201
|
Xu C, Nikolova O, Basom RS, Mitchell RM, Shaw R, Moser RD, Park H, Gurley KE, Kao MC, Green CL, Schaub FX, Diaz RL, Swan HA, Jang IS, Guinney J, Gadi VK, Margolin AA, Grandori C, Kemp CJ, Méndez E. Functional Precision Medicine Identifies Novel Druggable Targets and Therapeutic Options in Head and Neck Cancer. Clin Cancer Res 2018; 24:2828-2843. [PMID: 29599409 DOI: 10.1158/1078-0432.ccr-17-1339] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/06/2017] [Accepted: 03/20/2018] [Indexed: 01/07/2023]
Abstract
Purpose: Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, with high mortality and a lack of targeted therapies. To identify and prioritize druggable targets, we performed genome analysis together with genome-scale siRNA and oncology drug profiling using low-passage tumor cells derived from a patient with treatment-resistant HPV-negative HNSCC.Experimental Design: A tumor cell culture was established and subjected to whole-exome sequencing, RNA sequencing, comparative genome hybridization, and high-throughput phenotyping with a siRNA library covering the druggable genome and an oncology drug library. Secondary screens of candidate target genes were performed on the primary tumor cells and two nontumorigenic keratinocyte cell cultures for validation and to assess cancer specificity. siRNA screens of the kinome on two isogenic pairs of p53-mutated HNSCC cell lines were used to determine generalizability. Clinical utility was addressed by performing drug screens on two additional HNSCC cell cultures derived from patients enrolled in a clinical trial.Results: Many of the identified copy number aberrations and somatic mutations in the primary tumor were typical of HPV(-) HNSCC, but none pointed to obvious therapeutic choices. In contrast, siRNA profiling identified 391 candidate target genes, 35 of which were preferentially lethal to cancer cells, most of which were not genomically altered. Chemotherapies and targeted agents with strong tumor-specific activities corroborated the siRNA profiling results and included drugs that targeted the mitotic spindle, the proteasome, and G2-M kinases WEE1 and CHK1 We also show the feasibility of ex vivo drug profiling for patients enrolled in a clinical trial.Conclusions: High-throughput phenotyping with siRNA and drug libraries using patient-derived tumor cells prioritizes mutated driver genes and identifies novel drug targets not revealed by genomic profiling. Functional profiling is a promising adjunct to DNA sequencing for precision oncology. Clin Cancer Res; 24(12); 2828-43. ©2018 AACR.
Collapse
Affiliation(s)
- Chang Xu
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Olga Nikolova
- Computational Biology Program, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Ryan S Basom
- Shared Resources, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ryan M Mitchell
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, Washington
| | | | - Russell D Moser
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Heuijoon Park
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kay E Gurley
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Michael C Kao
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Carlos L Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | | | - In S Jang
- Sage Bionetworks, Seattle, Washington
| | | | - Vijayakrishna K Gadi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Seattle Cancer Care Alliance, Seattle, Washington
| | - Adam A Margolin
- Computational Biology Program, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | | | - Christopher J Kemp
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Eduardo Méndez
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Seattle Cancer Care Alliance, Seattle, Washington
| |
Collapse
|
202
|
Buljan M, Blattmann P, Aebersold R, Boutros M. Systematic characterization of pan-cancer mutation clusters. Mol Syst Biol 2018; 14:e7974. [PMID: 29572294 PMCID: PMC5866917 DOI: 10.15252/msb.20177974] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer genome sequencing has shown that driver genes can often be distinguished not only by the elevated mutation frequency but also by specific nucleotide positions that accumulate changes at a high rate. However, properties associated with a residue's potential to drive tumorigenesis when mutated have not yet been systematically investigated. Here, using a novel methodological approach, we identify and characterize a compendium of 180 hotspot residues within 160 human proteins which occur with a significant frequency and are likely to have functionally relevant impact. We find that such mutations (i) are more prominent in proteins that can exist in the on and off state, (ii) reflect the identity of a tumor of origin, and (iii) often localize within interfaces which mediate interactions with other proteins or ligands. Following, we further examine structural data for human protein complexes and identify a number of additional protein interfaces that accumulate cancer mutations at a high rate. Jointly, these analyses suggest that disruption and dysregulation of protein interactions can be instrumental in switching functions of cancer proteins and activating downstream changes.
Collapse
Affiliation(s)
- Marija Buljan
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Blattmann
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland .,Faculty of Science, University of Zurich, Zurich, Switzerland
| | - Michael Boutros
- Division Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany .,Department Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg University, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
203
|
Gao J, Mfuh A, Amako Y, Woo CM. Small Molecule Interactome Mapping by Photoaffinity Labeling Reveals Binding Site Hotspots for the NSAIDs. J Am Chem Soc 2018. [DOI: 10.1021/jacs.7b11639] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jinxu Gao
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Adelphe Mfuh
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Yuka Amako
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, United States
| | - Christina M. Woo
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St., Cambridge, Massachusetts 02138, United States
| |
Collapse
|
204
|
Nikolova O, Moser R, Kemp C, Gönen M, Margolin AA. Modeling gene-wise dependencies improves the identification of drug response biomarkers in cancer studies. Bioinformatics 2018; 33:1362-1369. [PMID: 28082455 DOI: 10.1093/bioinformatics/btw836] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/29/2016] [Indexed: 01/07/2023] Open
Abstract
Motivation In recent years, vast advances in biomedical technologies and comprehensive sequencing have revealed the genomic landscape of common forms of human cancer in unprecedented detail. The broad heterogeneity of the disease calls for rapid development of personalized therapies. Translating the readily available genomic data into useful knowledge that can be applied in the clinic remains a challenge. Computational methods are needed to aid these efforts by robustly analyzing genome-scale data from distinct experimental platforms for prioritization of targets and treatments. Results We propose a novel, biologically motivated, Bayesian multitask approach, which explicitly models gene-centric dependencies across multiple and distinct genomic platforms. We introduce a gene-wise prior and present a fully Bayesian formulation of a group factor analysis model. In supervised prediction applications, our multitask approach leverages similarities in response profiles of groups of drugs that are more likely to be related to true biological signal, which leads to more robust performance and improved generalization ability. We evaluate the performance of our method on molecularly characterized collections of cell lines profiled against two compound panels, namely the Cancer Cell Line Encyclopedia and the Cancer Therapeutics Response Portal. We demonstrate that accounting for the gene-centric dependencies enables leveraging information from multi-omic input data and improves prediction and feature selection performance. We further demonstrate the applicability of our method in an unsupervised dimensionality reduction application by inferring genes essential to tumorigenesis in the pancreatic ductal adenocarcinoma and lung adenocarcinoma patient cohorts from The Cancer Genome Atlas. Availability and Implementation : The code for this work is available at https://github.com/olganikolova/gbgfa. Contact : nikolova@ohsu.edu or margolin@ohsu.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Olga Nikolova
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA
| | - Russell Moser
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Christopher Kemp
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mehmet Gönen
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA.,Department of Industrial Engineering, Koç University, İstanbul, Turkey
| | - Adam A Margolin
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
205
|
Llano M, Peña-Hernandez MA. Defining Pharmacological Targets by Analysis of Virus-Host Protein Interactions. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 111:223-242. [PMID: 29459033 PMCID: PMC6322211 DOI: 10.1016/bs.apcsb.2017.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Viruses are obligate parasites that depend on cellular factors for replication. Pharmacological inhibition of essential viral proteins, mostly enzymes, is an effective therapeutic alternative in the absence of effective vaccines. However, this strategy commonly encounters drug resistance mechanisms that allow these pathogens to evade control. Due to the dependency on host factors for viral replication, pharmacological disruption of the host-pathogen protein-protein interactions (PPIs) is an important therapeutic alternative to block viral replication. In this review we discuss salient aspects of PPIs implicated in viral replication and advances in the development of small molecules that inhibit viral replication through antagonism of these interactions.
Collapse
Affiliation(s)
- Manuel Llano
- University of Texas at El Paso, El Paso, TX, United States.
| | - Mario A Peña-Hernandez
- University of Texas at El Paso, El Paso, TX, United States; Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolas de los Garza, Mexico
| |
Collapse
|
206
|
Rauscher B, Heigwer F, Henkel L, Hielscher T, Voloshanenko O, Boutros M. Toward an integrated map of genetic interactions in cancer cells. Mol Syst Biol 2018; 14:e7656. [PMID: 29467179 PMCID: PMC5820685 DOI: 10.15252/msb.20177656] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 01/20/2018] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
Cancer genomes often harbor hundreds of molecular aberrations. Such genetic variants can be drivers or passengers of tumorigenesis and create vulnerabilities for potential therapeutic exploitation. To identify genotype-dependent vulnerabilities, forward genetic screens in different genetic backgrounds have been conducted. We devised MINGLE, a computational framework to integrate CRISPR/Cas9 screens originating from different libraries building on approaches pioneered for genetic network discovery in model organisms. We applied this method to integrate and analyze data from 85 CRISPR/Cas9 screens in human cancer cells combining functional data with information on genetic variants to explore more than 2.1 million gene-background relationships. In addition to known dependencies, we identified new genotype-specific vulnerabilities of cancer cells. Experimental validation of predicted vulnerabilities identified GANAB and PRKCSH as new positive regulators of Wnt/β-catenin signaling. By clustering genes with similar genetic interaction profiles, we drew the largest genetic network in cancer cells to date. Our scalable approach highlights how diverse genetic screens can be integrated to systematically build informative maps of genetic interactions in cancer, which can grow dynamically as more data are included.
Collapse
Affiliation(s)
- Benedikt Rauscher
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Florian Heigwer
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Luisa Henkel
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oksana Voloshanenko
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
207
|
Abstract
A better understanding of proteostasis in health and disease requires robust methods to determine protein half-lives. Here we improve the precision and accuracy of peptide ion intensity-based quantification, enabling more accurate protein turnover determination in non-dividing cells by dynamic SILAC-based proteomics. This approach allows exact determination of protein half-lives ranging from 10 to >1000 h. We identified 4000-6000 proteins in several non-dividing cell types, corresponding to 9699 unique protein identifications over the entire data set. We observed similar protein half-lives in B-cells, natural killer cells and monocytes, whereas hepatocytes and mouse embryonic neurons show substantial differences. Our data set extends and statistically validates the previous observation that subunits of protein complexes tend to have coherent turnover. Moreover, analysis of different proteasome and nuclear pore complex assemblies suggests that their turnover rate is architecture dependent. These results illustrate that our approach allows investigating protein turnover and its implications in various cell types.
Collapse
|
208
|
Tan CSH, Go KD, Bisteau X, Dai L, Yong CH, Prabhu N, Ozturk MB, Lim YT, Sreekumar L, Lengqvist J, Tergaonkar V, Kaldis P, Sobota RM, Nordlund P. Thermal proximity coaggregation for system-wide profiling of protein complex dynamics in cells. Science 2018; 359:1170-1177. [DOI: 10.1126/science.aan0346] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 09/28/2017] [Accepted: 01/27/2018] [Indexed: 01/20/2023]
|
209
|
Liu F, Lössl P, Rabbitts BM, Balaban RS, Heck AJR. The interactome of intact mitochondria by cross-linking mass spectrometry provides evidence for coexisting respiratory supercomplexes. Mol Cell Proteomics 2018; 17:216-232. [PMID: 29222160 PMCID: PMC5795388 DOI: 10.1074/mcp.ra117.000470] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Indexed: 12/22/2022] Open
Abstract
Mitochondria exert an immense amount of cytophysiological functions, but the structural basis of most of these processes is still poorly understood. Here we use cross-linking mass spectrometry to probe the organization of proteins in native mouse heart mitochondria. Our approach provides the largest survey of mitochondrial protein interactions reported so far. In total, we identify 3,322 unique residue-to-residue contacts involving half of the mitochondrial proteome detected by bottom-up proteomics. The obtained mitochondrial protein interactome gives insights in the architecture and submitochondrial localization of defined protein assemblies, and reveals the mitochondrial localization of four proteins not yet included in the MitoCarta database. As one of the highlights, we show that the oxidative phosphorylation complexes I-V exist in close spatial proximity, providing direct evidence for supercomplex assembly in intact mitochondria. The specificity of these contacts is demonstrated by comparative analysis of mitochondria after high salt treatment, which disrupts the native supercomplexes and substantially changes the mitochondrial interactome.
Collapse
Affiliation(s)
- Fan Liu
- From the ‡Biomolecular Mass Spectrometry and Proteomics. Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584 CH, Utrecht, The Netherlands
- §Netherlands Proteomics Center, Padualaan 8, 3584 CH, Utrecht, The Netherlands
- ¶Leibniz Institute of Molecular Pharmacology (FMP Berlin), Robert-Rössle-Straβe 10, 13125 Berlin, Germany
| | - Philip Lössl
- From the ‡Biomolecular Mass Spectrometry and Proteomics. Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584 CH, Utrecht, The Netherlands
- §Netherlands Proteomics Center, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Beverley M Rabbitts
- ‖Laboratory of Cardiac Energetics, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Robert S Balaban
- ‖Laboratory of Cardiac Energetics, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Albert J R Heck
- From the ‡Biomolecular Mass Spectrometry and Proteomics. Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584 CH, Utrecht, The Netherlands;
- §Netherlands Proteomics Center, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| |
Collapse
|
210
|
Adler A, Kirchmeier P, Reinhard J, Brauner B, Dunger I, Fobo G, Frishman G, Montrone C, Mewes HW, Arnold M, Ruepp A. PhenoDis: a comprehensive database for phenotypic characterization of rare cardiac diseases. Orphanet J Rare Dis 2018; 13:22. [PMID: 29370821 PMCID: PMC5785853 DOI: 10.1186/s13023-018-0765-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/12/2018] [Indexed: 12/31/2022] Open
Abstract
Background Thoroughly annotated data resources are a key requirement in phenotype dependent analysis and diagnosis of diseases in the area of precision medicine. Recent work has shown that curation and systematic annotation of human phenome data can significantly improve the quality and selectivity for the interpretation of inherited diseases. We have therefore developed PhenoDis, a comprehensive, manually annotated database providing symptomatic, genetic and imprinting information about rare cardiac diseases. Results PhenoDis includes 214 rare cardiac diseases from Orphanet and 94 more from OMIM. For phenotypic characterization of the diseases, we performed manual annotation of diseases with articles from the biomedical literature. Detailed description of disease symptoms required the use of 2247 different terms from the Human Phenotype Ontology (HPO). Diseases listed in PhenoDis frequently cover a broad spectrum of symptoms with 28% from the branch of ‘cardiovascular abnormality’ and others from areas such as neurological (11.5%) and metabolism (6%). We collected extensive information on the frequency of symptoms in respective diseases as well as on disease-associated genes and imprinting data. The analysis of the abundance of symptoms in patient studies revealed that most of the annotated symptoms (71%) are found in less than half of the patients of a particular disease. Comprehensive and systematic characterization of symptoms including their frequency is a pivotal prerequisite for computer based prediction of diseases and disease causing genetic variants. To this end, PhenoDis provides in-depth annotation for a complete group of rare diseases, including information on pathogenic and likely pathogenic genetic variants for 206 diseases as listed in ClinVar. We integrated all results in an online database (http://mips.helmholtz-muenchen.de/phenodis/) with multiple search options and provide the complete dataset for download. Conclusion PhenoDis provides a comprehensive set of manually annotated rare cardiac diseases that enables computational approaches for disease prediction via decision support systems and phenotype-driven strategies for the identification of disease causing genes.
Collapse
Affiliation(s)
- Angela Adler
- Technische Universität München, Chair of Genome Oriented Bioinformatics, Center of Life and Food Science, D-85350, Freising-Weihenstephan, Germany
| | - Pia Kirchmeier
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - Julian Reinhard
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - Barbara Brauner
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - Irmtraud Dunger
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - Gisela Fobo
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - Goar Frishman
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - Corinna Montrone
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - H-Werner Mewes
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany.,Technische Universität München, Chair of Genome Oriented Bioinformatics, Center of Life and Food Science, D-85350, Freising-Weihenstephan, Germany
| | - Matthias Arnold
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany
| | - Andreas Ruepp
- Institute for Bioinformatics and Systems Biology (IBIS), Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), D-85764, Neuherberg, Germany.
| |
Collapse
|
211
|
Ribeiro DM, Zanzoni A, Cipriano A, Delli Ponti R, Spinelli L, Ballarino M, Bozzoni I, Tartaglia GG, Brun C. Protein complex scaffolding predicted as a prevalent function of long non-coding RNAs. Nucleic Acids Res 2018; 46:917-928. [PMID: 29165713 PMCID: PMC5778612 DOI: 10.1093/nar/gkx1169] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/03/2017] [Accepted: 11/07/2017] [Indexed: 11/14/2022] Open
Abstract
The human transcriptome contains thousands of long non-coding RNAs (lncRNAs). Characterizing their function is a current challenge. An emerging concept is that lncRNAs serve as protein scaffolds, forming ribonucleoproteins and bringing proteins in proximity. However, only few scaffolding lncRNAs have been characterized and the prevalence of this function is unknown. Here, we propose the first computational approach aimed at predicting scaffolding lncRNAs at large scale. We predicted the largest human lncRNA-protein interaction network to date using the catRAPID omics algorithm. In combination with tissue expression and statistical approaches, we identified 847 lncRNAs (∼5% of the long non-coding transcriptome) predicted to scaffold half of the known protein complexes and network modules. Lastly, we show that the association of certain lncRNAs to disease may involve their scaffolding ability. Overall, our results suggest for the first time that RNA-mediated scaffolding of protein complexes and modules may be a common mechanism in human cells.
Collapse
Affiliation(s)
- Diogo M Ribeiro
- Aix-Marseille Université, Inserm, TAGC UMR_S1090, Marseille, France
| | - Andreas Zanzoni
- Aix-Marseille Université, Inserm, TAGC UMR_S1090, Marseille, France
| | - Andrea Cipriano
- Dept. of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Riccardo Delli Ponti
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Lionel Spinelli
- Aix-Marseille Université, Inserm, TAGC UMR_S1090, Marseille, France
| | - Monica Ballarino
- Dept. of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Irene Bozzoni
- Dept. of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Gian Gaetano Tartaglia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- Institucio Catalana de Recerca i Estudis Avançats (ICREA), 23 Passeig Lluıs Companys, 08010 Barcelona, Spain
| | - Christine Brun
- Aix-Marseille Université, Inserm, TAGC UMR_S1090, Marseille, France
- CNRS, Marseille, France
| |
Collapse
|
212
|
Huang H, Luo Z, Qi S, Huang J, Xu P, Wang X, Gao L, Li F, Wang J, Zhao W, Gu W, Chen Z, Dai L, Dai J, Zhao Y. Landscape of the regulatory elements for lysine 2-hydroxyisobutyrylation pathway. Cell Res 2018; 28:111-125. [PMID: 29192674 PMCID: PMC5752845 DOI: 10.1038/cr.2017.149] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/02/2017] [Accepted: 08/16/2017] [Indexed: 02/05/2023] Open
Abstract
Short-chain fatty acids and their corresponding acyl-CoAs sit at the crossroads of metabolic pathways and play important roles in diverse cellular processes. They are also precursors for protein post-translational lysine acylation modifications. A noteworthy example is the newly identified lysine 2-hydroxyisobutyrylation (Khib) that is derived from 2-hydroxyisobutyrate and 2-hydroxyisobutyryl-CoA. Histone Khib has been shown to be associated with active gene expression in spermatogenic cells. However, the key elements that regulate this post-translational lysine acylation pathway remain unknown. This has hindered characterization of the mechanisms by which this modification exerts its biological functions. Here we show that Esa1p in budding yeast and its homologue Tip60 in human could add Khib to substrate proteins both in vitro and in vivo. In addition, we have identified HDAC2 and HDAC3 as the major enzymes to remove Khib. Moreover, we report the first global profiling of Khib proteome in mammalian cells, identifying 6 548 Khib sites on 1 725 substrate proteins. Our study has thus discovered both the "writers" and "erasers" for histone Khib marks, and major Khib protein substrates. These results not only illustrate the landscape of this new lysine acylation pathway, but also open new avenues for studying diverse functions of cellular metabolites associated with this pathway.
Collapse
Affiliation(s)
- He Huang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Zhouqing Luo
- Center for Synthetic Biology Engineering Research, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- MOE Key Laboratory of Bioinformatics and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shankang Qi
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Jing Huang
- MOE Key Laboratory of Bioinformatics and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Peng Xu
- MOE Key Laboratory of Bioinformatics and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiuxuan Wang
- Department of General Practice and Lab of PTM, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Li Gao
- Department of General Practice and Lab of PTM, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Fangyi Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Jian Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Wenhui Zhao
- Department of Biochemistry and Molecular Biology, Health Science Center and Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Peking University, Beijing 100191, China
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, 1130 Nicholas Avenue, New York, NY 10032, USA
| | - Zhucheng Chen
- MOE Key Laboratory of Bioinformatics and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lunzhi Dai
- Department of General Practice and Lab of PTM, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Junbiao Dai
- Center for Synthetic Biology Engineering Research, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- MOE Key Laboratory of Bioinformatics and Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yingming Zhao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
213
|
Meyer MJ, Beltrán JF, Liang S, Fragoza R, Rumack A, Liang J, Wei X, Yu H. Interactome INSIDER: a structural interactome browser for genomic studies. Nat Methods 2018; 15:107-114. [PMID: 29355848 PMCID: PMC6026581 DOI: 10.1038/nmeth.4540] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 10/22/2017] [Indexed: 02/07/2023]
Abstract
We present Interactome INSIDER, a tool to link genomic variant information with
structural protein-protein interactomes. Underlying this tool is the application of
machine learning to predict protein interaction interfaces for 185,957 protein
interactions with previously unresolved interfaces, in human and 7 model organisms,
including the entire experimentally determined human binary interactome. Predicted
interfaces exhibit similar functional properties as known interfaces, including enrichment
for disease mutations and recurrent cancer mutations. Through 2,164 de
novo mutagenesis experiments, we show that mutations of predicted and known
interface residues disrupt interactions at a similar rate, and much more frequently than
mutations outside of predicted interfaces. To spur functional genomic studies, Interactome
INSIDER (http://interactomeinsider.yulab.org) enables users to identify whether
variants or disease mutations are enriched in known and predicted interaction interfaces
at various resolutions. Users may explore known population variants, disease mutations,
and somatic cancer mutations, or upload their own set of mutations for this purpose.
Collapse
Affiliation(s)
- Michael J Meyer
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA.,Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York, USA
| | - Juan Felipe Beltrán
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Siqi Liang
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Robert Fragoza
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA.,Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| | - Aaron Rumack
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Jin Liang
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| | - Xiaomu Wei
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA.,Department of Medicine, Weill Cornell College of Medicine, New York, New York, USA
| | - Haiyuan Yu
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA.,Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
214
|
Pinto G, Radulovic M, Godovac-Zimmermann J. Spatial perspectives in the redox code-Mass spectrometric proteomics studies of moonlighting proteins. MASS SPECTROMETRY REVIEWS 2018; 37:81-100. [PMID: 27186965 DOI: 10.1002/mas.21508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/03/2016] [Indexed: 06/05/2023]
Abstract
The Redox Code involves specific, reversible oxidative changes in proteins that modulate protein tertiary structure, interactions, trafficking, and activity, and hence couple the proteome to the metabolic/oxidative state of cells. It is currently a major focus of study in cell biology. Recent studies of dynamic cellular spatial reorganization with MS-based subcellular-spatial-razor proteomics reveal that protein constituents of many subcellular structures, including mitochondria, the endoplasmic reticulum, the plasma membrane, and the extracellular matrix, undergo changes in their subcellular abundance/distribution in response to oxidative stress. These proteins are components of a diverse variety of functional processes spatially distributed across cells. Many of the same proteins are involved in response to suppression of DNA replication indicate that oxidative stress is strongly intertwined with DNA replication/proliferation. Both are replete with networks of moonlighting proteins that show coordinated changes in subcellular location and that include primary protein actuators of the redox code involved in the processing of NAD+ /NADH, NADP+ /NADPH, Cys/CySS, and GSH/GSSG redox couples. Small groups of key proteins such as {KPNA2, KPNB1, PCNA, PTMA, SET} constitute "spatial switches" that modulate many nuclear processes. Much of the functional response involves subcellular protein trafficking, including nuclear import/export processes, vesicle-mediated trafficking, the endoplasmic reticulum/Golgi pathway, chaperone-assisted processes, and other transport systems. This is not visible to measurements of total protein abundance by transcriptomics or proteomics. Comprehensive pictures of cellular function will require collection of data on the subcellular transport and local functions of many moonlighting proteins, especially of those with critical roles in spatial coordination across cells. The proteome-wide analysis of coordinated changes in abundance and trafficking of proteins offered by MS-based proteomics has a unique, crucial role to play in deciphering the complex adaptive systems that underlie cellular function. © 2016 Wiley Periodicals, Inc. Mass Spec Rev.
Collapse
Affiliation(s)
- Gabriella Pinto
- Division of Medicine, Center for Nephrology, Royal Free Campus, University College London, Rowland Hill Street, London, NW3 2PF, United Kingdom
| | - Marko Radulovic
- Insitute of Oncology and Radiology, Pasterova 14, Belgrade, 11000, Serbia
| | - Jasminka Godovac-Zimmermann
- Division of Medicine, Center for Nephrology, Royal Free Campus, University College London, Rowland Hill Street, London, NW3 2PF, United Kingdom
| |
Collapse
|
215
|
Benoit Bouvrette LP, Cody NAL, Bergalet J, Lefebvre FA, Diot C, Wang X, Blanchette M, Lécuyer E. CeFra-seq reveals broad asymmetric mRNA and noncoding RNA distribution profiles in Drosophila and human cells. RNA (NEW YORK, N.Y.) 2018; 24:98-113. [PMID: 29079635 PMCID: PMC5733575 DOI: 10.1261/rna.063172.117] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/13/2017] [Indexed: 05/26/2023]
Abstract
Cells are highly asymmetrical, a feature that relies on the sorting of molecular constituents, including proteins, lipids, and nucleic acids, to distinct subcellular locales. The localization of RNA molecules is an important layer of gene regulation required to modulate localized cellular activities, although its global prevalence remains unclear. We combine biochemical cell fractionation with RNA-sequencing (CeFra-seq) analysis to assess the prevalence and conservation of RNA asymmetric distribution on a transcriptome-wide scale in Drosophila and human cells. This approach reveals that the majority (∼80%) of cellular RNA species are asymmetrically distributed, whether considering coding or noncoding transcript populations, in patterns that are broadly conserved evolutionarily. Notably, a large number of Drosophila and human long noncoding RNAs and circular RNAs display enriched levels within specific cytoplasmic compartments, suggesting that these RNAs fulfill extra-nuclear functions. Moreover, fraction-specific mRNA populations exhibit distinctive sequence characteristics. Comparative analysis of mRNA fractionation profiles with that of their encoded proteins reveals a general lack of correlation in subcellular distribution, marked by strong cases of asymmetry. However, coincident distribution profiles are observed for mRNA/protein pairs related to a variety of functional protein modules, suggesting complex regulatory inputs of RNA localization to cellular organization.
Collapse
Affiliation(s)
- Louis Philip Benoit Bouvrette
- Institut de Recherches Clinique de Montréal (IRCM), Montréal H2W 1R7, Canada
- Département de Biochimie, Université de Montréal, Montréal H3C 3J7, Canada
| | - Neal A L Cody
- Institut de Recherches Clinique de Montréal (IRCM), Montréal H2W 1R7, Canada
| | - Julie Bergalet
- Institut de Recherches Clinique de Montréal (IRCM), Montréal H2W 1R7, Canada
| | - Fabio Alexis Lefebvre
- Institut de Recherches Clinique de Montréal (IRCM), Montréal H2W 1R7, Canada
- Département de Biochimie, Université de Montréal, Montréal H3C 3J7, Canada
| | - Cédric Diot
- Institut de Recherches Clinique de Montréal (IRCM), Montréal H2W 1R7, Canada
- Département de Biochimie, Université de Montréal, Montréal H3C 3J7, Canada
| | - Xiaofeng Wang
- Institut de Recherches Clinique de Montréal (IRCM), Montréal H2W 1R7, Canada
| | - Mathieu Blanchette
- McGill School of Computer Science, McGill University, Montréal H3A 0E9, Canada
| | - Eric Lécuyer
- Institut de Recherches Clinique de Montréal (IRCM), Montréal H2W 1R7, Canada
- Département de Biochimie, Université de Montréal, Montréal H3C 3J7, Canada
- Division of Experimental Medicine, McGill University, Montréal H4A 3J1, Canada
| |
Collapse
|
216
|
Song J, Carey M, Zhu H, Miao H, Ramírez JC, Wu H. Identifying the dynamic gene regulatory network during latent HIV-1 reactivation using high-dimensional ordinary differential equations. ACTA ACUST UNITED AC 2018; 11:135-153. [PMID: 34531927 DOI: 10.1504/ijcbdd.2018.10011910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Reactivation of latently infected cells has emerged as an important strategy for eradication of HIV. However, genetic mechanisms of regulation after reactivation remain unclear. We describe a five-step pipeline to study the dynamics of the gene regulatory network following a viral reactivation using high-dimensional ordinary differential equations. Our pipeline implements a combination of five different methods, by detecting temporally differentially expressed genes (step 1), clustering genes with similar temporal expression patterns into a small number of response modules (step2), performing a functional enrichment analysis within each gene response module (step 3), identifying a network structure based on the gene response modules using ordinary differential equations (ODE) and a high-dimensional variable selection technique (step 4), and obtaining a gene regulatory model based on refined parameter estimates using nonlinear least squares (step 5). We applied our pipeline to a time course gene expression data of latently infected T-cells following a latency-reversion.
Collapse
Affiliation(s)
- Jaejoon Song
- Department of Biostatistics, The University of Texas MD, Anderson Cancer Center, 1400 Pressler Street, Houston, TX, 77030, USA
| | - Michelle Carey
- Department of Mathematics and Statistics, McGill University, 805 Sherbrooke Street West, Montreal, Canada, H3A 0B9
| | - Hongjian Zhu
- Department of Biostatistics, The University of Texas School of Public Health, 1200 Pressler Street, Houston, TX, 77030, USA
| | - Hongyu Miao
- Department of Biostatistics, The University of Texas School of Public Health, 1200 Pressler Street, Houston, TX, 77030, USA
| | - Juan Camilo Ramírez
- Faculty of Computer Engineering, Universidad Antonio Nariño, Cl. 58a Bis 3794, Bogotá, Cundinamarca, Colombia
| | - Hulin Wu
- Department of Biostatistics, The University of Texas School of Public Health, 1200 Pressler Street, Houston, TX, USA, 77030, USA
| |
Collapse
|
217
|
Goebels F, Hu L, Bader G, Emili A. Automated Computational Inference of Multi-protein Assemblies from Biochemical Co-purification Data. Methods Mol Biol 2018; 1764:391-399. [PMID: 29605929 DOI: 10.1007/978-1-4939-7759-8_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Biology has amassed a wealth of information about the function of a multitude of protein-coding genes across species. The challenge now is to understand how all these proteins work together to form a living organism, and a crucial step for gaining this knowledge is a complete description of the molecular "wiring circuits" that underlie cellular processes. In this chapter, we describe a general computational framework for predicting multi-protein assemblies from biochemical co-fractionation data.
Collapse
Affiliation(s)
- Florian Goebels
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Lucas Hu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Gary Bader
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Andrew Emili
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
218
|
Mlecnik B, Galon J, Bindea G. Comprehensive functional analysis of large lists of genes and proteins. J Proteomics 2018; 171:2-10. [DOI: 10.1016/j.jprot.2017.03.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/13/2017] [Accepted: 03/19/2017] [Indexed: 01/16/2023]
|
219
|
Tian Z, Guo M, Wang C, Liu X, Wang S. Refine gene functional similarity network based on interaction networks. BMC Bioinformatics 2017; 18:550. [PMID: 29297381 PMCID: PMC5751769 DOI: 10.1186/s12859-017-1969-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In recent years, biological interaction networks have become the basis of some essential study and achieved success in many applications. Some typical networks such as protein-protein interaction networks have already been investigated systematically. However, little work has been available for the construction of gene functional similarity networks so far. In this research, we will try to build a high reliable gene functional similarity network to promote its further application. RESULTS Here, we propose a novel method to construct and refine the gene functional similarity network. It mainly contains three steps. First, we establish an integrated gene functional similarity networks based on different functional similarity calculation methods. Then, we construct a referenced gene-gene association network based on the protein-protein interaction networks. At last, we refine the spurious edges in the integrated gene functional similarity network with the help of the referenced gene-gene association network. Experiment results indicate that the refined gene functional similarity network (RGFSN) exhibits a scale-free, small world and modular architecture, with its degrees fit best to power law distribution. In addition, we conduct protein complex prediction experiment for human based on RGFSN and achieve an outstanding result, which implies it has high reliability and wide application significance. CONCLUSIONS Our efforts are insightful for constructing and refining gene functional similarity networks, which can be applied to build other high quality biological networks.
Collapse
Affiliation(s)
- Zhen Tian
- Department of computer Science and Engineering, Harbin Institute of Technology, Harbin, 150001 People’s Republic of China
| | - Maozu Guo
- Department of computer Science and Engineering, Harbin Institute of Technology, Harbin, 150001 People’s Republic of China
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing, 100044 People’s Republic of China
| | - Chunyu Wang
- Department of computer Science and Engineering, Harbin Institute of Technology, Harbin, 150001 People’s Republic of China
| | - Xiaoyan Liu
- Department of computer Science and Engineering, Harbin Institute of Technology, Harbin, 150001 People’s Republic of China
| | - Shiming Wang
- Department of computer Science and Engineering, Harbin Institute of Technology, Harbin, 150001 People’s Republic of China
| |
Collapse
|
220
|
Jeong H, Qian X, Yoon BJ. CUFID-query: accurate network querying through random walk based network flow estimation. BMC Bioinformatics 2017; 18:500. [PMID: 29297279 PMCID: PMC5751815 DOI: 10.1186/s12859-017-1899-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Functional modules in biological networks consist of numerous biomolecules and their complicated interactions. Recent studies have shown that biomolecules in a functional module tend to have similar interaction patterns and that such modules are often conserved across biological networks of different species. As a result, such conserved functional modules can be identified through comparative analysis of biological networks. RESULTS In this work, we propose a novel network querying algorithm based on the CUFID (Comparative network analysis Using the steady-state network Flow to IDentify orthologous proteins) framework combined with an efficient seed-and-extension approach. The proposed algorithm, CUFID-query, can accurately detect conserved functional modules as small subnetworks in the target network that are expected to perform similar functions to the given query functional module. The CUFID framework was recently developed for probabilistic pairwise global comparison of biological networks, and it has been applied to pairwise global network alignment, where the framework was shown to yield accurate network alignment results. In the proposed CUFID-query algorithm, we adopt the CUFID framework and extend it for local network alignment, specifically to solve network querying problems. First, in the seed selection phase, the proposed method utilizes the CUFID framework to compare the query and the target networks and to predict the probabilistic node-to-node correspondence between the networks. Next, the algorithm selects and greedily extends the seed in the target network by iteratively adding nodes that have frequent interactions with other nodes in the seed network, in a way that the conductance of the extended network is maximally reduced. Finally, CUFID-query removes irrelevant nodes from the querying results based on the personalized PageRank vector for the induced network that includes the fully extended network and its neighboring nodes. CONCLUSIONS Through extensive performance evaluation based on biological networks with known functional modules, we show that CUFID-query outperforms the existing state-of-the-art algorithms in terms of prediction accuracy and biological significance of the predictions.
Collapse
Affiliation(s)
- Hyundoo Jeong
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, 77843, TX, USA.,Department of Neuroogy, Baylor College of Medicine, Houston, TX, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Xiaoning Qian
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, 77843, TX, USA.,TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, TX, USA
| | - Byung-Jun Yoon
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, 77843, TX, USA. .,TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, TX, USA.
| |
Collapse
|
221
|
A High-Resolution Genome-Wide CRISPR/Cas9 Viability Screen Reveals Structural Features and Contextual Diversity of the Human Cell-Essential Proteome. Mol Cell Biol 2017; 38:MCB.00302-17. [PMID: 29038160 DOI: 10.1128/mcb.00302-17] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/11/2017] [Indexed: 11/20/2022] Open
Abstract
To interrogate genes essential for cell growth, proliferation and survival in human cells, we carried out a genome-wide clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 screen in a B-cell lymphoma line using a custom extended-knockout (EKO) library of 278,754 single-guide RNAs (sgRNAs) that targeted 19,084 RefSeq genes, 20,852 alternatively spliced exons, and 3,872 hypothetical genes. A new statistical analysis tool called robust analytics and normalization for knockout screens (RANKS) identified 2,280 essential genes, 234 of which were unique. Individual essential genes were validated experimentally and linked to ribosome biogenesis and stress responses. Essential genes exhibited a bimodal distribution across 10 different cell lines, consistent with a continuous variation in essentiality as a function of cell type. Genes essential in more lines had more severe fitness defects and encoded the evolutionarily conserved structural cores of protein complexes, whereas genes essential in fewer lines formed context-specific modules and encoded subunits at the periphery of essential complexes. The essentiality of individual protein residues across the proteome correlated with evolutionary conservation, structural burial, modular domains, and protein interaction interfaces. Many alternatively spliced exons in essential genes were dispensable and were enriched for disordered regions. Fitness defects were observed for 44 newly evolved hypothetical reading frames. These results illuminate the contextual nature and evolution of essential gene functions in human cells.
Collapse
|
222
|
Maruyama O, Kuwahara Y. RocSampler: regularizing overlapping protein complexes in protein-protein interaction networks. BMC Bioinformatics 2017; 18:491. [PMID: 29244010 PMCID: PMC5731504 DOI: 10.1186/s12859-017-1920-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Background In recent years, protein-protein interaction (PPI) networks have been well recognized as important resources to elucidate various biological processes and cellular mechanisms. In this paper, we address the problem of predicting protein complexes from a PPI network. This problem has two difficulties. One is related to small complexes, which contains two or three components. It is relatively difficult to identify them due to their simpler internal structure, but unfortunately complexes of such sizes are dominant in major protein complex databases, such as CYC2008. Another difficulty is how to model overlaps between predicted complexes, that is, how to evaluate different predicted complexes sharing common proteins because CYC2008 and other databases include such protein complexes. Thus, it is critical how to model overlaps between predicted complexes to identify them simultaneously. Results In this paper, we propose a sampling-based protein complex prediction method, RocSampler (Regularizing Overlapping Complexes), which exploits, as part of the whole scoring function, a regularization term for the overlaps of predicted complexes and that for the distribution of sizes of predicted complexes. We have implemented RocSampler in MATLAB and its executable file for Windows is available at the site, http://imi.kyushu-u.ac.jp/~om/software/RocSampler/. Conclusions We have applied RocSampler to five yeast PPI networks and shown that it is superior to other existing methods. This implies that the design of scoring functions including regularization terms is an effective approach for protein complex prediction.
Collapse
Affiliation(s)
- Osamu Maruyama
- Institute of Mathematics for Industry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| | - Yuki Kuwahara
- Graduate School of Mathematics, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| |
Collapse
|
223
|
Vilne B, Skogsberg J, Foroughi Asl H, Talukdar HA, Kessler T, Björkegren JLM, Schunkert H. Network analysis reveals a causal role of mitochondrial gene activity in atherosclerotic lesion formation. Atherosclerosis 2017; 267:39-48. [PMID: 29100060 DOI: 10.1016/j.atherosclerosis.2017.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/05/2017] [Accepted: 10/18/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS Mitochondrial damage and augmented production of reactive oxygen species (ROS) may represent an intermediate step by which hypercholesterolemia exacerbates atherosclerotic lesion formation. METHODS To test this hypothesis, in mice with severe but genetically reversible hypercholesterolemia (i.e. the so called Reversa mouse model), we performed time-resolved analyses of mitochondrial transcriptome in the aortic arch employing a systems-level network approach. RESULTS During hypercholesterolemia, we observed a massive down-regulation (>28%) of mitochondrial genes, specifically at the time of rapid atherosclerotic lesion expansion and foam cell formation, i.e. between 30 and 40 weeks of age. Both phenomena - down-regulation of mitochondrial genes and lesion expansion - were largely reversible by genetically lowering plasma cholesterol (by >80%, from 427 to 54 ± 31 mg/L) at 30 weeks. Co-expression network analysis revealed that both mitochondrial signature genes were highly connected in two modules, negatively correlating with lesion size and supported as causal for coronary artery disease (CAD) in humans, as expression-associated single nucleotide polymorphisms (eSNPs) representing their genes overlapped markedly with established disease risk loci. Within these modules, we identified the transcription factor estrogen related receptor (ERR)-α and its co-factors PGC1-α and -β, i.e. two members of the peroxisome proliferator-activated receptor γ co-activator 1 family of transcription regulators, as key regulatory genes. Together, these factors are known as major orchestrators of mitochondrial biogenesis and antioxidant responses. CONCLUSIONS Using a network approach, we demonstrate how hypercholesterolemia could hamper mitochondrial activity during atherosclerosis progression and pinpoint potential therapeutic targets to counteract these processes.
Collapse
Affiliation(s)
- Baiba Vilne
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; DZHK (German Research Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Josefin Skogsberg
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Hassan Foroughi Asl
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Husain Ahammad Talukdar
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Integrated Cardio Metabolic Center (ICMC), Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; DZHK (German Research Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Johan L M Björkegren
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Department of Physiology, Institute of Biomedicine and Translation Medicine, University of Tartu, Estonia; Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai New York, New York, USA; Clinical Gene Networks AB, Stockholm, Sweden.
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; DZHK (German Research Centre for Cardiovascular Research), Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
224
|
Top-down characterization of endogenous protein complexes with native proteomics. Nat Chem Biol 2017; 14:36-41. [PMID: 29131144 PMCID: PMC5726920 DOI: 10.1038/nchembio.2515] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/04/2017] [Indexed: 11/08/2022]
Abstract
Protein complexes exhibit great diversity in protein membership, post-translational modifications and noncovalent cofactors, enabling them to function as the actuators of many important biological processes. The exposition of these molecular features using current methods lacks either throughput or molecular specificity, ultimately limiting the use of protein complexes as direct analytical targets in a wide range of applications. Here, we apply native proteomics, enabled by a multistage tandem MS approach, to characterize 125 intact endogenous complexes and 217 distinct proteoforms derived from mouse heart and human cancer cell lines in discovery mode. The native conditions preserved soluble protein-protein interactions, high-stoichiometry noncovalent cofactors, covalent modifications to cysteines, and, remarkably, superoxide ligands bound to the metal cofactor of superoxide dismutase 2. These data enable precise compositional analysis of protein complexes as they exist in the cell and demonstrate a new approach that uses MS as a bridge to structural biology.
Collapse
|
225
|
Tommiska J, Känsäkoski J, Skibsbye L, Vaaralahti K, Liu X, Lodge EJ, Tang C, Yuan L, Fagerholm R, Kanters JK, Lahermo P, Kaunisto M, Keski-Filppula R, Vuoristo S, Pulli K, Ebeling T, Valanne L, Sankila EM, Kivirikko S, Lääperi M, Casoni F, Giacobini P, Phan-Hug F, Buki T, Tena-Sempere M, Pitteloud N, Veijola R, Lipsanen-Nyman M, Kaunisto K, Mollard P, Andoniadou CL, Hirsch JA, Varjosalo M, Jespersen T, Raivio T. Two missense mutations in KCNQ1 cause pituitary hormone deficiency and maternally inherited gingival fibromatosis. Nat Commun 2017; 8:1289. [PMID: 29097701 PMCID: PMC5668380 DOI: 10.1038/s41467-017-01429-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 09/14/2017] [Indexed: 01/05/2023] Open
Abstract
Familial growth hormone deficiency provides an opportunity to identify new genetic causes of short stature. Here we combine linkage analysis with whole-genome resequencing in patients with growth hormone deficiency and maternally inherited gingival fibromatosis. We report that patients from three unrelated families harbor either of two missense mutations, c.347G>T p.(Arg116Leu) or c.1106C>T p.(Pro369Leu), in KCNQ1, a gene previously implicated in the long QT interval syndrome. Kcnq1 is expressed in hypothalamic GHRH neurons and pituitary somatotropes. Co-expressing KCNQ1 with the KCNE2 β-subunit shows that both KCNQ1 mutants increase current levels in patch clamp analyses and are associated with reduced pituitary hormone secretion from AtT-20 cells. In conclusion, our results reveal a role for the KCNQ1 potassium channel in the regulation of human growth, and show that growth hormone deficiency associated with maternally inherited gingival fibromatosis is an allelic disorder with cardiac arrhythmia syndromes caused by KCNQ1 mutations.
Collapse
Affiliation(s)
- Johanna Tommiska
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland.,Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital (HUCH), 00029, Helsinki, Finland
| | - Johanna Känsäkoski
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Lasse Skibsbye
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Kirsi Vaaralahti
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Xiaonan Liu
- Institute of Biotechnology, Biocenter 3, University of Helsinki, 00014, Helsinki, Finland
| | - Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, King's College London, Floor 27 Tower Wing, Guy's Campus, London, SE1 9RT, UK
| | - Chuyi Tang
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Lei Yuan
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Rainer Fagerholm
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland.,Department of Obstetrics and Gynecology, HUCH, 00029, Helsinki, Finland
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, 22000, Copenhagen, Denmark.,Department of Cardiology, Herlev & Gentofte University Hospitals, University of Copenhagen, 22000, Copenhagen, Denmark
| | - Päivi Lahermo
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | - Mari Kaunisto
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science HiLIFE, University of Helsinki, 00014, Helsinki, Finland
| | | | - Sanna Vuoristo
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Kristiina Pulli
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Tapani Ebeling
- Department of Medicine, Oulu University Hospital, Finland and Research Unit of Internal Medicine, University of Oulu, 90014, Oulu, Finland
| | - Leena Valanne
- Helsinki Medical Imaging Center, HUCH, 00029, Helsinki, Finland
| | | | - Sirpa Kivirikko
- Department of Clinical Genetics, HUCH, 00029, Helsinki, Finland
| | - Mitja Lääperi
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland
| | - Filippo Casoni
- Inserm U1172, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, 59045, Lille, France.,University of Lille, School of Medicine, 59045, Lille, France
| | - Paolo Giacobini
- Inserm U1172, Jean-Pierre Aubert Research Center, Development and Plasticity of the Neuroendocrine Brain, 59045, Lille, France.,University of Lille, School of Medicine, 59045, Lille, France
| | - Franziska Phan-Hug
- Pediatrics, Division of Pediatric Endocrinology, Diabetology and Obesity, University Hospital Lausanne (CHUV), 1011, Lausanne, Switzerland
| | - Tal Buki
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Institute of Structural Biology, 69978, Ramat Aviv, Israel
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, 14071, Cordoba, Spain.,Instituto Maimonides de Investigacion Biomedica (IMIBIC/HURS), 14004, Cordoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Nelly Pitteloud
- Pediatrics, Division of Pediatric Endocrinology, Diabetology and Obesity, University Hospital Lausanne (CHUV), 1011, Lausanne, Switzerland
| | - Riitta Veijola
- Department of Children and Adolescents, Oulu University Hospital, 90029, Oulu, Finland.,Department of Pediatrics, PEDEGO Research Center, Medical Research Center, University of Oulu, 90014, Oulu, Finland
| | - Marita Lipsanen-Nyman
- Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital (HUCH), 00029, Helsinki, Finland
| | - Kari Kaunisto
- Department of Children and Adolescents, Oulu University Hospital, 90029, Oulu, Finland
| | - Patrice Mollard
- IGF, CNRS, INSERM, Univ. Montpellier, F-34094, Montpellier, France
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, King's College London, Floor 27 Tower Wing, Guy's Campus, London, SE1 9RT, UK.,Department of Internal Medicine III, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Joel A Hirsch
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Institute of Structural Biology, 69978, Ramat Aviv, Israel
| | - Markku Varjosalo
- Institute of Biotechnology, Biocenter 3, University of Helsinki, 00014, Helsinki, Finland
| | - Thomas Jespersen
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Taneli Raivio
- Faculty of Medicine, Department of Physiology, University of Helsinki, 00014, Helsinki, Finland. .,Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital (HUCH), 00029, Helsinki, Finland.
| |
Collapse
|
226
|
Nagamani S, Gaur AS, Tanneeru K, Muneeswaran G, Madugula SS, Consortium M, Druzhilovskiy D, Poroikov VV, Sastry GN. Molecular property diagnostic suite (MPDS): Development of disease-specific open source web portals for drug discovery. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2017; 28:913-926. [PMID: 29206500 DOI: 10.1080/1062936x.2017.1402819] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 06/07/2023]
Abstract
Molecular property diagnostic suite (MPDS) is a Galaxy-based open source drug discovery and development platform. MPDS web portals are designed for several diseases, such as tuberculosis, diabetes mellitus, and other metabolic disorders, specifically aimed to evaluate and estimate the drug-likeness of a given molecule. MPDS consists of three modules, namely data libraries, data processing, and data analysis tools which are configured and interconnected to assist drug discovery for specific diseases. The data library module encompasses vast information on chemical space, wherein the MPDS compound library comprises 110.31 million unique molecules generated from public domain databases. Every molecule is assigned with a unique ID and card, which provides complete information for the molecule. Some of the modules in the MPDS are specific to the diseases, while others are non-specific. Importantly, a suitably altered protocol can be effectively generated for another disease-specific MPDS web portal by modifying some of the modules. Thus, the MPDS suite of web portals shows great promise to emerge as disease-specific portals of great value, integrating chemoinformatics, bioinformatics, molecular modelling, and structure- and analogue-based drug discovery approaches.
Collapse
Affiliation(s)
- S Nagamani
- a Centre for Molecular Modeling , CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| | - A S Gaur
- a Centre for Molecular Modeling , CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| | - K Tanneeru
- a Centre for Molecular Modeling , CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| | - G Muneeswaran
- a Centre for Molecular Modeling , CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| | - S S Madugula
- a Centre for Molecular Modeling , CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| | | | | | - V V Poroikov
- b Institute of Biomedical Chemistry , Moscow , Russia
| | - G N Sastry
- a Centre for Molecular Modeling , CSIR-Indian Institute of Chemical Technology , Hyderabad , India
| |
Collapse
|
227
|
Systematic proteome and proteostasis profiling in human Trisomy 21 fibroblast cells. Nat Commun 2017; 8:1212. [PMID: 29089484 PMCID: PMC5663699 DOI: 10.1038/s41467-017-01422-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/15/2017] [Indexed: 12/17/2022] Open
Abstract
Down syndrome (DS) is mostly caused by a trisomy of the entire Chromosome 21 (Trisomy 21, T21). Here, we use SWATH mass spectrometry to quantify protein abundance and protein turnover in fibroblasts from a monozygotic twin pair discordant for T21, and to profile protein expression in 11 unrelated DS individuals and matched controls. The integration of the steady-state and turnover proteomic data indicates that protein-specific degradation of members of stoichiometric complexes is a major determinant of T21 gene dosage outcome, both within and between individuals. This effect is not apparent from genomic and transcriptomic data. The data also reveal that T21 results in extensive proteome remodeling, affecting proteins encoded by all chromosomes. Finally, we find broad, organelle-specific post-transcriptional effects such as significant downregulation of the mitochondrial proteome contributing to T21 hallmarks. Overall, we provide a valuable proteomic resource to understand the origin of DS phenotypic manifestations. Trisomy 21 (T21) is a major cause of Down syndrome but little is known about its impact on the cellular proteome. Here, the authors define the proteome of T21 fibroblasts and its turnover and also map proteomic differences in monozygotic T21-discordant twins, revealing extensive, organelle-specific changes caused by T21.
Collapse
|
228
|
Ryan CJ, Kennedy S, Bajrami I, Matallanas D, Lord CJ. A Compendium of Co-regulated Protein Complexes in Breast Cancer Reveals Collateral Loss Events. Cell Syst 2017; 5:399-409.e5. [PMID: 29032073 PMCID: PMC5660599 DOI: 10.1016/j.cels.2017.09.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/31/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022]
Abstract
Protein complexes are responsible for the bulk of activities within the cell, but how their behavior and abundance varies across tumors remains poorly understood. By combining proteomic profiles of breast tumors with a large-scale protein-protein interaction network, we have identified a set of 285 high-confidence protein complexes whose subunits have highly correlated protein abundance across tumor samples. We used this set to identify complexes that are reproducibly under- or overexpressed in specific breast cancer subtypes. We found that mutation or deletion of one subunit of a co-regulated complex was often associated with a collateral reduction in protein expression of additional complex members. This collateral loss phenomenon was typically evident from proteomic, but not transcriptomic, profiles, suggesting post-transcriptional control. Mutation of the tumor suppressor E-cadherin (CDH1) was associated with a collateral loss of members of the adherens junction complex, an effect we validated using an engineered model of E-cadherin loss.
Collapse
Affiliation(s)
- Colm J Ryan
- School of Computer Science, University College Dublin, Dublin 4, Ireland; Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland.
| | - Susan Kennedy
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Ilirjana Bajrami
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Christopher J Lord
- The Breast Cancer Now Toby Robins Breast Cancer Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
229
|
Elurbe DM, Paranjpe SS, Georgiou G, van Kruijsbergen I, Bogdanovic O, Gibeaux R, Heald R, Lister R, Huynen MA, van Heeringen SJ, Veenstra GJC. Regulatory remodeling in the allo-tetraploid frog Xenopus laevis. Genome Biol 2017; 18:198. [PMID: 29065907 PMCID: PMC5655803 DOI: 10.1186/s13059-017-1335-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 10/03/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Genome duplication has played a pivotal role in the evolution of many eukaryotic lineages, including the vertebrates. A relatively recent vertebrate genome duplication is that in Xenopus laevis, which resulted from the hybridization of two closely related species about 17 million years ago. However, little is known about the consequences of this duplication at the level of the genome, the epigenome, and gene expression. RESULTS The X. laevis genome consists of two subgenomes, referred to as L (long chromosomes) and S (short chromosomes), that originated from distinct diploid progenitors. Of the parental subgenomes, S chromosomes have degraded faster than L chromosomes from the point of genome duplication until the present day. Deletions appear to have the largest effect on pseudogene formation and loss of regulatory regions. Deleted regions are enriched for long DNA repeats and the flanking regions have high alignment scores, suggesting that non-allelic homologous recombination has played a significant role in the loss of DNA. To assess innovations in the X. laevis subgenomes we examined p300-bound enhancer peaks that are unique to one subgenome and absent from X. tropicalis. A large majority of new enhancers comprise transposable elements. Finally, to dissect early and late events following interspecific hybridization, we examined the epigenome and the enhancer landscape in X. tropicalis × X. laevis hybrid embryos. Strikingly, young X. tropicalis DNA transposons are derepressed and recruit p300 in hybrid embryos. CONCLUSIONS The results show that erosion of X. laevis genes and functional regulatory elements is associated with repeats and non-allelic homologous recombination and furthermore that young repeats have also contributed to the p300-bound regulatory landscape following hybridization and whole-genome duplication.
Collapse
Affiliation(s)
- Dei M Elurbe
- Radboud University Medical Center, Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands
| | - Sarita S Paranjpe
- Radboud University, Faculty of Science, Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands
| | - Georgios Georgiou
- Radboud University, Faculty of Science, Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands
| | - Ila van Kruijsbergen
- Radboud University, Faculty of Science, Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands
| | - Ozren Bogdanovic
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
- ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Perth, Australia
| | - Romain Gibeaux
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Rebecca Heald
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Ryan Lister
- Harry Perkins Institute of Medical Research and ARC Centre of Excellence in Plant Energy Biology, The University of Western Australia, Perth, WA, 6009, Australia
| | - Martijn A Huynen
- Radboud University Medical Center, Center for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands.
| | - Simon J van Heeringen
- Radboud University, Faculty of Science, Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands.
| | - Gert Jan C Veenstra
- Radboud University, Faculty of Science, Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
230
|
Stacey RG, Skinnider MA, Scott NE, Foster LJ. A rapid and accurate approach for prediction of interactomes from co-elution data (PrInCE). BMC Bioinformatics 2017; 18:457. [PMID: 29061110 PMCID: PMC5654062 DOI: 10.1186/s12859-017-1865-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/09/2017] [Indexed: 12/24/2022] Open
Abstract
Background An organism’s protein interactome, or complete network of protein-protein interactions, defines the protein complexes that drive cellular processes. Techniques for studying protein complexes have traditionally applied targeted strategies such as yeast two-hybrid or affinity purification-mass spectrometry to assess protein interactions. However, given the vast number of protein complexes, more scalable methods are necessary to accelerate interaction discovery and to construct whole interactomes. We recently developed a complementary technique based on the use of protein correlation profiling (PCP) and stable isotope labeling in amino acids in cell culture (SILAC) to assess chromatographic co-elution as evidence of interacting proteins. Importantly, PCP-SILAC is also capable of measuring protein interactions simultaneously under multiple biological conditions, allowing the detection of treatment-specific changes to an interactome. Given the uniqueness and high dimensionality of co-elution data, new tools are needed to compare protein elution profiles, control false discovery rates, and construct an accurate interactome. Results Here we describe a freely available bioinformatics pipeline, PrInCE, for the analysis of co-elution data. PrInCE is a modular, open-source library that is computationally inexpensive, able to use label and label-free data, and capable of detecting tens of thousands of protein-protein interactions. Using a machine learning approach, PrInCE offers greatly reduced run time, more predicted interactions at the same stringency, prediction of protein complexes, and greater ease of use over previous bioinformatics tools for co-elution data. PrInCE is implemented in Matlab (version R2017a). Source code and standalone executable programs for Windows and Mac OSX are available at https://github.com/fosterlab/PrInCE, where usage instructions can be found. An example dataset and output are also provided for testing purposes. Conclusions PrInCE is the first fast and easy-to-use data analysis pipeline that predicts interactomes and protein complexes from co-elution data. PrInCE allows researchers without bioinformatics expertise to analyze high-throughput co-elution datasets. Electronic supplementary material The online version of this article (10.1186/s12859-017-1865-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- R Greg Stacey
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada.
| | - Michael A Skinnider
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada
| | - Nichollas E Scott
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada.,Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, V6T 1Z4, Canada. .,Department of Biochemistry, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
231
|
Begum T, Ghosh TC, Basak S. Systematic Analyses and Prediction of Human Drug Side Effect Associated Proteins from the Perspective of Protein Evolution. Genome Biol Evol 2017; 9:337-350. [PMID: 28391292 PMCID: PMC5499873 DOI: 10.1093/gbe/evw301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2017] [Indexed: 12/20/2022] Open
Abstract
Identification of various factors involved in adverse drug reactions in target proteins to develop therapeutic drugs with minimal/no side effect is very important. In this context, we have performed a comparative evolutionary rate analyses between the genes exhibiting drug side-effect(s) (SET) and genes showing no side effect (NSET) with an aim to increase the prediction accuracy of SET/NSET proteins using evolutionary rate determinants. We found that SET proteins are more conserved than the NSET proteins. The rates of evolution between SET and NSET protein primarily depend upon their noncomplex (protein complex association number = 0) forming nature, phylogenetic age, multifunctionality, membrane localization, and transmembrane helix content irrespective of their essentiality, total druggability (total number of drugs/target), m-RNA expression level, and tissue expression breadth. We also introduced two novel terms—killer druggability (number of drugs with killing side effect(s)/target), essential druggability (number of drugs targeting essential proteins/target) to explain the evolutionary rate variation between SET and NSET proteins. Interestingly, we noticed that SET proteins are younger than NSET proteins and multifunctional younger SET proteins are candidates of acquiring killing side effects. We provide evidence that higher killer druggability, multifunctionality, and transmembrane helices support the conservation of SET proteins over NSET proteins in spite of their recent origin. By employing all these entities, our Support Vector Machine model predicts human SET/NSET proteins to a high degree of accuracy (∼86%).
Collapse
Affiliation(s)
- Tina Begum
- Bioinformatics Centre, Tripura University, Suryamaninagar, Tripura, India
| | | | - Surajit Basak
- Bioinformatics Centre, Tripura University, Suryamaninagar, Tripura, India.,Department of Molecular Biology & Bioinformatics, Tripura University, Suryamaninagar, Tripura, India
| |
Collapse
|
232
|
Cheung CHY, Hsu CL, Chen KP, Chong ST, Wu CH, Huang HC, Juan HF. MCM2-regulated functional networks in lung cancer by multi-dimensional proteomic approach. Sci Rep 2017; 7:13302. [PMID: 29038488 PMCID: PMC5643318 DOI: 10.1038/s41598-017-13440-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/25/2017] [Indexed: 12/17/2022] Open
Abstract
DNA replication control is vital for maintaining genome stability and the cell cycle, perhaps most notably during cell division. Malignancies often exhibit defective minichromosome maintenance protein 2 (MCM2), a cancer proliferation biomarker that serves as a licensing factor in the initiation of DNA replication. MCM2 is also known to be one of the ATPase active sites that facilitates conformational changes and drives DNA unwinding at the origin of DNA replication. However, the biological networks of MCM2 in lung cancer cells via protein phosphorylation remain unmapped. The RNA-seq datasets from The Cancer Genome Atlas (TCGA) revealed that MCM2 overexpression is correlated with poor survival rate in lung cancer patients. To uncover MCM2-regulated functional networks in lung cancer, we performed multi-dimensional proteomic approach by integrating analysis of the phosphoproteome and proteome, and identified a total of 2361 phosphorylation sites on 753 phosphoproteins, and 4672 proteins. We found that the deregulation of MCM2 is involved in lung cancer cell proliferation, the cell cycle, and migration. Furthermore, HMGA1S99 phosphorylation was found to be differentially expressed under MCM2 perturbation in opposite directions, and plays an important role in regulating lung cancer cell proliferation. This study therefore enhances our capacity to therapeutically target cancer-specific phosphoproteins.
Collapse
Affiliation(s)
- Chantal Hoi Yin Cheung
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 10617, Taiwan
| | - Chia-Lang Hsu
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Kai-Pu Chen
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 10617, Taiwan
| | - Siao-Ting Chong
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 10617, Taiwan
| | - Chang-Hsun Wu
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, 11221, Taiwan.
| | - Hsueh-Fen Juan
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 10617, Taiwan. .,Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan. .,Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
233
|
Gonçalves E, Fragoulis A, Garcia-Alonso L, Cramer T, Saez-Rodriguez J, Beltrao P. Widespread Post-transcriptional Attenuation of Genomic Copy-Number Variation in Cancer. Cell Syst 2017; 5:386-398.e4. [PMID: 29032074 PMCID: PMC5660600 DOI: 10.1016/j.cels.2017.08.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/21/2017] [Accepted: 08/23/2017] [Indexed: 12/28/2022]
Abstract
Copy-number variations (CNVs) are ubiquitous in cancer and often act as driver events, but the effects of CNVs on the proteome of tumors are poorly understood. Here, we analyze recently published genomics, transcriptomics, and proteomics datasets made available by CPTAC and TCGA consortia on 282 breast, ovarian, and colorectal tumor samples to investigate the impact of CNVs in the proteomes of these cells. We found that CNVs are buffered by post-transcriptional regulation in 23%-33% of proteins that are significantly enriched in protein complex members. Our analyses show that complex subunits are highly co-regulated, and some act as rate-limiting steps of complex assembly, as their depletion induces decreased abundance of other complex members. We identified 48 such rate-limiting interactions and experimentally confirmed our predictions on the interactions of AP3B1 with AP3M1 and GTF2E2 with GTF2E1. This study highlights the importance of post-transcriptional mechanisms in cancer that allow cells to cope with their altered genomes.
Collapse
Affiliation(s)
- Emanuel Gonçalves
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, UK
| | - Athanassios Fragoulis
- Molecular Tumor Biology, Department of General, Visceral and Transplantation Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Luz Garcia-Alonso
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, UK
| | - Thorsten Cramer
- Molecular Tumor Biology, Department of General, Visceral and Transplantation Surgery, RWTH University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands; ESCAM - European Surgery Center Aachen Maastricht, Germany and the Netherlands
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, UK; RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, 52057 Aachen, Germany.
| | - Pedro Beltrao
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, UK.
| |
Collapse
|
234
|
Learning causal networks with latent variables from multivariate information in genomic data. PLoS Comput Biol 2017; 13:e1005662. [PMID: 28968390 PMCID: PMC5685645 DOI: 10.1371/journal.pcbi.1005662] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 11/14/2017] [Accepted: 06/29/2017] [Indexed: 12/24/2022] Open
Abstract
Learning causal networks from large-scale genomic data remains challenging in absence of time series or controlled perturbation experiments. We report an information- theoretic method which learns a large class of causal or non-causal graphical models from purely observational data, while including the effects of unobserved latent variables, commonly found in many genomic datasets. Starting from a complete graph, the method iteratively removes dispensable edges, by uncovering significant information contributions from indirect paths, and assesses edge-specific confidences from randomization of available data. The remaining edges are then oriented based on the signature of causality in observational data. The approach and associated algorithm, miic, outperform earlier methods on a broad range of benchmark networks. Causal network reconstructions are presented at different biological size and time scales, from gene regulation in single cells to whole genome duplication in tumor development as well as long term evolution of vertebrates. Miic is publicly available at https://github.com/miicTeam/MIIC. The reconstruction of causal networks from genomic data is an important but challenging problem. Predicting key regulatory interactions or genomic alterations at the origin of human diseases can guide experimental investigation and ultimately inspire innovative therapy. However, causal relationships are difficult to establish without the possibility to directly perturb the organisms’ genome for ethical or practical reasons. Besides, unmeasured (latent) variables may be hidden in many genomic datasets and lead to spurious causal relationships between observed variables. We propose in this paper an efficient computational approach, miic, that overcomes these limitations and learns causal networks from non-perturbative (observational) data in the presence of latent variables. In addition, we assess the confidence of each predicted interaction and demonstrate the enhanced robustness and accuracy of miic compared to alternative existing methods. This approach can be applied on a wide range of datasets and provide new biological insights on regulatory networks from single cell expression data or genomic alterations during tumor development. Miic is implemented in an R package freely available to the scientific community under a General Public License.
Collapse
|
235
|
Mosher DF, Wilkerson EM, Turton KB, Hebert AS, Coon JJ. Proteomics of Eosinophil Activation. Front Med (Lausanne) 2017; 4:159. [PMID: 29034237 PMCID: PMC5626809 DOI: 10.3389/fmed.2017.00159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/13/2017] [Indexed: 12/30/2022] Open
Abstract
We recently identified and quantified >7,000 proteins in non-activated human peripheral blood eosinophils using liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) and described phosphoproteomic changes that accompany acute activation of eosinophils by interleukin-5 (IL5) (1). These data comprise a treasure trove of information about eosinophils. We illustrate the power of label-free LC-MS/MS quantification by considering four examples: complexity of eosinophil STATs, contribution of immunoproteasome subunits to eosinophil proteasomes, complement of integrin subunits, and contribution of platelet proteins originating from platelet-eosinophil complexes to the overall proteome. We describe how isobaric labeling enables robust sample-to-sample comparisons and relate the 220 phosphosites that changed significantly upon treatment with IL5 to previous studies of eosinophil activation. Finally, we review previous attempts to leverage the power of mass spectrometry to discern differences between eosinophils of healthy subjects and those with eosinophil-associated conditions and point out features of label-free quantification and isobaric labeling that are important in planning future mass spectrometric studies.
Collapse
Affiliation(s)
- Deane F Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, United States.,Department of Medicine, University of Wisconsin, Madison, WI, United States
| | - Emily M Wilkerson
- Department of Chemistry, University of Wisconsin, Madison, WI, United States
| | - Keren B Turton
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, United States
| | - Alexander S Hebert
- Department of Chemistry, University of Wisconsin, Madison, WI, United States
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, United States.,Department of Chemistry, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
236
|
Song R, Wang ZD, Schapira M. Disease Association and Druggability of WD40 Repeat Proteins. J Proteome Res 2017; 16:3766-3773. [DOI: 10.1021/acs.jproteome.7b00451] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Richard Song
- Structural
Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Zhong-Duo Wang
- Structural
Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Matthieu Schapira
- Structural
Genomics Consortium, University of Toronto, Toronto, Ontario M5G 1L7, Canada
- Department
of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
237
|
Besnard E, Hakre S, Kampmann M, Lim HW, Hosmane NN, Martin A, Bassik MC, Verschueren E, Battivelli E, Chan J, Svensson JP, Gramatica A, Conrad RJ, Ott M, Greene WC, Krogan NJ, Siliciano RF, Weissman JS, Verdin E. The mTOR Complex Controls HIV Latency. Cell Host Microbe 2017; 20:785-797. [PMID: 27978436 DOI: 10.1016/j.chom.2016.11.001] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 09/30/2016] [Accepted: 11/06/2016] [Indexed: 12/22/2022]
Abstract
A population of CD4 T lymphocytes harboring latent HIV genomes can persist in patients on antiretroviral therapy, posing a barrier to HIV eradication. To examine cellular complexes controlling HIV latency, we conducted a genome-wide screen with a pooled ultracomplex shRNA library and in vitro system modeling HIV latency and identified the mTOR complex as a modulator of HIV latency. Knockdown of mTOR complex subunits or pharmacological inhibition of mTOR activity suppresses reversal of latency in various HIV-1 latency models and HIV-infected patient cells. mTOR inhibitors suppress HIV transcription both through the viral transactivator Tat and via Tat-independent mechanisms. This inhibition occurs at least in part via blocking the phosphorylation of CDK9, a p-TEFb complex member that serves as a cofactor for Tat-mediated transcription. The control of HIV latency by mTOR signaling identifies a pathway that may have significant therapeutic opportunities.
Collapse
Affiliation(s)
- Emilie Besnard
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shweta Hakre
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Martin Kampmann
- Department of Cellular and Molecular Pharmacology, The California Institute for Quantitative Biomedical Research, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hyung W Lim
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nina N Hosmane
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Alyssa Martin
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Michael C Bassik
- Department of Cellular and Molecular Pharmacology, The California Institute for Quantitative Biomedical Research, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Erik Verschueren
- Department of Cellular and Molecular Pharmacology, The California Institute for Quantitative Biomedical Research, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Emilie Battivelli
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jonathan Chan
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - J Peter Svensson
- Karolinska Institutet, Department of Biosciences and Nutrition, Novum, 141 83 Huddinge, Sweden
| | - Andrea Gramatica
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ryan J Conrad
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Melanie Ott
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Warner C Greene
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nevan J Krogan
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, The California Institute for Quantitative Biomedical Research, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, The California Institute for Quantitative Biomedical Research, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Eric Verdin
- Gladstone Institute of Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
238
|
Hernandez C, Mella C, Navarro G, Olivera-Nappa A, Araya J. Protein complex prediction via dense subgraphs and false positive analysis. PLoS One 2017; 12:e0183460. [PMID: 28937982 PMCID: PMC5609739 DOI: 10.1371/journal.pone.0183460] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 08/04/2017] [Indexed: 01/04/2023] Open
Abstract
Many proteins work together with others in groups called complexes in order to achieve a specific function. Discovering protein complexes is important for understanding biological processes and predict protein functions in living organisms. Large-scale and throughput techniques have made possible to compile protein-protein interaction networks (PPI networks), which have been used in several computational approaches for detecting protein complexes. Those predictions might guide future biologic experimental research. Some approaches are topology-based, where highly connected proteins are predicted to be complexes; some propose different clustering algorithms using partitioning, overlaps among clusters for networks modeled with unweighted or weighted graphs; and others use density of clusters and information based on protein functionality. However, some schemes still require much processing time or the quality of their results can be improved. Furthermore, most of the results obtained with computational tools are not accompanied by an analysis of false positives. We propose an effective and efficient mining algorithm for discovering highly connected subgraphs, which is our base for defining protein complexes. Our representation is based on transforming the PPI network into a directed acyclic graph that reduces the number of represented edges and the search space for discovering subgraphs. Our approach considers weighted and unweighted PPI networks. We compare our best alternative using PPI networks from Saccharomyces cerevisiae (yeast) and Homo sapiens (human) with state-of-the-art approaches in terms of clustering, biological metrics and execution times, as well as three gold standards for yeast and two for human. Furthermore, we analyze false positive predicted complexes searching the PDBe (Protein Data Bank in Europe) database in order to identify matching protein complexes that have been purified and structurally characterized. Our analysis shows that more than 50 yeast protein complexes and more than 300 human protein complexes found to be false positives according to our prediction method, i.e., not described in the gold standard complex databases, in fact contain protein complexes that have been characterized structurally and documented in PDBe. We also found that some of these protein complexes have recently been classified as part of a Periodic Table of Protein Complexes. The latest version of our software is publicly available at http://doi.org/10.6084/m9.figshare.5297314.v1.
Collapse
Affiliation(s)
- Cecilia Hernandez
- Computer Science, University of Concepción, Concepción, Chile
- Center for Biotechnology and Bioengineering (CeBiB), Department of Computer Science, University of Chile, Santiago, Chile
- * E-mail:
| | - Carlos Mella
- Computer Science, University of Concepción, Concepción, Chile
| | - Gonzalo Navarro
- Center for Biotechnology and Bioengineering (CeBiB), Department of Computer Science, University of Chile, Santiago, Chile
| | - Alvaro Olivera-Nappa
- Center for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering and Biotechnology, University of Chile, Santiago, Chile
| | - Jaime Araya
- Computer Science, University of Concepción, Concepción, Chile
| |
Collapse
|
239
|
Havugimana PC, Hu P, Emili A. Protein complexes, big data, machine learning and integrative proteomics: lessons learned over a decade of systematic analysis of protein interaction networks. Expert Rev Proteomics 2017; 14:845-855. [PMID: 28918672 DOI: 10.1080/14789450.2017.1374179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OVERVIEW Elucidation of the networks of physical (functional) interactions present in cells and tissues is fundamental for understanding the molecular organization of biological systems, the mechanistic basis of essential and disease-related processes, and for functional annotation of previously uncharacterized proteins (via guilt-by-association or -correlation). After a decade in the field, we felt it timely to document our own experiences in the systematic analysis of protein interaction networks. Areas covered: Researchers worldwide have contributed innovative experimental and computational approaches that have driven the rapidly evolving field of 'functional proteomics'. These include mass spectrometry-based methods to characterize macromolecular complexes on a global-scale and sophisticated data analysis tools - most notably machine learning - that allow for the generation of high-quality protein association maps. Expert commentary: Here, we recount some key lessons learned, with an emphasis on successful workflows, and challenges, arising from our own and other groups' ongoing efforts to generate, interpret and report proteome-scale interaction networks in increasingly diverse biological contexts.
Collapse
Affiliation(s)
- Pierre C Havugimana
- a Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , ON , Canada.,b Department of Molecular Genetics , University of Toronto , Toronto , ON , Canada
| | - Pingzhao Hu
- c Department of Biochemistry and Medical Genetics , University of Manitoba , Winnipeg , MB , Canada
| | - Andrew Emili
- a Donnelly Centre for Cellular and Biomolecular Research , University of Toronto , Toronto , ON , Canada.,b Department of Molecular Genetics , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
240
|
Lam HC, Liu HJ, Baglini CV, Filippakis H, Alesi N, Nijmeh J, Du H, Lope AL, Cottrill KA, Handen A, Asara JM, Kwiatkowski DJ, Ben-Sahra I, Oldham WM, Chan SY, Henske EP. Rapamycin-induced miR-21 promotes mitochondrial homeostasis and adaptation in mTORC1 activated cells. Oncotarget 2017; 8:64714-64727. [PMID: 29029388 PMCID: PMC5630288 DOI: 10.18632/oncotarget.19947] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 06/25/2017] [Indexed: 12/24/2022] Open
Abstract
mTORC1 hyperactivation drives the multi-organ hamartomatous disease tuberous sclerosis complex (TSC). Rapamycin inhibits mTORC1, inducing partial tumor responses; however, the tumors regrow following treatment cessation. We discovered that the oncogenic miRNA, miR-21, is increased in Tsc2-deficient cells and, surprisingly, further increased by rapamycin. To determine the impact of miR-21 in TSC, we inhibited miR-21 in vitro. miR-21 inhibition significantly repressed the tumorigenic potential of Tsc2-deficient cells and increased apoptosis sensitivity. Tsc2-deficient cells' clonogenic and anchorage independent growth were reduced by ∼50% (p<0.01) and ∼75% (p<0.0001), respectively, and combined rapamycin treatment decreased soft agar growth by ∼90% (p<0.0001). miR-21 inhibition also increased sensitivity to apoptosis. Through a network biology-driven integration of RNAseq data, we discovered that miR-21 promotes mitochondrial adaptation and homeostasis in Tsc2-deficient cells. miR-21 inhibition reduced mitochondrial polarization and function in Tsc2-deficient cells, with and without co-treatment with rapamycin. Importantly, miR-21 inhibition limited Tsc2-deficient tumor growth in vivo, reducing tumor size by approximately 3-fold (p<0.0001). When combined with rapamcyin, miR-21 inhibition showed even more striking efficacy, both during treatment and after treatment cessation, with a 4-fold increase in median survival following rapamycin cessation (p=0.0008). We conclude that miR-21 promotes mTORC1-driven tumorigenesis via a mechanism that involves the mitochondria, and that miR-21 is a potential therapeutic target for TSC-associated hamartomas and other mTORC1-driven tumors, with the potential for synergistic efficacy when combined with rapalogs.
Collapse
Affiliation(s)
- Hilaire C. Lam
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Heng-Jia Liu
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Christian V. Baglini
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Harilaos Filippakis
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Nicola Alesi
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Nijmeh
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Heng Du
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Alicia Llorente Lope
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Katherine A. Cottrill
- Department of Medicine, Division of Cardiology, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Adam Handen
- Department of Medicine, Division of Cardiology, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - John M. Asara
- Department of Medicine, Division of Signal Transduction, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David J. Kwiatkowski
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - William M. Oldham
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephen Y. Chan
- Department of Medicine, Division of Cardiology, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Elizabeth P. Henske
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
241
|
Lapek JD, Greninger P, Morris R, Amzallag A, Pruteanu-Malinici I, Benes CH, Haas W. Detection of dysregulated protein-association networks by high-throughput proteomics predicts cancer vulnerabilities. Nat Biotechnol 2017; 35:983-989. [PMID: 28892078 PMCID: PMC5683351 DOI: 10.1038/nbt.3955] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/08/2017] [Indexed: 12/17/2022]
Abstract
The formation of protein complexes and the co-regulation of the cellular concentrations of proteins are essential mechanisms for cellular signaling and for maintaining homeostasis. Here we use isobaric-labeling multiplexed proteomics to analyze protein co-regulation and show that this allows the identification of protein-protein associations with high accuracy. We apply this 'interactome mapping by high-throughput quantitative proteome analysis' (IMAHP) method to a panel of 41 breast cancer cell lines and show that deviations of the observed protein co-regulations in specific cell lines from the consensus network affects cellular fitness. Furthermore, these aberrant interactions serve as biomarkers that predict the drug sensitivity of cell lines in screens across 195 drugs. We expect that IMAHP can be broadly used to gain insight into how changing landscapes of protein-protein associations affect the phenotype of biological systems.
Collapse
Affiliation(s)
- John D Lapek
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Patricia Greninger
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Robert Morris
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Arnaud Amzallag
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Iulian Pruteanu-Malinici
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Cyril H Benes
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, USA
| |
Collapse
|
242
|
Chatzileontiadou DSM, Samiotaki M, Alexopoulou AN, Cotsiki M, Panayotou G, Stamatiadi M, Balatsos NAA, Leonidas DD, Kontou M. Proteomic Analysis of Human Angiogenin Interactions Reveals Cytoplasmic PCNA as a Putative Binding Partner. J Proteome Res 2017; 16:3606-3622. [PMID: 28777577 DOI: 10.1021/acs.jproteome.7b00335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human Angiogenin (hAng) is a member of the ribonuclease A superfamily and a potent inducer of neovascularization. Protein interactions of hAng in the nucleus and cytoplasm of the human umbilical vein cell line EA.hy926 have been investigated by mass spectroscopy. Data are available via ProteomeXchange with identifiers PXD006583 and PXD006584. The first gel-free analysis of hAng immunoprecipitates revealed many statistically significant potential hAng-interacting proteins involved in crucial biological pathways. Surprisingly, proliferating cell nuclear antigen (PCNA), was found to be immunoprecipitated with hAng only in the cytoplasm. The hAng-PCNA interaction and colocalization in the specific cellular compartment was validated with immunoprecipitation, immunoblotting, and immunocytochemistry. The results revealed that PCNA is predominantly localized in the cytoplasm, while hAng is distributed both in the nucleus and in the cytoplasm. hAng and PCNA colocalize in the cytoplasm, suggesting that they may interact in this compartment.
Collapse
Affiliation(s)
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | | | - Marina Cotsiki
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | - George Panayotou
- Biomedical Sciences Research Center "Alexander Fleming" , Vari 16672, Greece
| | - Melina Stamatiadi
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Nikolaos A A Balatsos
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Demetres D Leonidas
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| | - Maria Kontou
- Department of Biochemistry and Biotechnology, University of Thessaly , Biopolis, 41500 Larissa, Greece
| |
Collapse
|
243
|
Han H, Braunschweig U, Gonatopoulos-Pournatzis T, Weatheritt RJ, Hirsch CL, Ha KCH, Radovani E, Nabeel-Shah S, Sterne-Weiler T, Wang J, O'Hanlon D, Pan Q, Ray D, Zheng H, Vizeacoumar F, Datti A, Magomedova L, Cummins CL, Hughes TR, Greenblatt JF, Wrana JL, Moffat J, Blencowe BJ. Multilayered Control of Alternative Splicing Regulatory Networks by Transcription Factors. Mol Cell 2017; 65:539-553.e7. [PMID: 28157508 DOI: 10.1016/j.molcel.2017.01.011] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/16/2016] [Accepted: 01/05/2017] [Indexed: 12/21/2022]
Abstract
Networks of coordinated alternative splicing (AS) events play critical roles in development and disease. However, a comprehensive knowledge of the factors that regulate these networks is lacking. We describe a high-throughput system for systematically linking trans-acting factors to endogenous RNA regulatory events. Using this system, we identify hundreds of factors associated with diverse regulatory layers that positively or negatively control AS events linked to cell fate. Remarkably, more than one-third of the regulators are transcription factors. Further analyses of the zinc finger protein Zfp871 and BTB/POZ domain transcription factor Nacc1, which regulate neural and stem cell AS programs, respectively, reveal roles in controlling the expression of specific splicing regulators. Surprisingly, these proteins also appear to regulate target AS programs via binding RNA. Our results thus uncover a large "missing cache" of splicing regulators among annotated transcription factors, some of which dually regulate AS through direct and indirect mechanisms.
Collapse
Affiliation(s)
- Hong Han
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | | | - Robert J Weatheritt
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Calley L Hirsch
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Kevin C H Ha
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ernest Radovani
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Syed Nabeel-Shah
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Juli Wang
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Dave O'Hanlon
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Qun Pan
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Debashish Ray
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Hong Zheng
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Frederick Vizeacoumar
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Alessandro Datti
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Lilia Magomedova
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Timothy R Hughes
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jack F Greenblatt
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jeffrey L Wrana
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jason Moffat
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Benjamin J Blencowe
- Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
244
|
Niu Y, Liu C, Moghimyfiroozabad S, Yang Y, Alavian KN. PrePhyloPro: phylogenetic profile-based prediction of whole proteome linkages. PeerJ 2017; 5:e3712. [PMID: 28875072 PMCID: PMC5578374 DOI: 10.7717/peerj.3712] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/28/2017] [Indexed: 02/05/2023] Open
Abstract
Direct and indirect functional links between proteins as well as their interactions as part of larger protein complexes or common signaling pathways may be predicted by analyzing the correlation of their evolutionary patterns. Based on phylogenetic profiling, here we present a highly scalable and time-efficient computational framework for predicting linkages within the whole human proteome. We have validated this method through analysis of 3,697 human pathways and molecular complexes and a comparison of our results with the prediction outcomes of previously published co-occurrency model-based and normalization methods. Here we also introduce PrePhyloPro, a web-based software that uses our method for accurately predicting proteome-wide linkages. We present data on interactions of human mitochondrial proteins, verifying the performance of this software. PrePhyloPro is freely available at http://prephylopro.org/phyloprofile/.
Collapse
Affiliation(s)
- Yulong Niu
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom.,Key Lab of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China.,School of Medicine, Department of Internal Medicine, Endocrinology, Yale University, New Haven, CT, United States of America
| | - Chengcheng Liu
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | | - Yi Yang
- Key Lab of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Kambiz N Alavian
- Department of Medicine, Division of Brain Sciences, Imperial College London, London, United Kingdom.,School of Medicine, Department of Internal Medicine, Endocrinology, Yale University, New Haven, CT, United States of America.,Department of Biology, The Bahá'í Institute for Higher Education (BIHE), Tehran, Iran
| |
Collapse
|
245
|
Babbi G, Martelli PL, Profiti G, Bovo S, Savojardo C, Casadio R. eDGAR: a database of Disease-Gene Associations with annotated Relationships among genes. BMC Genomics 2017; 18:554. [PMID: 28812536 PMCID: PMC5558190 DOI: 10.1186/s12864-017-3911-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Genetic investigations, boosted by modern sequencing techniques, allow dissecting the genetic component of different phenotypic traits. These efforts result in the compilation of lists of genes related to diseases and show that an increasing number of diseases is associated with multiple genes. Investigating functional relations among genes associated with the same disease contributes to highlighting molecular mechanisms of the pathogenesis. RESULTS We present eDGAR, a database collecting and organizing the data on gene/disease associations as derived from OMIM, Humsavar and ClinVar. For each disease-associated gene, eDGAR collects information on its annotation. Specifically, for lists of genes, eDGAR provides information on: i) interactions retrieved from PDB, BIOGRID and STRING; ii) co-occurrence in stable and functional structural complexes; iii) shared Gene Ontology annotations; iv) shared KEGG and REACTOME pathways; v) enriched functional annotations computed with NET-GE; vi) regulatory interactions derived from TRRUST; vii) localization on chromosomes and/or co-localisation in neighboring loci. The present release of eDGAR includes 2672 diseases, related to 3658 different genes, for a total number of 5729 gene-disease associations. 71% of the genes are linked to 621 multigenic diseases and eDGAR highlights their common GO terms, KEGG/REACTOME pathways, physical and regulatory interactions. eDGAR includes a network based enrichment method for detecting statistically significant functional terms associated to groups of genes. CONCLUSIONS eDGAR offers a resource to analyze disease-gene associations. In multigenic diseases genes can share physical interactions and/or co-occurrence in the same functional processes. eDGAR is freely available at: edgar.biocomp.unibo.it.
Collapse
Affiliation(s)
- Giulia Babbi
- Biocomputing Group, BiGeA, University of Bologna, Bologna, Italy
| | | | - Giuseppe Profiti
- Biocomputing Group, BiGeA, University of Bologna, Bologna, Italy
| | - Samuele Bovo
- Biocomputing Group, BiGeA, University of Bologna, Bologna, Italy
| | | | - Rita Casadio
- Biocomputing Group, BiGeA, University of Bologna, Bologna, Italy.,Interdepartmental Center «Giorgio Prodi» for Cancer Research, University of Bologna, Bologna, Italy
| |
Collapse
|
246
|
Tsherniak A, Vazquez F, Montgomery PG, Weir BA, Kryukov G, Cowley GS, Gill S, Harrington WF, Pantel S, Krill-Burger JM, Meyers RM, Ali L, Goodale A, Lee Y, Jiang G, Hsiao J, Gerath WFJ, Howell S, Merkel E, Ghandi M, Garraway LA, Root DE, Golub TR, Boehm JS, Hahn WC. Defining a Cancer Dependency Map. Cell 2017; 170:564-576.e16. [PMID: 28753430 DOI: 10.1016/j.cell.2017.06.010] [Citation(s) in RCA: 1581] [Impact Index Per Article: 225.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/09/2017] [Accepted: 06/07/2017] [Indexed: 12/15/2022]
Abstract
Most human epithelial tumors harbor numerous alterations, making it difficult to predict which genes are required for tumor survival. To systematically identify cancer dependencies, we analyzed 501 genome-scale loss-of-function screens performed in diverse human cancer cell lines. We developed DEMETER, an analytical framework that segregates on- from off-target effects of RNAi. 769 genes were differentially required in subsets of these cell lines at a threshold of six SDs from the mean. We found predictive models for 426 dependencies (55%) by nonlinear regression modeling considering 66,646 molecular features. Many dependencies fall into a limited number of classes, and unexpectedly, in 82% of models, the top biomarkers were expression based. We demonstrated the basis behind one such predictive model linking hypermethylation of the UBB ubiquitin gene to a dependency on UBC. Together, these observations provide a foundation for a cancer dependency map that facilitates the prioritization of therapeutic targets.
Collapse
Affiliation(s)
- Aviad Tsherniak
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Francisca Vazquez
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Phil G Montgomery
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Barbara A Weir
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Gregory Kryukov
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Glenn S Cowley
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Stanley Gill
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | | | - Sasha Pantel
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | | | - Robin M Meyers
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Levi Ali
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Amy Goodale
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Yenarae Lee
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Guozhi Jiang
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Jessica Hsiao
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | | | - Sara Howell
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Erin Merkel
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Mahmoud Ghandi
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Levi A Garraway
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA, USA; Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD, USA
| | - David E Root
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Todd R Golub
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA, USA; Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD, USA
| | - Jesse S Boehm
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| |
Collapse
|
247
|
Genome-wide predicting disease-related protein complexes by walking on the heterogeneous network based on data integration and laplacian normalization. Comput Biol Chem 2017; 69:41-47. [DOI: 10.1016/j.compbiolchem.2017.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/08/2017] [Accepted: 04/12/2017] [Indexed: 11/20/2022]
|
248
|
Safari-Alighiarloo N, Taghizadeh M, Tabatabaei SM, Shahsavari S, Namaki S, Khodakarim S, Rezaei-Tavirani M. Identification of new key genes for type 1 diabetes through construction and analysis of protein-protein interaction networks based on blood and pancreatic islet transcriptomes. J Diabetes 2017; 9:764-777. [PMID: 27625010 DOI: 10.1111/1753-0407.12483] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 08/17/2016] [Accepted: 09/08/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic β-cells are destroyed by infiltrating immune cells. Bilateral cooperation of pancreatic β-cells and immune cells has been proposed in the progression of T1D, but as yet no systems study has investigated this possibility. The aims of the study were to elucidate the underlying molecular mechanisms and identify key genes associated with T1D risk using a network biology approach. METHODS Interactome (protein-protein interaction [PPI]) and transcriptome data were integrated to construct networks of differentially expressed genes in peripheral blood mononuclear cells (PBMCs) and pancreatic β-cells. Centrality, modularity, and clique analyses of networks were used to get more meaningful biological information. RESULTS Analysis of genes expression profiles revealed several cytokines and chemokines in β-cells and their receptors in PBMCs, which is supports the dialogue between these two tissues in terms of PPIs. Functional modules and complexes analysis unraveled most significant biological pathways such as immune response, apoptosis, spliceosome, proteasome, and pathways of protein synthesis in the tissues. Finally, Y-box binding protein 1 (YBX1), SRSF protein kinase 1 (SRPK1), proteasome subunit alpha1/ 3, (PSMA1/3), X-ray repair cross complementing 6 (XRCC6), Cbl proto-oncogene (CBL), SRC proto-oncogene, non-receptor tyrosine kinase (SRC), phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), phospholipase C gamma 1 (PLCG1), SHC adaptor protein1 (SHC1) and ubiquitin conjugating enzyme E2 N (UBE2N) were identified as key markers that were hub-bottleneck genes involved in functional modules and complexes. CONCLUSIONS This study provide new insights into network biomarkers that may be considered potential therapeutic targets.
Collapse
Affiliation(s)
- Nahid Safari-Alighiarloo
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghizadeh
- Bioinformatics Department, Institute of Biochemistry and Biophysics, Tehran University, Tehran, Iran
| | - Seyyed Mohammad Tabatabaei
- Medical Informatics Department, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soodeh Shahsavari
- Biostatistics Department, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Namaki
- Department of Immunology, Faculty of Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soheila Khodakarim
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
249
|
Climente-González H, Porta-Pardo E, Godzik A, Eyras E. The Functional Impact of Alternative Splicing in Cancer. Cell Rep 2017; 20:2215-2226. [DOI: 10.1016/j.celrep.2017.08.012] [Citation(s) in RCA: 364] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/15/2017] [Accepted: 07/26/2017] [Indexed: 12/29/2022] Open
|
250
|
Mazza A, Klockmeier K, Wanker E, Sharan R. An integer programming framework for inferring disease complexes from network data. Bioinformatics 2017; 32:i271-i277. [PMID: 27307626 PMCID: PMC4908347 DOI: 10.1093/bioinformatics/btw263] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MOTIVATION Unraveling the molecular mechanisms that underlie disease calls for methods that go beyond the identification of single causal genes to inferring larger protein assemblies that take part in the disease process. RESULTS Here, we develop an exact, integer-programming-based method for associating protein complexes with disease. Our approach scores proteins based on their proximity in a protein-protein interaction network to a prior set that is known to be relevant for the studied disease. These scores are combined with interaction information to infer densely interacting protein complexes that are potentially disease-associated. We show that our method outperforms previous ones and leads to predictions that are well supported by current experimental data and literature knowledge. AVAILABILITY AND IMPLEMENTATION The datasets we used, the executables and the results are available at www.cs.tau.ac.il/roded/disease_complexes.zip CONTACT roded@post.tau.ac.il.
Collapse
Affiliation(s)
- Arnon Mazza
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | | | - Erich Wanker
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Roded Sharan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|