cells at various stages of maturation that are essential for skeletal development, growth, and maintenance. Over the past decade, many of the signaling cascades that regulate this lineage have been elucidated; however, little is known of the networks that coordinate, modulate, and transmit these signals. Here, we identify a gene network specific to the osteoblast-lineage through the reconstruction of a bone co-expression network using microarray profiles collected on 96 Hybrid Mouse Diversity Panel (HMDP) inbred strains. Of the 21 modules that comprised the bone network, module 9 (M9) contained genes that were highly correlated with prototypical osteoblast maker genes and were more highly expressed in osteoblasts relative to other bone cells. In addition, the M9 contained many of the key genes that define the osteoblast-lineage, which together suggested that it was specific to this lineage. To use the M9 to identify novel osteoblast genes and highlight its biological relevance, we knocked-down the expression of its two most connected “hub” genes, Maged1 and Pard6g. Their perturbation altered both osteoblast proliferation and differentiation. Furthermore, we demonstrated the mice deficient in Maged1 had decreased bone mineral density (BMD). It was also discovered that a local expression quantitative trait locus (eQTL) regulating the Wnt signaling antagonist Sfrp1 was a key driver of the M9. We also show that the M9 is associated with BMD in the HMDP and is enriched for genes implicated in the regulation of human BMD through genome-wide association studies. In conclusion, we have identified a physiologically relevant gene network and used it to discover novel genes and regulatory mechanisms involved in the function of osteoblast-lineage cells. Our results highlight the power of harnessing natural genetic variation to generate co-expression networks that can be used to gain insight into the function of specific cell-types. The osteoblast-lineage consists of a range of cells from osteogenic precursors that mature into bone-forming osteoblasts to osteocytes that are entombed in bone. Each cell in the lineage serves a number of distinct and critical roles in the growth and maintenance of the skeleton, as well as many extra-skeletal functions. Over the last decade, many of the major regulatory pathways governing the differentiation and activity of these cells have been discovered. In contrast, little is known regarding the composition or function of gene networks within the lineage. The goal of this study was to increase our understanding of how genes are organized into networks in osteoblasts. Towards this goal, we used microarray gene expression profiles from bone to identify a group of genes that formed a network specific to the osteoblast-lineage. We used the knowledge of this network to identify novel genes that are important for regulating various aspects of osteoblast function. These data improve our understanding of the gene networks operative in cells of the osteoblast-lineage.
Collapse
Affiliation(s)
- Gina Calabrese
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Brian J. Bennett
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Luz Orozco
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hyun M. Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eleazar Eskin
- Department of Computer Science, University of California Los Angeles, Los Angeles, California, United States of America
| | - Carlos Dombret
- Unité de Recherche en Physiologie Moléculaire (URPHYM), Namur Research Institute for Life Sciences (NARILIS), FUNDP School of Medicine, University of Namur, Namur, Belgium
| | - Olivier De Backer
- Unité de Recherche en Physiologie Moléculaire (URPHYM), Namur Research Institute for Life Sciences (NARILIS), FUNDP School of Medicine, University of Namur, Namur, Belgium
| | - Aldons J. Lusis
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, California, United States of America
| | - Charles R. Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
196
|
Alawieh A, Zaraket FA, Li JL, Mondello S, Nokkari A, Razafsha M, Fadlallah B, Boustany RM, Kobeissy FH. Systems biology, bioinformatics, and biomarkers in neuropsychiatry. Front Neurosci 2012; 6:187. [PMID: 23269912 PMCID: PMC3529307 DOI: 10.3389/fnins.2012.00187] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 12/06/2012] [Indexed: 11/13/2022] Open
Abstract
Although neuropsychiatric (NP) disorders are among the top causes of disability worldwide with enormous financial costs, they can still be viewed as part of the most complex disorders that are of unknown etiology and incomprehensible pathophysiology. The complexity of NP disorders arises from their etiologic heterogeneity and the concurrent influence of environmental and genetic factors. In addition, the absence of rigid boundaries between the normal and diseased state, the remarkable overlap of symptoms among conditions, the high inter-individual and inter-population variations, and the absence of discriminative molecular and/or imaging biomarkers for these diseases makes difficult an accurate diagnosis. Along with the complexity of NP disorders, the practice of psychiatry suffers from a "top-down" method that relied on symptom checklists. Although checklist diagnoses cost less in terms of time and money, they are less accurate than a comprehensive assessment. Thus, reliable and objective diagnostic tools such as biomarkers are needed that can detect and discriminate among NP disorders. The real promise in understanding the pathophysiology of NP disorders lies in bringing back psychiatry to its biological basis in a systemic approach which is needed given the NP disorders' complexity to understand their normal functioning and response to perturbation. This approach is implemented in the systems biology discipline that enables the discovery of disease-specific NP biomarkers for diagnosis and therapeutics. Systems biology involves the use of sophisticated computer software "omics"-based discovery tools and advanced performance computational techniques in order to understand the behavior of biological systems and identify diagnostic and prognostic biomarkers specific for NP disorders together with new targets of therapeutics. In this review, we try to shed light on the need of systems biology, bioinformatics, and biomarkers in neuropsychiatry, and illustrate how the knowledge gained through these methodologies can be translated into clinical use providing clinicians with improved ability to diagnose, manage, and treat NP patients.
Collapse
Affiliation(s)
- Ali Alawieh
- Department of Biochemistry, College of Medicine, American University of Beirut Beirut, Lebanon
| | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Affiliation(s)
- Xiaoke Ma
- School of Computer Science and Technology, Xidian University, No. 2 South TaiBai Road, Xi'an, Shaanxi 710071, P.R. China
| | | |
Collapse
|
198
|
Jamil K, Jayaraman A, Rao R, Raju S. In silico evidence of signaling pathways of notch mediated networks in leukemia. Comput Struct Biotechnol J 2012; 1:e201207005. [PMID: 24688641 PMCID: PMC3962152 DOI: 10.5936/csbj.201207005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 11/04/2012] [Accepted: 11/07/2012] [Indexed: 11/22/2022] Open
Abstract
Notch signaling plays a critical role in cell fate determination and maintenance of progenitors in many developmental systems. Notch receptors have been shown to be expressed on hematopoietic progenitor cells as well as to various degrees in peripheral blood T and B lymphocytes, monocytes, and neutrophils. Our aim was to understand the protein interaction network, using Notch1 protein name as query in STRING database and we generated a model to assess the significance of Notch1 associated proteins in Acute Lymphoblastic Leukemia (ALL). We further analyzed the expression levels of the genes encoding hub proteins, using Oncomine database, to determine their significance in leukemogenesis. Of the forty two hub genes, we observed that sixteen genes were underexpressed and eleven genes were overexpressed in T-cell Acute Lymphoblastic samples in comparison to their expression levels in normal cells. Of these, we found three novel genes which have not been reported earlier- KAT2B, PSEN1 (underexpressed) and CDH2 (overexpressed).These three identified genes may provide new insights into the abnormal hematopoietic process observed in Leukemia as these genes are involved in Notch signaling and cell adhesion processes. It is evident that experimental validation of the protein interactors in leukemic cells could help in the identification of new diagnostic markers for leukemia.
Collapse
Affiliation(s)
- Kaiser Jamil
- Centre for Biotechnology and Bioinformatics, School of Life sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), 6th Floor, Budha Bhawan, M.G. Road, Secunderabad 500003, Andhra Pradesh, India
| | - Archana Jayaraman
- Centre for Biotechnology and Bioinformatics, School of Life sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), 6th Floor, Budha Bhawan, M.G. Road, Secunderabad 500003, Andhra Pradesh, India
| | - Raghunatha Rao
- Oncology Department, Nizams Institute of Medical Sciences ( NIMS), Panjagutta, Hyderabad 500082, Andhra Pradesh, India
| | - Suryanarayana Raju
- Oncology Department, Nizams Institute of Medical Sciences ( NIMS), Panjagutta, Hyderabad 500082, Andhra Pradesh, India
| |
Collapse
|
199
|
Gaspar ME, Csermely P. Rigidity and flexibility of biological networks. Brief Funct Genomics 2012; 11:443-56. [DOI: 10.1093/bfgp/els023] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
200
|
Affiliation(s)
- Nagasuma Chandra
- Indian Institute of Science, Department of Biochemistry,
Bangalore – 560012, India ,
| | - Jyothi Padiadpu
- Indian Institute of Science, Department of Biochemistry,
Bangalore – 560012, India
| |
Collapse
|