1
|
Li Y, Herold T, Mansmann U, Hornung R. Does combining numerous data types in multi-omics data improve or hinder performance in survival prediction? Insights from a large-scale benchmark study. BMC Med Inform Decis Mak 2024; 24:244. [PMID: 39223659 PMCID: PMC11370316 DOI: 10.1186/s12911-024-02642-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Predictive modeling based on multi-omics data, which incorporates several types of omics data for the same patients, has shown potential to outperform single-omics predictive modeling. Most research in this domain focuses on incorporating numerous data types, despite the complexity and cost of acquiring them. The prevailing assumption is that increasing the number of data types necessarily improves predictive performance. However, the integration of less informative or redundant data types could potentially hinder this performance. Therefore, identifying the most effective combinations of omics data types that enhance predictive performance is critical for cost-effective and accurate predictions. METHODS In this study, we systematically evaluated the predictive performance of all 31 possible combinations including at least one of five genomic data types (mRNA, miRNA, methylation, DNAseq, and copy number variation) using 14 cancer datasets with right-censored survival outcomes, publicly available from the TCGA database. We employed various prediction methods and up-weighted clinical data in every model to leverage their predictive importance. Harrell's C-index and the integrated Brier Score were used as performance measures. To assess the robustness of our findings, we performed a bootstrap analysis at the level of the included datasets. Statistical testing was conducted for key results, limiting the number of tests to ensure a low risk of false positives. RESULTS Contrary to expectations, we found that using only mRNA data or a combination of mRNA and miRNA data was sufficient for most cancer types. For some cancer types, the additional inclusion of methylation data led to improved prediction results. Far from enhancing performance, the introduction of more data types most often resulted in a decline in performance, which varied between the two performance measures. CONCLUSIONS Our findings challenge the prevailing notion that combining multiple omics data types in multi-omics survival prediction improves predictive performance. Thus, the widespread approach in multi-omics prediction of incorporating as many data types as possible should be reconsidered to avoid suboptimal prediction results and unnecessary expenditure.
Collapse
Affiliation(s)
- Yingxia Li
- Institute for Medical Information Processing, Biometry and Epidemiology, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Tobias Herold
- Laboratory for Leukemia Diagnostics, Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Ulrich Mansmann
- Institute for Medical Information Processing, Biometry and Epidemiology, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Roman Hornung
- Institute for Medical Information Processing, Biometry and Epidemiology, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
- Munich Center for Machine Learning (MCML), Munich, Germany
| |
Collapse
|
2
|
Yu KKH, Basu S, Baquer G, Ahn R, Gantchev J, Jindal S, Regan MS, Abou-Mrad Z, Prabhu MC, Williams MJ, D'Souza AD, Malinowski SW, Hopland K, Elhanati Y, Stopka SA, Stortchevoi A, He Z, Sun J, Chen Y, Espejo AB, Chow KH, Yerrum S, Kao PL, Kerrigan BP, Norberg L, Nielsen D, Puduvalli VK, Huse J, Beroukhim R, Kim YSB, Goswami S, Boire A, Frisken S, Cima MJ, Holdhoff M, Lucas CHG, Bettegowda C, Levine SS, Bale TA, Brennan C, Reardon DA, Lang FF, Antonio Chiocca E, Ligon KL, White FM, Sharma P, Tabar V, Agar NYR. Investigative needle core biopsies for multi-omics in Glioblastoma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.29.23300541. [PMID: 38234840 PMCID: PMC10793534 DOI: 10.1101/2023.12.29.23300541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Glioblastoma (GBM) is a primary brain cancer with an abysmal prognosis and few effective therapies. The ability to investigate the tumor microenvironment before and during treatment would greatly enhance both understanding of disease response and progression, as well as the delivery and impact of therapeutics. Stereotactic biopsies are a routine surgical procedure performed primarily for diagnostic histopathologic purposes. The role of investigative biopsies - tissue sampling for the purpose of understanding tumor microenvironmental responses to treatment using integrated multi-modal molecular analyses ('Multi-omics") has yet to be defined. Secondly, it is unknown whether comparatively small tissue samples from brain biopsies can yield sufficient information with such methods. Here we adapt stereotactic needle core biopsy tissue in two separate patients. In the first patient with recurrent GBM we performed highly resolved multi-omics analysis methods including single cell RNA sequencing, spatial-transcriptomics, metabolomics, proteomics, phosphoproteomics, T-cell clonotype analysis, and MHC Class I immunopeptidomics from biopsy tissue that was obtained from a single procedure. In a second patient we analyzed multi-regional core biopsies to decipher spatial and genomic variance. We also investigated the utility of stereotactic biopsies as a method for generating patient derived xenograft models in a separate patient cohort. Dataset integration across modalities showed good correspondence between spatial modalities, highlighted immune cell associated metabolic pathways and revealed poor correlation between RNA expression and the tumor MHC Class I immunopeptidome. In conclusion, stereotactic needle biopsy cores are of sufficient quality to generate multi-omics data, provide data rich insight into a patient's disease process and tumor immune microenvironment and can be of value in evaluating treatment responses. One sentence summary Integrative multi-omics analysis of stereotactic needle core biopsies in glioblastoma.
Collapse
|
3
|
Winkler S, Winkler I, Figaschewski M, Tiede T, Nordheim A, Kohlbacher O. De novo identification of maximally deregulated subnetworks based on multi-omics data with DeRegNet. BMC Bioinformatics 2022; 23:139. [PMID: 35439941 PMCID: PMC9020058 DOI: 10.1186/s12859-022-04670-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Background With a growing amount of (multi-)omics data being available, the extraction of knowledge from these datasets is still a difficult problem. Classical enrichment-style analyses require predefined pathways or gene sets that are tested for significant deregulation to assess whether the pathway is functionally involved in the biological process under study. De novo identification of these pathways can reduce the bias inherent in predefined pathways or gene sets. At the same time, the definition and efficient identification of these pathways de novo from large biological networks is a challenging problem. Results We present a novel algorithm, DeRegNet, for the identification of maximally deregulated subnetworks on directed graphs based on deregulation scores derived from (multi-)omics data. DeRegNet can be interpreted as maximum likelihood estimation given a certain probabilistic model for de-novo subgraph identification. We use fractional integer programming to solve the resulting combinatorial optimization problem. We can show that the approach outperforms related algorithms on simulated data with known ground truths. On a publicly available liver cancer dataset we can show that DeRegNet can identify biologically meaningful subgraphs suitable for patient stratification. DeRegNet can also be used to find explicitly multi-omics subgraphs which we demonstrate by presenting subgraphs with consistent methylation-transcription patterns. DeRegNet is freely available as open-source software. Conclusion The proposed algorithmic framework and its available implementation can serve as a valuable heuristic hypothesis generation tool contextualizing omics data within biomolecular networks.
Collapse
Affiliation(s)
- Sebastian Winkler
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany. .,International Max Planck Research School (IMPRS) "From Molecules to Organism", Tübingen, Germany.
| | - Ivana Winkler
- International Max Planck Research School (IMPRS) "From Molecules to Organism", Tübingen, Germany.,Interfaculty Institute for Cell Biology (IFIZ), University of Tuebingen, Tübingen, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Figaschewski
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany
| | - Thorsten Tiede
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany
| | - Alfred Nordheim
- Interfaculty Institute for Cell Biology (IFIZ), University of Tuebingen, Tübingen, Germany.,Leibniz Institute on Aging (FLI), Jena, Germany
| | - Oliver Kohlbacher
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany.,Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tübingen, Germany.,Translational Bioinformatics, University Hospital Tuebingen, Tübingen, Germany
| |
Collapse
|
4
|
Heo YJ, Hwa C, Lee GH, Park JM, An JY. Integrative Multi-Omics Approaches in Cancer Research: From Biological Networks to Clinical Subtypes. Mol Cells 2021; 44:433-443. [PMID: 34238766 PMCID: PMC8334347 DOI: 10.14348/molcells.2021.0042] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/09/2021] [Accepted: 05/12/2021] [Indexed: 11/27/2022] Open
Abstract
Multi-omics approaches are novel frameworks that integrate multiple omics datasets generated from the same patients to better understand the molecular and clinical features of cancers. A wide range of emerging omics and multi-view clustering algorithms now provide unprecedented opportunities to further classify cancers into subtypes, improve the survival prediction and therapeutic outcome of these subtypes, and understand key pathophysiological processes through different molecular layers. In this review, we overview the concept and rationale of multi-omics approaches in cancer research. We also introduce recent advances in the development of multi-omics algorithms and integration methods for multiple-layered datasets from cancer patients. Finally, we summarize the latest findings from large-scale multi-omics studies of various cancers and their implications for patient subtyping and drug development.
Collapse
Affiliation(s)
- Yong Jin Heo
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Korea
- Department of Integrated Biomedical and Life Science, Korea University, Seoul 02841, Korea
| | - Chanwoong Hwa
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Korea
| | - Gang-Hee Lee
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Korea
| | - Jae-Min Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Korea
| | - Joon-Yong An
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02841, Korea
- Department of Integrated Biomedical and Life Science, Korea University, Seoul 02841, Korea
| |
Collapse
|
5
|
Mantini G, Pham TV, Piersma SR, Jimenez CR. Computational Analysis of Phosphoproteomics Data in Multi-Omics Cancer Studies. Proteomics 2020; 21:e1900312. [PMID: 32875713 DOI: 10.1002/pmic.201900312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/09/2020] [Indexed: 12/24/2022]
Abstract
Multiple types of molecular data for the same set of clinical samples are increasingly available and may be analyzed jointly in an integrative analysis to maximize comprehensive biological insight. This analysis is important as separate analyses of individual omics data types usually do not fully explain disease phenotypes. An increasing number of studies have now been focusing on multi-omics data integration, yet not many studies have included phosphoproteomics data, an important layer for understanding signaling pathways. Multi-omics integration methods with phosphoproteomics data are reviewed in the context of cancer research as well as multi-omics methods papers that would be promising to apply to phosphoproteomics data. Analysis of individual data types is still the major approach even in large cohort proteogenomics studies. Hence, a section is dedicated on possible integrative methods for multi-omics and phosphoproteomics data. In summary, this review provides the readers with both currently used integrative methods previously applied to phosphoproteomics and multi-omics data integration and other algorithms for multi-omics data integration promising for future application to phosphoproteomics data.
Collapse
Affiliation(s)
- Giulia Mantini
- Department of Medical Oncology, OncoProteomics Laboratory, CCA 1-60, Amsterdam UMC VUmc-location, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Thang V Pham
- Department of Medical Oncology, OncoProteomics Laboratory, CCA 1-60, Amsterdam UMC VUmc-location, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Sander R Piersma
- Department of Medical Oncology, OncoProteomics Laboratory, CCA 1-60, Amsterdam UMC VUmc-location, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Connie R Jimenez
- Department of Medical Oncology, OncoProteomics Laboratory, CCA 1-60, Amsterdam UMC VUmc-location, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| |
Collapse
|
6
|
Li M, Shin J, Risgaard RD, Parries MJ, Wang J, Chasman D, Liu S, Roy S, Bhattacharyya A, Zhao X. Identification of FMR1-regulated molecular networks in human neurodevelopment. Genome Res 2020; 30:361-374. [PMID: 32179589 PMCID: PMC7111522 DOI: 10.1101/gr.251405.119] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 02/21/2020] [Indexed: 12/17/2022]
Abstract
RNA-binding proteins (RNA-BPs) play critical roles in development and disease to regulate gene expression. However, genome-wide identification of their targets in primary human cells has been challenging. Here, we applied a modified CLIP-seq strategy to identify genome-wide targets of the FMRP translational regulator 1 (FMR1), a brain-enriched RNA-BP, whose deficiency leads to Fragile X Syndrome (FXS), the most prevalent inherited intellectual disability. We identified FMR1 targets in human dorsal and ventral forebrain neural progenitors and excitatory and inhibitory neurons differentiated from human pluripotent stem cells. In parallel, we measured the transcriptomes of the same four cell types upon FMR1 gene deletion. We discovered that FMR1 preferentially binds long transcripts in human neural cells. FMR1 targets include genes unique to human neural cells and associated with clinical phenotypes of FXS and autism. Integrative network analysis using graph diffusion and multitask clustering of FMR1 CLIP-seq and transcriptional targets reveals critical pathways regulated by FMR1 in human neural development. Our results demonstrate that FMR1 regulates a common set of targets among different neural cell types but also operates in a cell type-specific manner targeting distinct sets of genes in human excitatory and inhibitory neural progenitors and neurons. By defining molecular subnetworks and validating specific high-priority genes, we identify novel components of the FMR1 regulation program. Our results provide new insights into gene regulation by a critical neuronal RNA-BP in human neurodevelopment.
Collapse
Affiliation(s)
- Meng Li
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.,Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Junha Shin
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Ryan D Risgaard
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.,Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Molly J Parries
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.,Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Jianyi Wang
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.,Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Deborah Chasman
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Shuang Liu
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Sushmita Roy
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.,Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.,Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA.,Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
7
|
Chiang S, Shinohara H, Huang JH, Tsai HK, Okada M. Inferring the transcriptional regulatory mechanism of signal-dependent gene expression via an integrative computational approach. FEBS Lett 2020; 594:1477-1496. [PMID: 32052437 DOI: 10.1002/1873-3468.13757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/26/2019] [Accepted: 01/20/2020] [Indexed: 11/10/2022]
Abstract
Eukaryotic transcription factors (TFs) coordinate different upstream signals to regulate the expression of their target genes. To unveil this regulatory network in B-cell receptor signaling, we developed a computational pipeline to systematically analyze the extracellular signal-regulated kinase (ERK)- and IκB kinase (IKK)-dependent transcriptome responses. We combined a bilinear regression method and kinetic modeling to identify the signal-to-TF and TF-to-gene dynamics, respectively. We input a set of time-course experimental data for B cells and concentrated on transcriptional activators. The results show that the combination of TFs differentially controlled by ERK and IKK could contribute divergent expression dynamics in orchestrating the B-cell response. Our findings provide insights into the regulatory mechanisms underlying signal-dependent gene expression in eukaryotic cells.
Collapse
Affiliation(s)
- Sufeng Chiang
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan.,Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | | | - Jia-Hsin Huang
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Huai-Kuang Tsai
- Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, Taiwan.,Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Mariko Okada
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory of Cell Systems, Institute for Protein Research, Osaka University, Suita, Japan
| |
Collapse
|
8
|
Bag AK, Mandloi S, Jarmalavicius S, Mondal S, Kumar K, Mandal C, Walden P, Chakrabarti S, Mandal C. Connecting signaling and metabolic pathways in EGF receptor-mediated oncogenesis of glioblastoma. PLoS Comput Biol 2019; 15:e1007090. [PMID: 31386654 PMCID: PMC6684045 DOI: 10.1371/journal.pcbi.1007090] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 05/13/2019] [Indexed: 12/21/2022] Open
Abstract
As malignant transformation requires synchronization of growth-driving signaling (S) and metabolic (M) pathways, defining cancer-specific S-M interconnected networks (SMINs) could lead to better understanding of oncogenic processes. In a systems-biology approach, we developed a mathematical model for SMINs in mutated EGF receptor (EGFRvIII) compared to wild-type EGF receptor (EGFRwt) expressing glioblastoma multiforme (GBM). Starting with experimentally validated human protein-protein interactome data for S-M pathways, and incorporating proteomic data for EGFRvIII and EGFRwt GBM cells and patient transcriptomic data, we designed a dynamic model for EGFR-driven GBM-specific information flow. Key nodes and paths identified by in silico perturbation were validated experimentally when inhibition of signaling pathway proteins altered expression of metabolic proteins as predicted by the model. This demonstrated capacity of the model to identify unknown connections between signaling and metabolic pathways, explain the robustness of oncogenic SMINs, predict drug escape, and assist identification of drug targets and the development of combination therapies. Complex and highly dynamic interconnected networks allow cancer to take different routes and circumvent chemotherapy. Therefore, understanding these context-specific networks and their dynamics of molecular interactions driven by different oncogenic signaling and metabolic pathways is very much needed to predict drug targets and the effect of therapeutics. We incorporated high-throughput transcriptome and proteome data into mathematical models to deduce properties of cancer cells through systems biology approach. Here we report the development, testing and validation of an integrated systems biology model of information flow between signaling and metabolic pathways to understand the regulation of the interconnection between them in cancer. Our model efficiently identified unique connections and key nodes important in signaling-metabolic information flow. We predicted some potential novel targets before performing actual drug tests. We have successfully applied this model to identify the interconnections altered in the constitutive signaling of the mutated EGFR by comparing EGF-dependent and wild-type EGFR signaling in glioblastoma multiforme.
Collapse
Affiliation(s)
- Arup K. Bag
- Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Sapan Mandloi
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Saulius Jarmalavicius
- Department of Dermatology, Venerology and Allergology, Charité– Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susmita Mondal
- Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Krishna Kumar
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Chhabinath Mandal
- National Institute of Pharmaceutical Education and Research, Kolkata, India
| | - Peter Walden
- Department of Dermatology, Venerology and Allergology, Charité– Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- * E-mail: (PW); , (SC); , (CM)
| | - Saikat Chakrabarti
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Kolkata, India
- * E-mail: (PW); , (SC); , (CM)
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
- * E-mail: (PW); , (SC); , (CM)
| |
Collapse
|
9
|
Reece AS, Hulse GK. Impacts of cannabinoid epigenetics on human development: reflections on Murphy et. al. 'cannabinoid exposure and altered DNA methylation in rat and human sperm' epigenetics 2018; 13: 1208-1221. Epigenetics 2019; 14:1041-1056. [PMID: 31293213 PMCID: PMC6773386 DOI: 10.1080/15592294.2019.1633868] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent data from the Kollins lab (‘Cannabinoid exposure and altered DNA methylation in rat and human sperm’ Epigenetics 2018; 13: 1208–1221) indicated epigenetic effects of cannabis use on sperm in man parallel those in rats and showed substantial shifts in both hypo- and hyper-DNA methylation with the latter predominating. This provides one likely mechanism for the transgenerational transmission of epigenomic instability with sperm as the vector. It therefore contributes important pathophysiological insights into the probable mechanisms underlying the epidemiology of prenatal cannabis exposure potentially explaining diverse features of cannabis-related teratology including effects on the neuraxis, cardiovasculature, immune stimulation, secondary genomic instability and carcinogenesis related to both adult and pediatric cancers. The potentially inheritable and therefore multigenerational nature of these defects needs to be carefully considered in the light of recent teratological and neurobehavioural trends in diverse jurisdictions such as the USA nationally, Hawaii, Colorado, Canada, France and Australia, particularly relating to mental retardation, age-related morbidity and oncogenesis including inheritable cancerogenesis. Increasing demonstrations that the epigenome can respond directly and in real time and retain memories of environmental exposures of many kinds implies that the genome-epigenome is much more sensitive to environmental toxicants than has been generally realized. Issues of long-term multigenerational inheritance amplify these concerns. Further research particularly on the epigenomic toxicology of many cannabinoids is also required.
Collapse
Affiliation(s)
- Albert Stuart Reece
- Division of Psychiatry, University of Western Australia , Crawley , Western Australia Australia.,School of Medical and Health Sciences, Edith Cowan University , Joondalup , Western Australia , Australia
| | - Gary Kenneth Hulse
- Division of Psychiatry, University of Western Australia , Crawley , Western Australia Australia.,School of Medical and Health Sciences, Edith Cowan University , Joondalup , Western Australia , Australia
| |
Collapse
|
10
|
Schwartz GW, Petrovic J, Zhou Y, Faryabi RB. Differential Integration of Transcriptome and Proteome Identifies Pan-Cancer Prognostic Biomarkers. Front Genet 2018; 9:205. [PMID: 29971090 PMCID: PMC6018483 DOI: 10.3389/fgene.2018.00205] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022] Open
Abstract
High-throughput analysis of the transcriptome and proteome individually are used to interrogate complex oncogenic processes in cancer. However, an outstanding challenge is how to combine these complementary, yet partially disparate data sources to accurately identify tumor-specific gene products and clinical biomarkers. Here, we introduce inteGREAT for robust and scalable differential integration of high-throughput measurements. With inteGREAT, each data source is represented as a co-expression network, which is analyzed to characterize the local and global structure of each node across networks. inteGREAT scores the degree by which the topology of each gene in both transcriptome and proteome networks are conserved within a tumor type, yet different from other normal or malignant cells. We demonstrated the high performance of inteGREAT based on several analyses: deconvolving synthetic networks, rediscovering known diagnostic biomarkers, establishing relationships between tumor lineages, and elucidating putative prognostic biomarkers which we experimentally validated. Furthermore, we introduce the application of a clumpiness measure to quantitatively describe tumor lineage similarity. Together, inteGREAT not only infers functional and clinical insights from the integration of transcriptomic and proteomic data sources in cancer, but also can be readily applied to other heterogeneous high-throughput data sources. inteGREAT is open source and available to download from https://github.com/faryabib/inteGREAT.
Collapse
Affiliation(s)
- Gregory W. Schwartz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Jelena Petrovic
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Yeqiao Zhou
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Robert B. Faryabi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Institute for Biomedical Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
11
|
Mertins P, Przybylski D, Yosef N, Qiao J, Clauser K, Raychowdhury R, Eisenhaure TM, Maritzen T, Haucke V, Satoh T, Akira S, Carr SA, Regev A, Hacohen N, Chevrier N. An Integrative Framework Reveals Signaling-to-Transcription Events in Toll-like Receptor Signaling. Cell Rep 2018; 19:2853-2866. [PMID: 28658630 DOI: 10.1016/j.celrep.2017.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 04/11/2017] [Accepted: 06/01/2017] [Indexed: 10/19/2022] Open
Abstract
Building an integrated view of cellular responses to environmental cues remains a fundamental challenge due to the complexity of intracellular networks in mammalian cells. Here, we introduce an integrative biochemical and genetic framework to dissect signal transduction events using multiple data types and, in particular, to unify signaling and transcriptional networks. Using the Toll-like receptor (TLR) system as a model cellular response, we generate multifaceted datasets on physical, enzymatic, and functional interactions and integrate these data to reveal biochemical paths that connect TLR4 signaling to transcription. We define the roles of proximal TLR4 kinases, identify and functionally test two dozen candidate regulators, and demonstrate a role for Ap1ar (encoding the Gadkin protein) and its binding partner, Picalm, potentially linking vesicle transport with pro-inflammatory responses. Our study thus demonstrates how deciphering dynamic cellular responses by integrating datasets on various regulatory layers defines key components and higher-order logic underlying signaling-to-transcription pathways.
Collapse
Affiliation(s)
- Philipp Mertins
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Dariusz Przybylski
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Nir Yosef
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA; Department of Electrical Engineering and Computer Science and Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jana Qiao
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Karl Clauser
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | | | - Thomas M Eisenhaure
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Tanja Maritzen
- Molecular Physiology and Cell Biology Section, Leibniz-Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Volker Haucke
- Molecular Physiology and Cell Biology Section, Leibniz-Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Takashi Satoh
- WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Shizuo Akira
- WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Steven A Carr
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Department of Biology, MIT, Cambridge, MA 02142, USA.
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA; Center for Immunology and Inflammatory Diseases and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Nicolas Chevrier
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA.
| |
Collapse
|
12
|
Kedaigle AJ, Fraenkel E. Discovering Altered Regulation and Signaling Through Network-based Integration of Transcriptomic, Epigenomic, and Proteomic Tumor Data. Methods Mol Biol 2018; 1711:13-26. [PMID: 29344883 PMCID: PMC6309679 DOI: 10.1007/978-1-4939-7493-1_2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
With the extraordinary rise in available biological data, biologists and clinicians need unbiased tools for data integration in order to reach accurate, succinct conclusions. Network biology provides one such method for high-throughput data integration, but comes with its own set of algorithmic problems and needed expertise. We provide a step-by-step guide for using Omics Integrator, a software package designed for the integration of transcriptomic, epigenomic, and proteomic data. Omics Integrator can be found at http://fraenkel.mit.edu/omicsintegrator .
Collapse
Affiliation(s)
- Amanda J Kedaigle
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ernest Fraenkel
- Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
13
|
Ursu O, Gosline SJC, Beeharry N, Fink L, Bhattacharjee V, Huang SSC, Zhou Y, Yen T, Fraenkel E. Network modeling of kinase inhibitor polypharmacology reveals pathways targeted in chemical screens. PLoS One 2017; 12:e0185650. [PMID: 29023490 PMCID: PMC5638242 DOI: 10.1371/journal.pone.0185650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 09/15/2017] [Indexed: 01/22/2023] Open
Abstract
Small molecule screens are widely used to prioritize pharmaceutical development. However, determining the pathways targeted by these molecules is challenging, since the compounds are often promiscuous. We present a network strategy that takes into account the polypharmacology of small molecules in order to generate hypotheses for their broader mode of action. We report a screen for kinase inhibitors that increase the efficacy of gemcitabine, the first-line chemotherapy for pancreatic cancer. Eight kinase inhibitors emerge that are known to affect 201 kinases, of which only three kinases have been previously identified as modifiers of gemcitabine toxicity. In this work, we use the SAMNet algorithm to identify pathways linking these kinases and genetic modifiers of gemcitabine toxicity with transcriptional and epigenetic changes induced by gemcitabine that we measure using DNaseI-seq and RNA-seq. SAMNet uses a constrained optimization algorithm to connect genes from these complementary datasets through a small set of protein-protein and protein-DNA interactions. The resulting network recapitulates known pathways including DNA repair, cell proliferation and the epithelial-to-mesenchymal transition. We use the network to predict genes with important roles in the gemcitabine response, including six that have already been shown to modify gemcitabine efficacy in pancreatic cancer and ten novel candidates. Our work reveals the important role of polypharmacology in the activity of these chemosensitizing agents.
Collapse
Affiliation(s)
- Oana Ursu
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Sara J. C. Gosline
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Neil Beeharry
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Lauren Fink
- Cancer Biology Program, Fox Chase Cancer Center; Philadelphia, Pennsylvania, United States of America
| | | | - Shao-shan Carol Huang
- Plant Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Yan Zhou
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Tim Yen
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
14
|
Sychev ZE, Hu A, DiMaio TA, Gitter A, Camp ND, Noble WS, Wolf-Yadlin A, Lagunoff M. Integrated systems biology analysis of KSHV latent infection reveals viral induction and reliance on peroxisome mediated lipid metabolism. PLoS Pathog 2017; 13:e1006256. [PMID: 28257516 PMCID: PMC5352148 DOI: 10.1371/journal.ppat.1006256] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 03/15/2017] [Accepted: 02/22/2017] [Indexed: 12/22/2022] Open
Abstract
Kaposi’s Sarcoma associated Herpesvirus (KSHV), an oncogenic, human gamma-herpesvirus, is the etiological agent of Kaposi’s Sarcoma the most common tumor of AIDS patients world-wide. KSHV is predominantly latent in the main KS tumor cell, the spindle cell, a cell of endothelial origin. KSHV modulates numerous host cell-signaling pathways to activate endothelial cells including major metabolic pathways involved in lipid metabolism. To identify the underlying cellular mechanisms of KSHV alteration of host signaling and endothelial cell activation, we identified changes in the host proteome, phosphoproteome and transcriptome landscape following KSHV infection of endothelial cells. A Steiner forest algorithm was used to integrate the global data sets and, together with transcriptome based predicted transcription factor activity, cellular networks altered by latent KSHV were predicted. Several interesting pathways were identified, including peroxisome biogenesis. To validate the predictions, we showed that KSHV latent infection increases the number of peroxisomes per cell. Additionally, proteins involved in peroxisomal lipid metabolism of very long chain fatty acids, including ABCD3 and ACOX1, are required for the survival of latently infected cells. In summary, novel cellular pathways altered during herpesvirus latency that could not be predicted by a single systems biology platform, were identified by integrated proteomics and transcriptomics data analysis and when correlated with our metabolomics data revealed that peroxisome lipid metabolism is essential for KSHV latent infection of endothelial cells. Kaposi’s Sarcoma herpesvirus (KSHV) is the etiologic agent of Kaposi’s Sarcoma, the most common tumor of AIDS patients. KSHV modulates host cell signaling and metabolism to maintain a life-long latent infection. To unravel the underlying cellular mechanisms modulated by KSHV, we used multiple global systems biology platforms to identify and integrate changes in both cellular protein expression and transcription following KSHV infection of endothelial cells, the relevant cell type for KS tumors. The analysis identified several interesting pathways including peroxisome biogenesis. Peroxisomes are small cytoplasmic organelles involved in redox reactions and lipid metabolism. KSHV latent infection increases the number of peroxisomes per cell and proteins involved in peroxisomal lipid metabolism are required for the survival of latently infected cells. In summary, through integration of multiple global systems biology analyses we were able to identify novel pathways that could not be predicted by one platform alone and found that lipid metabolism in a small cytoplasmic organelle is necessary for the survival of latent infection with a herpesvirus.
Collapse
Affiliation(s)
- Zoi E. Sychev
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Alex Hu
- Department of Genome Science, University of Washington, Seattle, Washington, United States of America
| | - Terri A. DiMaio
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison and Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Nathan D. Camp
- Department of Genome Science, University of Washington, Seattle, Washington, United States of America
| | - William S. Noble
- Department of Genome Science, University of Washington, Seattle, Washington, United States of America
| | - Alejandro Wolf-Yadlin
- Department of Genome Science, University of Washington, Seattle, Washington, United States of America
- * E-mail: (ML); (AWY)
| | - Michael Lagunoff
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (ML); (AWY)
| |
Collapse
|
15
|
Vyse S, Desmond H, Huang PH. Advances in mass spectrometry based strategies to study receptor tyrosine kinases. IUCRJ 2017; 4:119-130. [PMID: 28250950 PMCID: PMC5330522 DOI: 10.1107/s2052252516020546] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/27/2016] [Indexed: 06/06/2023]
Abstract
Receptor tyrosine kinases (RTKs) are key transmembrane environmental sensors that are capable of transmitting extracellular information into phenotypic responses, including cell proliferation, survival and metabolism. Advances in mass spectrometry (MS)-based phosphoproteomics have been instrumental in providing the foundations of much of our current understanding of RTK signalling networks and activation dynamics. Furthermore, new insights relating to the deregulation of RTKs in disease, for instance receptor co-activation and kinome reprogramming, have largely been identified using phosphoproteomic-based strategies. This review outlines the current approaches employed in phosphoproteomic workflows, including phosphopeptide enrichment and MS data-acquisition methods. Here, recent advances in the application of MS-based phosphoproteomics to bridge critical gaps in our knowledge of RTK signalling are focused on. The current limitations of the technology are discussed and emerging areas such as computational modelling, high-throughput phospho-proteomic workflows and next-generation single-cell approaches to further our understanding in new areas of RTK biology are highlighted.
Collapse
Affiliation(s)
- Simon Vyse
- Division of Cancer Biology, The Institute of Cancer Research, London SW3 6JB, England
| | - Howard Desmond
- Division of Cancer Biology, The Institute of Cancer Research, London SW3 6JB, England
| | - Paul H. Huang
- Division of Cancer Biology, The Institute of Cancer Research, London SW3 6JB, England
| |
Collapse
|
16
|
Kang UB, Marto JA. Leucine-rich repeat kinase 2 and Parkinson's disease. Proteomics 2016; 17. [PMID: 27723254 DOI: 10.1002/pmic.201600092] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 12/21/2022]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a large multidomain protein that is expressed in many tissues and participates in numerous biological pathways. Mutations in LRRK2 are recognized as genetic risk factors for familial Parkinson's disease (PD) and may also represent causal factors in the more common sporadic form of PD. The structure of LRRK2 comprises a combination of GTPase, kinase, and scaffolding domains. This functional diversity, combined with a potentially central role in genetic and idiopathic PD motivates significant effort to further credential LRRK2 as a therapeutic target. Here, we review the current understanding for LRRK2 function in normal physiology and PD, with emphasis on insight gained from proteomic approaches.
Collapse
Affiliation(s)
- Un-Beom Kang
- Department of Cancer Biology and Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jarrod A Marto
- Department of Cancer Biology and Blais Proteomics Center, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Drake JM, Paull EO, Graham NA, Lee JK, Smith BA, Titz B, Stoyanova T, Faltermeier CM, Uzunangelov V, Carlin DE, Fleming DT, Wong CK, Newton Y, Sudha S, Vashisht AA, Huang J, Wohlschlegel JA, Graeber TG, Witte ON, Stuart JM. Phosphoproteome Integration Reveals Patient-Specific Networks in Prostate Cancer. Cell 2016; 166:1041-1054. [PMID: 27499020 DOI: 10.1016/j.cell.2016.07.007] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/15/2016] [Accepted: 07/07/2016] [Indexed: 12/19/2022]
Abstract
We used clinical tissue from lethal metastatic castration-resistant prostate cancer (CRPC) patients obtained at rapid autopsy to evaluate diverse genomic, transcriptomic, and phosphoproteomic datasets for pathway analysis. Using Tied Diffusion through Interacting Events (TieDIE), we integrated differentially expressed master transcriptional regulators, functionally mutated genes, and differentially activated kinases in CRPC tissues to synthesize a robust signaling network consisting of druggable kinase pathways. Using MSigDB hallmark gene sets, six major signaling pathways with phosphorylation of several key residues were significantly enriched in CRPC tumors after incorporation of phosphoproteomic data. Individual autopsy profiles developed using these hallmarks revealed clinically relevant pathway information potentially suitable for patient stratification and targeted therapies in late stage prostate cancer. Here, we describe phosphorylation-based cancer hallmarks using integrated personalized signatures (pCHIPS) that shed light on the diversity of activated signaling pathways in metastatic CRPC while providing an integrative, pathway-based reference for drug prioritization in individual patients.
Collapse
Affiliation(s)
- Justin M Drake
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Rutgers Cancer Institute of New Jersey and Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA.
| | - Evan O Paull
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Nicholas A Graham
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - John K Lee
- Division of Hematology and Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bryan A Smith
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bjoern Titz
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tanya Stoyanova
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA 94304, USA
| | - Claire M Faltermeier
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vladislav Uzunangelov
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Daniel E Carlin
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel Teo Fleming
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Christopher K Wong
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Yulia Newton
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sud Sudha
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ajay A Vashisht
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jiaoti Huang
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas G Graeber
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Owen N Witte
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Joshua M Stuart
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
18
|
Nagaraj AB, Joseph P, Kovalenko O, Singh S, Armstrong A, Redline R, Resnick K, Zanotti K, Waggoner S, DiFeo A. Critical role of Wnt/β-catenin signaling in driving epithelial ovarian cancer platinum resistance. Oncotarget 2016; 6:23720-34. [PMID: 26125441 PMCID: PMC4695147 DOI: 10.18632/oncotarget.4690] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/01/2015] [Indexed: 01/06/2023] Open
Abstract
Resistance to platinum-based chemotherapy is the major barrier to treating epithelial ovarian cancer. To improve patient outcomes, it is critical to identify the underlying mechanisms that promote platinum resistance. Emerging evidence supports the concept that platinum-based therapies are able to eliminate the bulk of differentiated cancer cells, but are unable to eliminate cancer initiating cells (CIC). To date, the relevant pathways that regulate ovarian CICs remain elusive. Several correlative studies have shown that Wnt/β-catenin pathway activation is associated with poor outcomes in patients with high-grade serous ovarian cancer (HGSOC). However, the functional relevance of these findings remain to be delineated. We have uncovered that Wnt/β-catenin pathway activation is a critical driver of HGSOC chemotherapy resistance, and targeted inhibition of this pathway, which eliminates CICs, represents a novel and effective treatment for chemoresistant HGSOC. Here we show that Wnt/β-catenin signaling is activated in ovarian CICs, and targeted inhibition of β-catenin potently sensitized cells to cisplatin and decreased CIC tumor sphere formation. Furthermore, the Wnt/β-catenin specific inhibitor iCG-001 potently sensitized cells to cisplatin and decreased stem-cell frequency in platinum resistant cells. Taken together, our data is the first report providing evidence that the Wnt/β-catenin signaling pathway maintains stem-like properties and drug resistance of primary HGSOC PDX derived platinum resistant models, and therapeutic targeting of this pathway with iCG-001/PRI-724, which has been shown to be well tolerated in Phase I trials, may be an effective treatment option.
Collapse
Affiliation(s)
- Anil Belur Nagaraj
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Peronne Joseph
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Olga Kovalenko
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Sareena Singh
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Amy Armstrong
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Raymond Redline
- Department of Pathology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Kimberly Resnick
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Kristine Zanotti
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Steven Waggoner
- Department of Gynecology, Division of Gynecological Oncology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Analisa DiFeo
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
19
|
Revealing disease-associated pathways by network integration of untargeted metabolomics. Nat Methods 2016; 13:770-6. [PMID: 27479327 DOI: 10.1038/nmeth.3940] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 06/17/2016] [Indexed: 12/11/2022]
Abstract
Uncovering the molecular context of dysregulated metabolites is crucial to understand pathogenic pathways. However, their system-level analysis has been limited owing to challenges in global metabolite identification. Most metabolite features detected by untargeted metabolomics carried out by liquid-chromatography-mass spectrometry cannot be uniquely identified without additional, time-consuming experiments. We report a network-based approach, prize-collecting Steiner forest algorithm for integrative analysis of untargeted metabolomics (PIUMet), that infers molecular pathways and components via integrative analysis of metabolite features, without requiring their identification. We demonstrated PIUMet by analyzing changes in metabolism of sphingolipids, fatty acids and steroids in a Huntington's disease model. Additionally, PIUMet enabled us to elucidate putative identities of altered metabolite features in diseased cells, and infer experimentally undetected, disease-associated metabolites and dysregulated proteins. Finally, we established PIUMet's ability for integrative analysis of untargeted metabolomics data with proteomics data, demonstrating that this approach elicits disease-associated metabolites and proteins that cannot be inferred by individual analysis of these data.
Collapse
|
20
|
Network Modeling Identifies Patient-specific Pathways in Glioblastoma. Sci Rep 2016; 6:28668. [PMID: 27354287 PMCID: PMC4926112 DOI: 10.1038/srep28668] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/08/2016] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma is the most aggressive type of malignant human brain tumor. Molecular profiling experiments have revealed that these tumors are extremely heterogeneous. This heterogeneity is one of the principal challenges for developing targeted therapies. We hypothesize that despite the diverse molecular profiles, it might still be possible to identify common signaling changes that could be targeted in some or all tumors. Using a network modeling approach, we reconstruct the altered signaling pathways from tumor-specific phosphoproteomic data and known protein-protein interactions. We then develop a network-based strategy for identifying tumor specific proteins and pathways that were predicted by the models but not directly observed in the experiments. Among these hidden targets, we show that the ERK activator kinase1 (MEK1) displays increased phosphorylation in all tumors. By contrast, protein numb homolog (NUMB) is present only in the subset of the tumors that are the most invasive. Additionally, increased S100A4 is associated with only one of the tumors. Overall, our results demonstrate that despite the heterogeneity of the proteomic data, network models can identify common or tumor specific pathway-level changes. These results represent an important proof of principle that can improve the target selection process for tumor specific treatments.
Collapse
|
21
|
Chitforoushzadeh Z, Ye Z, Sheng Z, LaRue S, Fry RC, Lauffenburger DA, Janes KA. TNF-insulin crosstalk at the transcription factor GATA6 is revealed by a model that links signaling and transcriptomic data tensors. Sci Signal 2016; 9:ra59. [PMID: 27273097 DOI: 10.1126/scisignal.aad3373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Signal transduction networks coordinate transcriptional programs activated by diverse extracellular stimuli, such as growth factors and cytokines. Cells receive multiple stimuli simultaneously, and mapping how activation of the integrated signaling network affects gene expression is a challenge. We stimulated colon adenocarcinoma cells with various combinations of the cytokine tumor necrosis factor (TNF) and the growth factors insulin and epidermal growth factor (EGF) to investigate signal integration and transcriptional crosstalk. We quantitatively linked the proteomic and transcriptomic data sets by implementing a structured computational approach called tensor partial least squares regression. This statistical model accurately predicted transcriptional signatures from signaling arising from single and combined stimuli and also predicted time-dependent contributions of signaling events. Specifically, the model predicted that an early-phase, AKT-associated signal downstream of insulin repressed a set of transcripts induced by TNF. Through bioinformatics and cell-based experiments, we identified the AKT-repressed signal as glycogen synthase kinase 3 (GSK3)-catalyzed phosphorylation of Ser(37) on the long form of the transcription factor GATA6. Phosphorylation of GATA6 on Ser(37) promoted its degradation, thereby preventing GATA6 from repressing transcripts that are induced by TNF and attenuated by insulin. Our analysis showed that predictive tensor modeling of proteomic and transcriptomic data sets can uncover pathway crosstalk that produces specific patterns of gene expression in cells receiving multiple stimuli.
Collapse
Affiliation(s)
- Zeinab Chitforoushzadeh
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA. Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Zi Ye
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Ziran Sheng
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Silvia LaRue
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
22
|
Tuncbag N, Gosline SJC, Kedaigle A, Soltis AR, Gitter A, Fraenkel E. Network-Based Interpretation of Diverse High-Throughput Datasets through the Omics Integrator Software Package. PLoS Comput Biol 2016; 12:e1004879. [PMID: 27096930 PMCID: PMC4838263 DOI: 10.1371/journal.pcbi.1004879] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 03/23/2016] [Indexed: 02/07/2023] Open
Abstract
High-throughput, ‘omic’ methods provide sensitive measures of biological responses to perturbations. However, inherent biases in high-throughput assays make it difficult to interpret experiments in which more than one type of data is collected. In this work, we introduce Omics Integrator, a software package that takes a variety of ‘omic’ data as input and identifies putative underlying molecular pathways. The approach applies advanced network optimization algorithms to a network of thousands of molecular interactions to find high-confidence, interpretable subnetworks that best explain the data. These subnetworks connect changes observed in gene expression, protein abundance or other global assays to proteins that may not have been measured in the screens due to inherent bias or noise in measurement. This approach reveals unannotated molecular pathways that would not be detectable by searching pathway databases. Omics Integrator also provides an elegant framework to incorporate not only positive data, but also negative evidence. Incorporating negative evidence allows Omics Integrator to avoid unexpressed genes and avoid being biased toward highly-studied hub proteins, except when they are strongly implicated by the data. The software is comprised of two individual tools, Garnet and Forest, that can be run together or independently to allow a user to perform advanced integration of multiple types of high-throughput data as well as create condition-specific subnetworks of protein interactions that best connect the observed changes in various datasets. It is available at http://fraenkel.mit.edu/omicsintegrator and on GitHub at https://github.com/fraenkel-lab/OmicsIntegrator.
Collapse
Affiliation(s)
- Nurcan Tuncbag
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Sara J. C. Gosline
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Amanda Kedaigle
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Anthony R. Soltis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Anthony Gitter
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
23
|
Suwala AK, Hanaford A, Kahlert UD, Maciaczyk J. Clipping the Wings of Glioblastoma: Modulation of WNT as a Novel Therapeutic Strategy. J Neuropathol Exp Neurol 2016; 75:388-96. [PMID: 26979081 DOI: 10.1093/jnen/nlw013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant brain tumor and has a dismal prognosis. Aberrant WNT signaling is known to promote glioma cell growth and dissemination and resistance to conventional radio- and chemotherapy. Moreover, a population of cancer stem-like cells that promote glioma growth and recurrence are strongly dependent on WNT signaling. Here, we discuss the role and mechanisms of aberrant canonical and noncanonical WNT signaling in GBM. We present current clinical approaches aimed at modulating WNT activity and evaluate their clinical perspective as a novel treatment option for GBM.
Collapse
Affiliation(s)
- Abigail K Suwala
- From the Department of Neurosurgery, University Medical Center Düsseldorf, Düsseldorf, Germany (AKS, UDK, JM); and Division of Neuropathology, Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (AH)
| | - Allison Hanaford
- From the Department of Neurosurgery, University Medical Center Düsseldorf, Düsseldorf, Germany (AKS, UDK, JM); and Division of Neuropathology, Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (AH)
| | - Ulf D Kahlert
- From the Department of Neurosurgery, University Medical Center Düsseldorf, Düsseldorf, Germany (AKS, UDK, JM); and Division of Neuropathology, Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (AH)
| | - Jaroslaw Maciaczyk
- From the Department of Neurosurgery, University Medical Center Düsseldorf, Düsseldorf, Germany (AKS, UDK, JM); and Division of Neuropathology, Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland (AH).
| |
Collapse
|
24
|
Abstract
Currently, gliomas are diagnosed by neuroimaging, and refined diagnosis requires resection or biopsy to obtain tumour tissue for histopathological classification and grading. Blood-derived biomarkers, therefore, would be useful as minimally invasive markers that could support diagnosis and enable monitoring of tumour growth and response to treatment. Such circulating biomarkers could distinguish true progression from therapy-associated changes such as radiation necrosis, and help evaluate the persistence or disappearance of a therapeutic target, such as an oncoprotein or a targetable gene mutation, after targeted therapy. Unlike for other tumours, circulating biomarkers for gliomas are still being defined and are not yet in use in clinical practice. Circulating tumour DNA (ctDNA) isolated from plasma has been shown to reflect the mutational status of glioblastoma, and extracellular vesicles (EVs) containing ctDNA, microRNA and proteins function as rapidly adapting reservoirs for glioma biomarkers such as typical DNA mutations, regulatory microRNAs and oncoproteins. Ideally, circulating tumour cells could enable profiling of the whole-tumour genome, but they are difficult to detect and can reflect only a single cell type of the heterogeneous tumour composition, whereas EVs reflect the complex heterogeneity of the whole tumour, as well as its adaptations to therapy. Although all categories of potential blood-derived biomarkers need to be developed further, findings from other tumour types suggest that EVs are the most promising biomarkers.
Collapse
|
25
|
Terfve CDA, Wilkes EH, Casado P, Cutillas PR, Saez-Rodriguez J. Large-scale models of signal propagation in human cells derived from discovery phosphoproteomic data. Nat Commun 2015; 6:8033. [PMID: 26354681 PMCID: PMC4579397 DOI: 10.1038/ncomms9033] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/09/2015] [Indexed: 12/27/2022] Open
Abstract
Mass spectrometry is widely used to probe the proteome and its modifications in an untargeted manner, with unrivalled coverage. Applied to phosphoproteomics, it has tremendous potential to interrogate phospho-signalling and its therapeutic implications. However, this task is complicated by issues of undersampling of the phosphoproteome and challenges stemming from its high-content but low-sample-throughput nature. Hence, methods using such data to reconstruct signalling networks have been limited to restricted data sets and insights (for example, groups of kinases likely to be active in a sample). We propose a new method to handle high-content discovery phosphoproteomics data on perturbation by putting it in the context of kinase/phosphatase-substrate knowledge, from which we derive and train logic models. We show, on a data set obtained through perturbations of cancer cells with small-molecule inhibitors, that this method can study the targets and effects of kinase inhibitors, and reconcile insights obtained from multiple data sets, a common issue with these data. Phosphoproteomics can offer significant insight into cell signalling and how signalling is modified in response to perturbations. Here the authors develop a new tool for the analysis of high-content phosphoproteomics in the context of kinase/phosphatase-substrate knowledge, which is used to train logic models.
Collapse
Affiliation(s)
- Camille D A Terfve
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Edmund H Wilkes
- Integrative Cell Signalling and Proteomics, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Pedro Casado
- Integrative Cell Signalling and Proteomics, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Pedro R Cutillas
- Integrative Cell Signalling and Proteomics, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| |
Collapse
|
26
|
Abstract
Cell signaling pathways control cells' responses to their environment through an intricate network of proteins and small molecules partitioned by intracellular structures, such as the cytoskeleton and nucleus. Our understanding of these pathways has been revised recently with the advent of more advanced experimental techniques; no longer are signaling pathways viewed as linear cascades of information flowing from membrane-bound receptors to the nucleus. Instead, such pathways must be understood in the context of networks, and studying such networks requires an integration of computational and experimental approaches. This understanding is becoming more important in designing novel therapies for diseases such as cancer. Using the MAPK (mitogen-activated protein kinase) and PI3K (class I phosphoinositide-3' kinase) pathways as case studies of cellular signaling, we give an overview of these pathways and their functions. We then describe, using a number of case studies, how computational modeling has aided in understanding these pathways' deregulation in cancer, and how such understanding can be used to optimally tailor current therapies or help design new therapies against cancer.
Collapse
Affiliation(s)
- Julio Saez-Rodriguez
- Current address: Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, D-52074 Aachen, Germany;
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SD, United Kingdom;
| | - Aidan MacNamara
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire CB10 1SD, United Kingdom;
| | - Simon Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom;
| |
Collapse
|
27
|
Budak G, Eren Ozsoy O, Aydin Son Y, Can T, Tuncbag N. Reconstruction of the temporal signaling network in Salmonella-infected human cells. Front Microbiol 2015; 6:730. [PMID: 26257716 PMCID: PMC4507143 DOI: 10.3389/fmicb.2015.00730] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/03/2015] [Indexed: 12/02/2022] Open
Abstract
Salmonella enterica is a bacterial pathogen that usually infects its host through food sources. Translocation of the pathogen proteins into the host cells leads to changes in the signaling mechanism either by activating or inhibiting the host proteins. Given that the bacterial infection modifies the response network of the host, a more coherent view of the underlying biological processes and the signaling networks can be obtained by using a network modeling approach based on the reverse engineering principles. In this work, we have used a published temporal phosphoproteomic dataset of Salmonella-infected human cells and reconstructed the temporal signaling network of the human host by integrating the interactome and the phosphoproteomic dataset. We have combined two well-established network modeling frameworks, the Prize-collecting Steiner Forest (PCSF) approach and the Integer Linear Programming (ILP) based edge inference approach. The resulting network conserves the information on temporality, direction of interactions, while revealing hidden entities in the signaling, such as the SNARE binding, mTOR signaling, immune response, cytoskeleton organization, and apoptosis pathways. Targets of the Salmonella effectors in the host cells such as CDC42, RHOA, 14-3-3δ, Syntaxin family, Oxysterol-binding proteins were included in the reconstructed signaling network although they were not present in the initial phosphoproteomic data. We believe that integrated approaches, such as the one presented here, have a high potential for the identification of clinical targets in infectious diseases, especially in the Salmonella infections.
Collapse
Affiliation(s)
- Gungor Budak
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Oyku Eren Ozsoy
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Yesim Aydin Son
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| | - Tolga Can
- Department of Computer Engineering, College of Engineering, Middle East Technical University Ankara, Turkey
| | - Nurcan Tuncbag
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University Ankara, Turkey
| |
Collapse
|
28
|
Kamdje AHN, Etet PFS, Vecchio L, Tagne RS, Amvene JM, Muller JM, Krampera M, Lukong KE. New targeted therapies for breast cancer: A focus on tumor microenvironmental signals and chemoresistant breast cancers. World J Clin Cases 2014; 2:769-86. [PMID: 25516852 PMCID: PMC4266825 DOI: 10.12998/wjcc.v2.i12.769] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/12/2014] [Accepted: 09/23/2014] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the most frequent female malignancy worldwide. Current strategies in breast cancer therapy, including classical chemotherapy, hormone therapy, and targeted therapies, are usually associated with chemoresistance and serious adverse effects. Advances in our understanding of changes affecting the interactome in advanced and chemoresistant breast tumors have provided novel therapeutic targets, including, cyclin dependent kinases, mammalian target of rapamycin, Notch, Wnt and Shh. Inhibitors of these molecules recently entered clinical trials in mono- and combination therapy in metastatic and chemo-resistant breast cancers. Anticancer epigenetic drugs, mainly histone deacetylase inhibitors and DNA methyltransferase inhibitors, also entered clinical trials. Because of the complexity and heterogeneity of breast cancer, the future in therapy lies in the application of individualized tailored regimens. Emerging therapeutic targets and the implications for personalized-based therapy development in breast cancer are herein discussed.
Collapse
|
29
|
Chasman D, Ho YH, Berry DB, Nemec CM, MacGilvray ME, Hose J, Merrill AE, Lee MV, Will JL, Coon JJ, Ansari AZ, Craven M, Gasch AP. Pathway connectivity and signaling coordination in the yeast stress-activated signaling network. Mol Syst Biol 2014; 10:759. [PMID: 25411400 PMCID: PMC4299600 DOI: 10.15252/msb.20145120] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Stressed cells coordinate a multi-faceted response spanning many levels of physiology. Yet
knowledge of the complete stress-activated regulatory network as well as design principles for
signal integration remains incomplete. We developed an experimental and computational approach to
integrate available protein interaction data with gene fitness contributions, mutant transcriptome
profiles, and phospho-proteome changes in cells responding to salt stress, to infer the
salt-responsive signaling network in yeast. The inferred subnetwork presented many novel predictions
by implicating new regulators, uncovering unrecognized crosstalk between known pathways, and
pointing to previously unknown ‘hubs’ of signal integration. We exploited these
predictions to show that Cdc14 phosphatase is a central hub in the network and that modification of
RNA polymerase II coordinates induction of stress-defense genes with reduction of growth-related
transcripts. We find that the orthologous human network is enriched for cancer-causing genes,
underscoring the importance of the subnetwork's predictions in understanding stress
biology.
Collapse
Affiliation(s)
- Deborah Chasman
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yi-Hsuan Ho
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - David B Berry
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Corey M Nemec
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | | | - James Hose
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna E Merrill
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - M Violet Lee
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessica L Will
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA Department of Biological Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Aseem Z Ansari
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark Craven
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, USA Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Audrey P Gasch
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
30
|
Affiliation(s)
- H Steven Wiley
- Pacific Northwest National Laboratory, 902 Battelle Boulevard, K8-96, Richland 99352, WA, USA.
| |
Collapse
|
31
|
Beste MT, Pfäffle-Doyle N, Prentice EA, Morris SN, Lauffenburger DA, Isaacson KB, Griffith LG. Molecular network analysis of endometriosis reveals a role for c-Jun-regulated macrophage activation. Sci Transl Med 2014; 6:222ra16. [PMID: 24500404 DOI: 10.1126/scitranslmed.3007988] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Clinical management of endometriosis is limited by the complex relationship between symptom severity, heterogeneous surgical presentation, and variability in clinical outcomes. As a complement to visual classification schemes, molecular profiles of disease activity may improve risk stratification to better inform treatment decisions and identify new approaches to targeted treatment. We use a network analysis of information flow within and between inflammatory cells to discern consensus behaviors characterizing patient subpopulations. Unsupervised multivariate analysis of cytokine profiles quantified by multiplex immunoassays identified a subset of patients with a shared "consensus signature" of 13 elevated cytokines that was associated with common clinical features of endometriosis, but was not observed among patient subpopulations defined by morphologic presentation alone. Enrichment analysis of consensus markers reinforced the primacy of peritoneal macrophage infiltration and activation, which was demonstrably elevated in ex vivo cultures. Although familiar targets of the nuclear factor κB family emerged among overrepresented transcriptional binding sites for consensus markers, our analysis provides evidence for an unexpected contribution from c-Jun, c-Fos, and AP-1 effectors of mitogen-associated kinase signaling. Their crucial involvement in propagation of macrophage-driven inflammatory networks was confirmed via targeted inhibition of upstream kinases. Collectively, these analyses suggest a clinically relevant inflammatory network that may serve as an objective measure for guiding treatment decisions for endometriosis management, and in the future may provide a mechanistic endpoint for assessing efficacy of new agents aimed at curtailing inflammatory mechanisms that drive disease progression.
Collapse
Affiliation(s)
- Michael T Beste
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Osmanbeyoglu HU, Pelossof R, Bromberg JF, Leslie CS. Linking signaling pathways to transcriptional programs in breast cancer. Genome Res 2014; 24:1869-80. [PMID: 25183703 PMCID: PMC4216927 DOI: 10.1101/gr.173039.114] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer cells acquire genetic and epigenetic alterations that often lead to dysregulation of oncogenic signal transduction pathways, which in turn alters downstream transcriptional programs. Numerous methods attempt to deduce aberrant signaling pathways in tumors from mRNA data alone, but these pathway analysis approaches remain qualitative and imprecise. In this study, we present a statistical method to link upstream signaling to downstream transcriptional response by exploiting reverse phase protein array (RPPA) and mRNA expression data in The Cancer Genome Atlas (TCGA) breast cancer project. Formally, we use an algorithm called affinity regression to learn an interaction matrix between upstream signal transduction proteins and downstream transcription factors (TFs) that explains target gene expression. The trained model can then predict the TF activity, given a tumor sample’s protein expression profile, or infer the signaling protein activity, given a tumor sample’s gene expression profile. Breast cancers are comprised of molecularly distinct subtypes that respond differently to pathway-targeted therapies. We trained our model on the TCGA breast cancer data set and identified subtype-specific and common TF regulators of gene expression. We then used the trained tumor model to predict signaling protein activity in a panel of breast cancer cell lines for which gene expression and drug response data was available. Correlations between inferred protein activities and drug responses in breast cancer cell lines grouped several drugs that are clinically used in combination. Finally, inferred protein activity predicted the clinical outcome within the METABRIC Luminal A cohort, identifying high- and low-risk patient groups within this heterogeneous subtype.
Collapse
Affiliation(s)
- Hatice U Osmanbeyoglu
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Raphael Pelossof
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Jacqueline F Bromberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York 10065, USA
| | - Christina S Leslie
- Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA;
| |
Collapse
|
33
|
Abstract
Phosphorylation of serine, threonine and tyrosine plays significant roles in cellular signal transduction and in modifying multiple protein functions. Phosphoproteins are coordinated and regulated by a network of kinases, phosphatases and phospho-binding proteins, which modify the phosphorylation states, recognize unique phosphopeptides, or target proteins for degradation. Detailed and complete information on the structure and dynamics of these networks is required to better understand fundamental mechanisms of cellular processes and diseases. High-throughput technologies have been developed to investigate phosphoproteomes in model organisms and human diseases. Among them, mass spectrometry (MS)-based technologies are the major platforms and have been widely applied, which has led to explosive growth of phosphoproteomic data in recent years. New bioinformatics tools are needed to analyze and make sense of these data. Moreover, most research has focused on individual phosphoproteins and kinases. To gain a more complete knowledge of cellular processes, systems biology approaches, including pathways and networks modeling, have to be applied to integrate all components of the phosphorylation machinery, including kinases, phosphatases, their substrates, and phospho-binding proteins. This review presents the latest developments of bioinformatics methods and attempts to apply systems biology to analyze phosphoproteomics data generated by MS-based technologies. Challenges and future directions in this field will be also discussed.
Collapse
|
34
|
Xie L, Ge X, Tan H, Xie L, Zhang Y, Hart T, Yang X, Bourne PE. Towards structural systems pharmacology to study complex diseases and personalized medicine. PLoS Comput Biol 2014; 10:e1003554. [PMID: 24830652 PMCID: PMC4022462 DOI: 10.1371/journal.pcbi.1003554] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genome-Wide Association Studies (GWAS), whole genome sequencing, and high-throughput omics techniques have generated vast amounts of genotypic and molecular phenotypic data. However, these data have not yet been fully explored to improve the effectiveness and efficiency of drug discovery, which continues along a one-drug-one-target-one-disease paradigm. As a partial consequence, both the cost to launch a new drug and the attrition rate are increasing. Systems pharmacology and pharmacogenomics are emerging to exploit the available data and potentially reverse this trend, but, as we argue here, more is needed. To understand the impact of genetic, epigenetic, and environmental factors on drug action, we must study the structural energetics and dynamics of molecular interactions in the context of the whole human genome and interactome. Such an approach requires an integrative modeling framework for drug action that leverages advances in data-driven statistical modeling and mechanism-based multiscale modeling and transforms heterogeneous data from GWAS, high-throughput sequencing, structural genomics, functional genomics, and chemical genomics into unified knowledge. This is not a small task, but, as reviewed here, progress is being made towards the final goal of personalized medicines for the treatment of complex diseases.
Collapse
Affiliation(s)
- Lei Xie
- Department of Computer Science, Hunter College, The City University of New York, New York, New York, United States of America
- Ph.D. Program in Computer Science, Biology, and Biochemistry, The Graduate Center, The City University of New York, New York, New York, United States of America
- * E-mail:
| | - Xiaoxia Ge
- Department of Computer Science, Hunter College, The City University of New York, New York, New York, United States of America
| | - Hepan Tan
- Department of Computer Science, Hunter College, The City University of New York, New York, New York, United States of America
| | - Li Xie
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Yinliang Zhang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Thomas Hart
- Department of Biological Sciences, Hunter College, The City University of New York, New York, New York, United States of America
| | - Xiaowei Yang
- School of Public Health, Hunter College, The City University of New York, New York, New York, United States of America
| | - Philip E. Bourne
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
35
|
Bolouri H. Modeling genomic regulatory networks with big data. Trends Genet 2014; 30:182-91. [PMID: 24630831 DOI: 10.1016/j.tig.2014.02.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 02/06/2023]
Abstract
High-throughput sequencing, large-scale data generation projects, and web-based cloud computing are changing how computational biology is performed, who performs it, and what biological insights it can deliver. I review here the latest developments in available data, methods, and software, focusing on the modeling and analysis of the gene regulatory interactions in cells. Three key findings are: (i) although sophisticated computational resources are increasingly available to bench biologists, tailored ongoing education is necessary to avoid the erroneous use of these resources. (ii) Current models of the regulation of gene expression are far too simplistic and need updating. (iii) Integrative computational analysis of large-scale datasets is becoming a fundamental component of molecular biology. I discuss current and near-term opportunities and challenges related to these three points.
Collapse
Affiliation(s)
- Hamid Bolouri
- Division of Human Biology, Fred Hutchinson Cancer Research Center (FHCRC), 1100 Fairview Avenue North, PO Box 19024, Seattle, WA 98109, USA.
| |
Collapse
|
36
|
Li J, Shan F, Xiong G, Wang JM, Wang WL, Xu X, Bai Y. Transcriptional regulation of miR-146b by C/EBPβ LAP2 in esophageal cancer cells. Biochem Biophys Res Commun 2014; 446:267-71. [PMID: 24589738 DOI: 10.1016/j.bbrc.2014.02.096] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 02/22/2014] [Indexed: 12/27/2022]
Abstract
Recent clinical study indicated that up-regulation of miR-146b was associated with poor overall survival of patients in esophageal squamous cell carcinoma. However, the underlying mechanism of miR-146b dysregulation remains to be explored. Here we report that miR-146b promotes cell proliferation and inhibits cell apoptosis in esophageal cancer cell lines. Mechanismly, two C/EBPβ binding motifs are located in the miR-146b promoter conserved region. Among the three isoforms of C/EBPβ, C/EBPβ LAP2 positively regulated miR-146b expression and increases miR-146b levels in a dose-dependent manner through transcription activation of miR-146b gene. Together, these results suggest a miR-146b regulatory mechanism involving C/EBPβ, which may contribute to the up-regulation of miR-146b in esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Junxia Li
- Department of Medical Genetics, Third Military Medical University, Chongqing, People's Republic of China
| | - Fabo Shan
- Department of Pathophysiology and High Altitude Physiology, Third Military Medical University, Chongqing, People's Republic of China
| | - Gang Xiong
- Department of Thoracic and Cardiac Surgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Ju-Ming Wang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Lin Wang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Xueqing Xu
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China.
| | - Yun Bai
- Department of Medical Genetics, Third Military Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
37
|
Abstract
Traditionally, scientific research has focused on studying individual events, such as single mutations, gene function, or the effect that mutating one protein has on a biological phenotype. A range of technologies is beginning to provide information that will enable a holistic view of how genomic and epigenetic aberrations in cancer cells can alter the homeostasis of signalling networks within these cells, between cancer cells and the local microenvironment, and at the organ and organism level. This process, termed Systems Biology, needs to be integrated with an iterative approach wherein hypotheses and predictions that arise from modelling are refined and constrained by experimental evaluation. Systems biology approaches will be vital for developing and implementing effective strategies to deliver personalized cancer therapy. Specifically, these approaches will be important to select those patients who are most likely to benefit from targeted therapies and for the development and implementation of rational combinatorial therapies. Systems biology can help to increase therapy efficacy or bypass the emergence of resistance, thus converting the current-often short term-effects of targeted therapies into durable responses, ultimately to improve patient quality of life and provide a cure.
Collapse
|
38
|
Stevens A, De Leonibus C, Hanson D, Dowsey AW, Whatmore A, Meyer S, Donn RP, Chatelain P, Banerjee I, Cosgrove KE, Clayton PE, Dunne MJ. Network analysis: a new approach to study endocrine disorders. J Mol Endocrinol 2014; 52:R79-93. [PMID: 24085748 DOI: 10.1530/jme-13-0112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Systems biology is the study of the interactions that occur between the components of individual cells - including genes, proteins, transcription factors, small molecules, and metabolites, and their relationships to complex physiological and pathological processes. The application of systems biology to medicine promises rapid advances in both our understanding of disease and the development of novel treatment options. Network biology has emerged as the primary tool for studying systems biology as it utilises the mathematical analysis of the relationships between connected objects in a biological system and allows the integration of varied 'omic' datasets (including genomics, metabolomics, proteomics, etc.). Analysis of network biology generates interactome models to infer and assess function; to understand mechanisms, and to prioritise candidates for further investigation. This review provides an overview of network methods used to support this research and an insight into current applications of network analysis applied to endocrinology. A wide spectrum of endocrine disorders are included ranging from congenital hyperinsulinism in infancy, through childhood developmental and growth disorders, to the development of metabolic diseases in early and late adulthood, such as obesity and obesity-related pathologies. In addition to providing a deeper understanding of diseases processes, network biology is also central to the development of personalised treatment strategies which will integrate pharmacogenomics with systems biology of the individual.
Collapse
Affiliation(s)
- A Stevens
- Faculty of Medical and Human Sciences, Institute of Human Development, University of Manchester, Manchester, UK Manchester Academic Health Science Centre, Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, 5th Floor, Oxford Road, Manchester M13 9WL, UK Paediatric and Adolescent Oncology, The University of Manchester, Manchester M13 9WL, UK Stem Cell and Leukaemia Proteomics Laboratory, School of Cancer and Imaging Sciences, The University of Manchester, Manchester M20 4BX, UK Musculoskeletal Research Group, NIHR BRU, University of Manchester, Manchester M13 9PT, UK Department Pediatrie, Hôpital Mère-Enfant, Université Claude Bernard, 69677 Lyon, France Faculty of Life Sciences, University of Manchester, Manchester M13 9NT, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
GITTER ANTHONY, BRAUNSTEIN ALFREDO, PAGNANI ANDREA, BALDASSI CARLO, BORGS CHRISTIAN, CHAYES JENNIFER, ZECCHINA RICCARDO, FRAENKEL ERNEST. Sharing information to reconstruct patient-specific pathways in heterogeneous diseases. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2014:39-50. [PMID: 24297532 PMCID: PMC3910098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Advances in experimental techniques resulted in abundant genomic, transcriptomic, epigenomic, and proteomic data that have the potential to reveal critical drivers of human diseases. Complementary algorithmic developments enable researchers to map these data onto protein-protein interaction networks and infer which signaling pathways are perturbed by a disease. Despite this progress, integrating data across different biological samples or patients remains a substantial challenge because samples from the same disease can be extremely heterogeneous. Somatic mutations in cancer are an infamous example of this heterogeneity. Although the same signaling pathways may be disrupted in a cancer patient cohort, the distribution of mutations is long-tailed, and many driver mutations may only be detected in a small fraction of patients. We developed a computational approach to account for heterogeneous data when inferring signaling pathways by sharing information across the samples. Our technique builds upon the prize-collecting Steiner forest problem, a network optimization algorithm that extracts pathways from a protein-protein interaction network. We recover signaling pathways that are similar across all samples yet still reflect the unique characteristics of each biological sample. Leveraging data from related tumors improves our ability to recover the disrupted pathways and reveals patient-specific pathway perturbations in breast cancer.
Collapse
Affiliation(s)
- ANTHONY GITTER
- Microsoft Research, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - ALFREDO BRAUNSTEIN
- DISAT and Center for Computational Sciences, Politecnico di Torino, Turin, Italy
- Human Genetics Foundation, Turin, Italy
| | - ANDREA PAGNANI
- DISAT and Center for Computational Sciences, Politecnico di Torino, Turin, Italy
- Human Genetics Foundation, Turin, Italy
| | - CARLO BALDASSI
- DISAT and Center for Computational Sciences, Politecnico di Torino, Turin, Italy
- Human Genetics Foundation, Turin, Italy
| | | | | | - RICCARDO ZECCHINA
- DISAT and Center for Computational Sciences, Politecnico di Torino, Turin, Italy
- Human Genetics Foundation, Turin, Italy
| | - ERNEST FRAENKEL
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
40
|
Dudley E, Bond AE. Phosphoproteomic Techniques and Applications. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 95:25-69. [DOI: 10.1016/b978-0-12-800453-1.00002-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
41
|
Patil A, Kumagai Y, Liang KC, Suzuki Y, Nakai K. Linking transcriptional changes over time in stimulated dendritic cells to identify gene networks activated during the innate immune response. PLoS Comput Biol 2013; 9:e1003323. [PMID: 24244133 PMCID: PMC3820512 DOI: 10.1371/journal.pcbi.1003323] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 09/21/2013] [Indexed: 01/09/2023] Open
Abstract
The innate immune response is primarily mediated by the Toll-like receptors functioning through the MyD88-dependent and TRIF-dependent pathways. Despite being widely studied, it is not yet completely understood and systems-level analyses have been lacking. In this study, we identified a high-probability network of genes activated during the innate immune response using a novel approach to analyze time-course gene expression profiles of activated immune cells in combination with a large gene regulatory and protein-protein interaction network. We classified the immune response into three consecutive time-dependent stages and identified the most probable paths between genes showing a significant change in expression at each stage. The resultant network contained several novel and known regulators of the innate immune response, many of which did not show any observable change in expression at the sampled time points. The response network shows the dominance of genes from specific functional classes during different stages of the immune response. It also suggests a role for the protein phosphatase 2a catalytic subunit α in the regulation of the immunoproteasome during the late phase of the response. In order to clarify the differences between the MyD88-dependent and TRIF-dependent pathways in the innate immune response, time-course gene expression profiles from MyD88-knockout and TRIF-knockout dendritic cells were analyzed. Their response networks suggest the dominance of the MyD88-dependent pathway in the innate immune response, and an association of the circadian regulators and immunoproteasomal degradation with the TRIF-dependent pathway. The response network presented here provides the most probable associations between genes expressed in the early and the late phases of the innate immune response, while taking into account the intermediate regulators. We propose that the method described here can also be used in the identification of time-dependent gene sub-networks in other biological systems.
Collapse
Affiliation(s)
- Ashwini Patil
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yutaro Kumagai
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kuo-ching Liang
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, The University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
42
|
Wilson JL, Hemann MT, Fraenkel E, Lauffenburger DA. Integrated network analyses for functional genomic studies in cancer. Semin Cancer Biol 2013; 23:213-8. [PMID: 23811269 DOI: 10.1016/j.semcancer.2013.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/11/2013] [Accepted: 06/13/2013] [Indexed: 11/24/2022]
Abstract
RNA-interference (RNAi) studies hold great promise for functional investigation of the significance of genetic variations and mutations, as well as potential synthetic lethalities, for understanding and treatment of cancer, yet technical and conceptual issues currently diminish the potential power of this approach. While numerous research groups are usefully employing this kind of functional genomic methodology to identify molecular mediators of disease severity, response, and resistance to treatment, findings are generally confounded by "off-target" effects. These effects arise from a variety of issues beyond non-specific reagent behavior, such as biological cross-talk and feedback processes so thus can occur even with specific perturbation. Interpreting RNAi results in a network framework instead of merely as individual "hits" or "targets" leverages contributions from all hit/target contributions to pathways via their relationships with other network nodes. This interpretation can ameliorate dependence upon individual reagent performance and increase confidence in biological validation. Here we provide background on RNAi studies in cancer applications, review key challenges with functional genomics, and motivate the use of network models grounded in pathway analyses.
Collapse
Affiliation(s)
- Jennifer L Wilson
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, USA.
| | | | | | | |
Collapse
|
43
|
Dattolo E, Gu J, Bayer PE, Mazzuca S, Serra IA, Spadafora A, Bernardo L, Natali L, Cavallini A, Procaccini G. Acclimation to different depths by the marine angiosperm Posidonia oceanica: transcriptomic and proteomic profiles. FRONTIERS IN PLANT SCIENCE 2013; 4:195. [PMID: 23785376 PMCID: PMC3683636 DOI: 10.3389/fpls.2013.00195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/27/2013] [Indexed: 05/11/2023]
Abstract
For seagrasses, seasonal and daily variations in light and temperature represent the mains factors driving their distribution along the bathymetric cline. Changes in these environmental factors, due to climatic and anthropogenic effects, can compromise their survival. In a framework of conservation and restoration, it becomes crucial to improve our knowledge about the physiological plasticity of seagrass species along environmental gradients. Here, we aimed to identify differences in transcriptomic and proteomic profiles, involved in the acclimation along the depth gradient in the seagrass Posidonia oceanica, and to improve the available molecular resources in this species, which is an important requisite for the application of eco-genomic approaches. To do that, from plant growing in shallow (-5 m) and deep (-25 m) portions of a single meadow, (i) we generated two reciprocal Expressed Sequences Tags (EST) libraries using a Suppressive Subtractive Hybridization (SSH) approach, to obtain depth/specific transcriptional profiles, and (ii) we identified proteins differentially expressed, using the highly innovative USIS mass spectrometry methodology, coupled with 1D-SDS electrophoresis and labeling free approach. Mass spectra were searched in the open source Global Proteome Machine (GPM) engine against plant databases and with the X!Tandem algorithm against a local database. Transcriptional analysis showed both quantitative and qualitative differences between depths. EST libraries had only the 3% of transcripts in common. A total of 315 peptides belonging to 64 proteins were identified by mass spectrometry. ATP synthase subunits were among the most abundant proteins in both conditions. Both approaches identified genes and proteins in pathways related to energy metabolism, transport and genetic information processing, that appear to be the most involved in depth acclimation in P. oceanica. Their putative rules in acclimation to depth were discussed.
Collapse
Affiliation(s)
- Emanuela Dattolo
- Functional and Evolutionary Ecology Lab, Stazione Zoologica Anton DohrnNapoli, Italy
| | - Jenny Gu
- Evolutionary Bioinformatics Group, Institute for Evolution and Biodiversity, University of MünsterMünster, Germany
| | - Philipp E. Bayer
- Evolutionary Bioinformatics Group, Institute for Evolution and Biodiversity, University of MünsterMünster, Germany
| | - Silvia Mazzuca
- Laboratorio di Proteomica, Dipartimento di Chimica e Tecnologie Chimiche, Università della CalabriaArcavacata di Rende (CS), Italy
- *Correspondence: Silvia Mazzuca, Associate Professor in Plant Biology, Laboratorio di Proteomica, Dipartimento di Chimica e Tecnologie Chimiche, Università della Calabria, Ponte Bucci, 12 A, 87036 Arcavacata di Rende (CS), Italy e-mail:
| | - Ilia A. Serra
- Laboratorio di Proteomica, Dipartimento di Chimica e Tecnologie Chimiche, Università della CalabriaArcavacata di Rende (CS), Italy
| | - Antonia Spadafora
- Laboratorio di Proteomica, Dipartimento di Chimica e Tecnologie Chimiche, Università della CalabriaArcavacata di Rende (CS), Italy
| | - Letizia Bernardo
- Laboratorio di Proteomica, Dipartimento di Chimica e Tecnologie Chimiche, Università della CalabriaArcavacata di Rende (CS), Italy
| | - Lucia Natali
- Dipartimento di Scienze Agrarie, Alimentari ed Agro-ambientali, Università di PisaPisa, Italy
| | - Andrea Cavallini
- Dipartimento di Scienze Agrarie, Alimentari ed Agro-ambientali, Università di PisaPisa, Italy
| | - Gabriele Procaccini
- Functional and Evolutionary Ecology Lab, Stazione Zoologica Anton DohrnNapoli, Italy
| |
Collapse
|