1
|
van Gerwen J, Masson SWC, Cutler HB, Vegas AD, Potter M, Stöckli J, Madsen S, Nelson ME, Humphrey SJ, James DE. The genetic and dietary landscape of the muscle insulin signalling network. eLife 2024; 12:RP89212. [PMID: 38329473 PMCID: PMC10942587 DOI: 10.7554/elife.89212] [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] [Indexed: 02/09/2024] Open
Abstract
Metabolic disease is caused by a combination of genetic and environmental factors, yet few studies have examined how these factors influence signal transduction, a key mediator of metabolism. Using mass spectrometry-based phosphoproteomics, we quantified 23,126 phosphosites in skeletal muscle of five genetically distinct mouse strains in two dietary environments, with and without acute in vivo insulin stimulation. Almost half of the insulin-regulated phosphoproteome was modified by genetic background on an ordinary diet, and high-fat high-sugar feeding affected insulin signalling in a strain-dependent manner. Our data revealed coregulated subnetworks within the insulin signalling pathway, expanding our understanding of the pathway's organisation. Furthermore, associating diverse signalling responses with insulin-stimulated glucose uptake uncovered regulators of muscle insulin responsiveness, including the regulatory phosphosite S469 on Pfkfb2, a key activator of glycolysis. Finally, we confirmed the role of glycolysis in modulating insulin action in insulin resistance. Our results underscore the significance of genetics in shaping global signalling responses and their adaptability to environmental changes, emphasising the utility of studying biological diversity with phosphoproteomics to discover key regulatory mechanisms of complex traits.
Collapse
Affiliation(s)
- Julian van Gerwen
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Stewart WC Masson
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Harry B Cutler
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Alexis Diaz Vegas
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Meg Potter
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Jacqueline Stöckli
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Søren Madsen
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Marin E Nelson
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
- Faculty of Medicine and Health, University of SydneySydneyAustralia
| |
Collapse
|
2
|
Pernice S, Maglione A, Tortarolo D, Sirovich R, Clerico M, Rolla S, Beccuti M, Cordero F. A new computational workflow to guide personalized drug therapy. J Biomed Inform 2023; 148:104546. [PMID: 37984546 DOI: 10.1016/j.jbi.2023.104546] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVE Computational models are at the forefront of the pursuit of personalized medicine thanks to their descriptive and predictive abilities. In the presence of complex and heterogeneous data, patient stratification is a prerequisite for effective precision medicine, since disease development is often driven by individual variability and unpredictable environmental events. Herein, we present GreatNectorworkflow as a valuable tool for (i) the analysis and clustering of patient-derived longitudinal data, and (ii) the simulation of the resulting model of patient-specific disease dynamics. METHODS GreatNectoris designed by combining an analytic strategy composed of CONNECTOR, a data-driven framework for the inspection of longitudinal data, and an unsupervised methodology to stratify the subjects with GreatMod, a quantitative modeling framework based on the Petri Net formalism and its generalizations. RESULTS To illustrate GreatNectorcapabilities, we exploited longitudinal data of four immune cell populations collected from Multiple Sclerosis patients. Our main results report that the T-cell dynamics after alemtuzumab treatment separate non-responders versus responders patients, and the patients in the non-responders group are characterized by an increase of the Th17 concentration around 36 months. CONCLUSION GreatNectoranalysis was able to stratify individual patients into three model meta-patients whose dynamics suggested insight into patient-tailored interventions.
Collapse
Affiliation(s)
- Simone Pernice
- Department of Computer Science, University of Turin, Corso Svizzera 185, Turin, 10149, Italy; CINI Infolife laboratory, Turin, Italy
| | - Alessandro Maglione
- Department of Clinical and Biological Sciences, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Dora Tortarolo
- Department of Computer Science, University of Turin, Corso Svizzera 185, Turin, 10149, Italy
| | - Roberta Sirovich
- Department of Mathematics, University of Turin, Via Carlo Alberto 10, Turin, 10123, Italy
| | - Marinella Clerico
- Department of Clinical and Biological Sciences, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Simona Rolla
- Department of Clinical and Biological Sciences, Regione Gonzole 10, Orbassano, 10043, Italy.
| | - Marco Beccuti
- Department of Computer Science, University of Turin, Corso Svizzera 185, Turin, 10149, Italy; CINI Infolife laboratory, Turin, Italy
| | - Francesca Cordero
- Department of Computer Science, University of Turin, Corso Svizzera 185, Turin, 10149, Italy; CINI Infolife laboratory, Turin, Italy
| |
Collapse
|
3
|
Flanary VL, Fisher JL, Wilk EJ, Howton TC, Lasseigne BN. Computational Advancements in Cancer Combination Therapy Prediction. JCO Precis Oncol 2023; 7:e2300261. [PMID: 37824797 DOI: 10.1200/po.23.00261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/20/2023] [Accepted: 08/15/2023] [Indexed: 10/14/2023] Open
Abstract
Given the high attrition rate of de novo drug discovery and limited efficacy of single-agent therapies in cancer treatment, combination therapy prediction through in silico drug repurposing has risen as a time- and cost-effective alternative for identifying novel and potentially efficacious therapies for cancer. The purpose of this review is to provide an introduction to computational methods for cancer combination therapy prediction and to summarize recent studies that implement each of these methods. A systematic search of the PubMed database was performed, focusing on studies published within the past 10 years. Our search included reviews and articles of ongoing and retrospective studies. We prioritized articles with findings that suggest considerations for improving combination therapy prediction methods over providing a meta-analysis of all currently available cancer combination therapy prediction methods. Computational methods used for drug combination therapy prediction in cancer research include networks, regression-based machine learning, classifier machine learning models, and deep learning approaches. Each method class has its own advantages and disadvantages, so careful consideration is needed to determine the most suitable class when designing a combination therapy prediction method. Future directions to improve current combination therapy prediction technology include incorporation of disease pathobiology, drug characteristics, patient multiomics data, and drug-drug interactions to determine maximally efficacious and tolerable drug regimens for cancer. As computational methods improve in their capability to integrate patient, drug, and disease data, more comprehensive models can be developed to more accurately predict safe and efficacious combination drug therapies for cancer and other complex diseases.
Collapse
Affiliation(s)
- Victoria L Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer L Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Elizabeth J Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Timothy C Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| | - Brittany N Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
4
|
Lövfors W, Magnusson R, Jönsson C, Gustafsson M, Olofsson CS, Cedersund G, Nyman E. A comprehensive mechanistic model of adipocyte signaling with layers of confidence. NPJ Syst Biol Appl 2023; 9:24. [PMID: 37286693 DOI: 10.1038/s41540-023-00282-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/17/2023] [Indexed: 06/09/2023] Open
Abstract
Adipocyte signaling, normally and in type 2 diabetes, is far from fully understood. We have earlier developed detailed dynamic mathematical models for several well-studied, partially overlapping, signaling pathways in adipocytes. Still, these models only cover a fraction of the total cellular response. For a broader coverage of the response, large-scale phosphoproteomic data and systems level knowledge on protein interactions are key. However, methods to combine detailed dynamic models with large-scale data, using information about the confidence of included interactions, are lacking. We have developed a method to first establish a core model by connecting existing models of adipocyte cellular signaling for: (1) lipolysis and fatty acid release, (2) glucose uptake, and (3) the release of adiponectin. Next, we use publicly available phosphoproteome data for the insulin response in adipocytes together with prior knowledge on protein interactions, to identify phosphosites downstream of the core model. In a parallel pairwise approach with low computation time, we test whether identified phosphosites can be added to the model. We iteratively collect accepted additions into layers and continue the search for phosphosites downstream of these added layers. For the first 30 layers with the highest confidence (311 added phosphosites), the model predicts independent data well (70-90% correct), and the predictive capability gradually decreases when we add layers of decreasing confidence. In total, 57 layers (3059 phosphosites) can be added to the model with predictive ability kept. Finally, our large-scale, layered model enables dynamic simulations of systems-wide alterations in adipocytes in type 2 diabetes.
Collapse
Affiliation(s)
- William Lövfors
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden.
- Department of Mathematics, Linköping University, Linköping, Sweden.
- School of Medical Sciences and Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Rasmus Magnusson
- School of Bioscience, Systems Biology Research Center, University of Skövde, Skövde, Sweden
| | - Cecilia Jönsson
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Mika Gustafsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Charlotta S Olofsson
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Gunnar Cedersund
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden.
- School of Medical Sciences and Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.
| | - Elin Nyman
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden.
| |
Collapse
|
5
|
Hemedan AA, Schneider R, Ostaszewski M. Applications of Boolean modeling to study the dynamics of a complex disease and therapeutics responses. FRONTIERS IN BIOINFORMATICS 2023; 3:1189723. [PMID: 37325771 PMCID: PMC10267406 DOI: 10.3389/fbinf.2023.1189723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023] Open
Abstract
Computational modeling has emerged as a critical tool in investigating the complex molecular processes involved in biological systems and diseases. In this study, we apply Boolean modeling to uncover the molecular mechanisms underlying Parkinson's disease (PD), one of the most prevalent neurodegenerative disorders. Our approach is based on the PD-map, a comprehensive molecular interaction diagram that captures the key mechanisms involved in the initiation and progression of PD. Using Boolean modeling, we aim to gain a deeper understanding of the disease dynamics, identify potential drug targets, and simulate the response to treatments. Our analysis demonstrates the effectiveness of this approach in uncovering the intricacies of PD. Our results confirm existing knowledge about the disease and provide valuable insights into the underlying mechanisms, ultimately suggesting potential targets for therapeutic intervention. Moreover, our approach allows us to parametrize the models based on omics data for further disease stratification. Our study highlights the value of computational modeling in advancing our understanding of complex biological systems and diseases, emphasizing the importance of continued research in this field. Furthermore, our findings have potential implications for the development of novel therapies for PD, which is a pressing public health concern. Overall, this study represents a significant step forward in the application of computational modeling to the investigation of neurodegenerative diseases, and underscores the power of interdisciplinary approaches in tackling challenging biomedical problems.
Collapse
|
6
|
Utharala R, Grab A, Vafaizadeh V, Peschke N, Ballinger M, Turei D, Tuechler N, Ma W, Ivanova O, Ortiz AG, Saez-Rodriguez J, Merten CA. A microfluidic Braille valve platform for on-demand production, combinatorial screening and sorting of chemically distinct droplets. Nat Protoc 2022; 17:2920-2965. [PMID: 36261631 DOI: 10.1038/s41596-022-00740-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022]
Abstract
Droplet microfluidics is a powerful tool for a variety of biological applications including single-cell genetics, antibody discovery and directed evolution. All these applications make use of genetic libraries, illustrating the difficulty of generating chemically distinct droplets for screening applications. This protocol describes our Braille Display valving platform for on-demand generation of droplets with different chemical contents (16 different reagents and combinations thereof), as well as sorting droplets with different chemical properties, on the basis of fluorescence signals. The Braille Display platform is compact, versatile and cost efficient (only ~US$1,000 on top of a standard droplet microfluidics setup). The procedure includes manufacturing of microfluidic chips, assembly of custom hardware, co-encapsulation of cells and drugs into droplets, fluorescence detection of readout signals and data analysis using shared, freely available LabVIEW and Python packages. As a first application, we demonstrate the complete workflow for screening cancer cell drug sensitivities toward 74 conditions. Furthermore, we describe here an assay enabling the normalization of the observed drug sensitivity to the number of cancer cells per droplet, which additionally increases the robustness of the system. As a second application, we also demonstrate the sorting of droplets according to enzymatic activity. The drug screening application can be completed within 2 d; droplet sorting takes ~1 d; and all preparatory steps for manufacturing molds, chips and setting up the Braille controller can be accomplished within 1 week.
Collapse
Affiliation(s)
- Ramesh Utharala
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Anna Grab
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, DKFZ Heidelberg and Translational Myeloma Research Group, Department of Internal Medicine V, University Hospital, Heidelberg, Germany
| | - Vida Vafaizadeh
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nicolas Peschke
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Martine Ballinger
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Denes Turei
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | - Nadine Tuechler
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Wenwei Ma
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Olga Ivanova
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
| | | | - Julio Saez-Rodriguez
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg University, Heidelberg, Germany
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Aachen, Germany
| | - Christoph A Merten
- Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
7
|
Kim M, Kim E. Mathematical model of the cell signaling pathway based on the extended Boolean network model with a stochastic process. BMC Bioinformatics 2022; 23:515. [PMID: 36451112 PMCID: PMC9710037 DOI: 10.1186/s12859-022-05077-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND In cell signaling pathways, proteins interact with each other to determine cell fate in response to either cell-extrinsic (micro-environmental) or intrinsic cues. One of the well-studied pathways, the mitogen-activated protein kinase (MAPK) signaling pathway, regulates cell processes such as differentiation, proliferation, apoptosis, and survival in response to various micro-environmental stimuli in eukaryotes. Upon micro-environmental stimulus, receptors on the cell membrane become activated. Activated receptors initiate a cascade of protein activation in the MAPK pathway. This activation involves protein binding, creating scaffold proteins, which are known to facilitate effective MAPK signaling transduction. RESULTS This paper presents a novel mathematical model of a cell signaling pathway coordinated by protein scaffolding. The model is based on the extended Boolean network approach with stochastic processes. Protein production or decay in a cell was modeled considering the stochastic process, whereas the protein-protein interactions were modeled based on the extended Boolean network approach. Our model fills a gap in the binary set applied to previous models. The model simultaneously considers the stochastic process directly. Using the model, we simulated a simplified mitogen-activated protein kinase (MAPK) signaling pathway upon stimulation of both a single receptor at the initial time and multiple receptors at several time points. Our simulations showed that the signal is amplified as it travels down to the pathway from the receptor, generating substantially amplified downstream ERK activity. The noise generated by the stochastic process of protein self-activity in the model was also amplified as the signaling propagated through the pathway. CONCLUSIONS The signaling transduction in a simplified MAPK signaling pathway could be explained by a mathematical model based on the extended Boolean network model with a stochastic process. The model simulations demonstrated signaling amplifications when it travels downstream, which was already observed in experimental settings. We also highlight the importance of stochastic activity in regulating protein inactivation.
Collapse
Affiliation(s)
- Minsoo Kim
- grid.35541.360000000121053345Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, South Korea
| | - Eunjung Kim
- grid.35541.360000000121053345Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, South Korea
| |
Collapse
|
8
|
Martins dos Santos V, Anton M, Szomolay B, Ostaszewski M, Arts I, Benfeitas R, Dominguez Del Angel V, Domínguez-Romero E, Ferk P, Fey D, Goble C, Golebiewski M, Gruden K, Heil KF, Hermjakob H, Kahlem P, Klapa MI, Koehorst J, Kolodkin A, Kutmon M, Leskošek B, Moretti S, Müller W, Pagni M, Rezen T, Rocha M, Rozman D, Šafránek D, T. Scott W, Sheriff RSM, Suarez Diez M, Van Steen K, Westerhoff HV, Wittig U, Wolstencroft K, Zupanic A, Evelo CT, Hancock JM. Systems Biology in ELIXIR: modelling in the spotlight. F1000Res 2022; 11:ELIXIR-1265. [PMID: 36742342 PMCID: PMC9871403 DOI: 10.12688/f1000research.126734.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
In this white paper, we describe the founding of a new ELIXIR Community - the Systems Biology Community - and its proposed future contributions to both ELIXIR and the broader community of systems biologists in Europe and worldwide. The Community believes that the infrastructure aspects of systems biology - databases, (modelling) tools and standards development, as well as training and access to cloud infrastructure - are not only appropriate components of the ELIXIR infrastructure, but will prove key components of ELIXIR's future support of advanced biological applications and personalised medicine. By way of a series of meetings, the Community identified seven key areas for its future activities, reflecting both future needs and previous and current activities within ELIXIR Platforms and Communities. These are: overcoming barriers to the wider uptake of systems biology; linking new and existing data to systems biology models; interoperability of systems biology resources; further development and embedding of systems medicine; provisioning of modelling as a service; building and coordinating capacity building and training resources; and supporting industrial embedding of systems biology. A set of objectives for the Community has been identified under four main headline areas: Standardisation and Interoperability, Technology, Capacity Building and Training, and Industrial Embedding. These are grouped into short-term (3-year), mid-term (6-year) and long-term (10-year) objectives.
Collapse
Affiliation(s)
- Vitor Martins dos Santos
- Laboratory of Bioprocess Engineering, Wageningen University & Research, Wageningen, 6708 PB, The Netherlands
| | - Mihail Anton
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, SE-41258, Sweden
| | - Barbara Szomolay
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, L-4367, Luxembourg
| | - Ilja Arts
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | | | | | - Polonca Ferk
- Faculty of Medicine, Institute for Biostatistics and Medical Informatics, Centre ELIXIR-SI, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | - Dirk Fey
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, 4, Ireland
| | - Carole Goble
- Department of Computer Science, The University of Manchester, Manchester, M13 9PL, UK
| | - Martin Golebiewski
- Heidelberg Institute for Theoretical Studies - HITS, Heidelberg, 69118, Germany
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, SI-1000, Slovenia
| | | | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, UK
| | - Pascal Kahlem
- Scientific Network Management SL, Barcelona, 08015, Spain
| | - Maria I. Klapa
- Metabolic Engineering & Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research & Technology - Hellas (FORTH/ICE-HT), Patras, 26504, Greece
| | - Jasper Koehorst
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, 6708WE, The Netherlands
| | - Alexey Kolodkin
- Competence Center for Methodology and Statistics; Transversal Translational Medicine, Translational Medicine Operations Hub, Luxembourg Institute of Health, Strassen, L-1445, Luxembourg
- ISBE.NL, VU University of Amsterdam, Amsterdam, The Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6200 MD, The Netherlands
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Brane Leskošek
- Faculty of Medicine, Institute for Biostatistics and Medical Informatics, Centre ELIXIR-SI, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | | | - Wolfgang Müller
- Heidelberg Institute for Theoretical Studies - HITS, Heidelberg, 69118, Germany
| | - Marco Pagni
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Tadeja Rezen
- Faculty of Medicine, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | - Miguel Rocha
- Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Damjana Rozman
- Faculty of Medicine, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | - David Šafránek
- Faculty of Informatics, Masaryk University, Brno, 602 00, Czech Republic
| | - William T. Scott
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, 6708WE, The Netherlands
- UNLOCK, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| | - Rahuman S. Malik Sheriff
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, UK
| | - Maria Suarez Diez
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, 6708WE, The Netherlands
| | - Kristel Van Steen
- BIO3 - Laboratory for Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, 3000, Belgium
- BIO3 - Systems Genetics, GIGA-R Medical Genomics, University of Liege, Liege, 4000, Belgium
| | | | - Ulrike Wittig
- Heidelberg Institute for Theoretical Studies - HITS, Heidelberg, 69118, Germany
| | - Katherine Wolstencroft
- Leiden Institute of Advanced Computer Science, Leiden University, Leiden, 2333 CA, The Netherlands
| | - Anze Zupanic
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, SI-1000, Slovenia
| | - Chris T. Evelo
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - John M. Hancock
- Faculty of Medicine, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| |
Collapse
|
9
|
Martins dos Santos V, Anton M, Szomolay B, Ostaszewski M, Arts I, Benfeitas R, Dominguez Del Angel V, Domínguez-Romero E, Ferk P, Fey D, Goble C, Golebiewski M, Gruden K, Heil KF, Hermjakob H, Kahlem P, Klapa MI, Koehorst J, Kolodkin A, Kutmon M, Leskošek B, Moretti S, Müller W, Pagni M, Rezen T, Rocha M, Rozman D, Šafránek D, T. Scott W, Sheriff RSM, Suarez Diez M, Van Steen K, Westerhoff HV, Wittig U, Wolstencroft K, Zupanic A, Evelo CT, Hancock JM. Systems Biology in ELIXIR: modelling in the spotlight. F1000Res 2022; 11:ELIXIR-1265. [PMID: 36742342 PMCID: PMC9871403 DOI: 10.12688/f1000research.126734.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
In this white paper, we describe the founding of a new ELIXIR Community - the Systems Biology Community - and its proposed future contributions to both ELIXIR and the broader community of systems biologists in Europe and worldwide. The Community believes that the infrastructure aspects of systems biology - databases, (modelling) tools and standards development, as well as training and access to cloud infrastructure - are not only appropriate components of the ELIXIR infrastructure, but will prove key components of ELIXIR's future support of advanced biological applications and personalised medicine. By way of a series of meetings, the Community identified seven key areas for its future activities, reflecting both future needs and previous and current activities within ELIXIR Platforms and Communities. These are: overcoming barriers to the wider uptake of systems biology; linking new and existing data to systems biology models; interoperability of systems biology resources; further development and embedding of systems medicine; provisioning of modelling as a service; building and coordinating capacity building and training resources; and supporting industrial embedding of systems biology. A set of objectives for the Community has been identified under four main headline areas: Standardisation and Interoperability, Technology, Capacity Building and Training, and Industrial Embedding. These are grouped into short-term (3-year), mid-term (6-year) and long-term (10-year) objectives.
Collapse
Affiliation(s)
- Vitor Martins dos Santos
- Laboratory of Bioprocess Engineering, Wageningen University & Research, Wageningen, 6708 PB, The Netherlands
| | - Mihail Anton
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, SE-41258, Sweden
| | - Barbara Szomolay
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, L-4367, Luxembourg
| | - Ilja Arts
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Rui Benfeitas
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | | | | | - Polonca Ferk
- Faculty of Medicine, Institute for Biostatistics and Medical Informatics, Centre ELIXIR-SI, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | - Dirk Fey
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, 4, Ireland
| | - Carole Goble
- Department of Computer Science, The University of Manchester, Manchester, M13 9PL, UK
| | - Martin Golebiewski
- Heidelberg Institute for Theoretical Studies - HITS, Heidelberg, 69118, Germany
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, SI-1000, Slovenia
| | | | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, UK
| | - Pascal Kahlem
- Scientific Network Management SL, Barcelona, 08015, Spain
| | - Maria I. Klapa
- Metabolic Engineering & Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research & Technology - Hellas (FORTH/ICE-HT), Patras, 26504, Greece
| | - Jasper Koehorst
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, 6708WE, The Netherlands
| | - Alexey Kolodkin
- Competence Center for Methodology and Statistics; Transversal Translational Medicine, Translational Medicine Operations Hub, Luxembourg Institute of Health, Strassen, L-1445, Luxembourg
- ISBE.NL, VU University of Amsterdam, Amsterdam, The Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6200 MD, The Netherlands
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Brane Leskošek
- Faculty of Medicine, Institute for Biostatistics and Medical Informatics, Centre ELIXIR-SI, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | | | - Wolfgang Müller
- Heidelberg Institute for Theoretical Studies - HITS, Heidelberg, 69118, Germany
| | - Marco Pagni
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Tadeja Rezen
- Faculty of Medicine, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | - Miguel Rocha
- Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Damjana Rozman
- Faculty of Medicine, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| | - David Šafránek
- Faculty of Informatics, Masaryk University, Brno, 602 00, Czech Republic
| | - William T. Scott
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, 6708WE, The Netherlands
- UNLOCK, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| | - Rahuman S. Malik Sheriff
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, CB10 1SD, UK
| | - Maria Suarez Diez
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Wageningen, 6708WE, The Netherlands
| | - Kristel Van Steen
- BIO3 - Laboratory for Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, 3000, Belgium
- BIO3 - Systems Genetics, GIGA-R Medical Genomics, University of Liege, Liege, 4000, Belgium
| | | | - Ulrike Wittig
- Heidelberg Institute for Theoretical Studies - HITS, Heidelberg, 69118, Germany
| | - Katherine Wolstencroft
- Leiden Institute of Advanced Computer Science, Leiden University, Leiden, 2333 CA, The Netherlands
| | - Anze Zupanic
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, SI-1000, Slovenia
| | - Chris T. Evelo
- Department of Bioinformatics - BiGCaT, NUTRIM, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - John M. Hancock
- Faculty of Medicine, University of Ljubljana, Ljubljana, SI-1000, Slovenia
| |
Collapse
|
10
|
Hassan M, Awan FM, Naz A, deAndrés-Galiana EJ, Alvarez O, Cernea A, Fernández-Brillet L, Fernández-Martínez JL, Kloczkowski A. Innovations in Genomics and Big Data Analytics for Personalized Medicine and Health Care: A Review. Int J Mol Sci 2022; 23:4645. [PMID: 35563034 PMCID: PMC9104788 DOI: 10.3390/ijms23094645] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Big data in health care is a fast-growing field and a new paradigm that is transforming case-based studies to large-scale, data-driven research. As big data is dependent on the advancement of new data standards, technology, and relevant research, the future development of big data applications holds foreseeable promise in the modern day health care revolution. Enormously large, rapidly growing collections of biomedical omics-data (genomics, proteomics, transcriptomics, metabolomics, glycomics, etc.) and clinical data create major challenges and opportunities for their analysis and interpretation and open new computational gateways to address these issues. The design of new robust algorithms that are most suitable to properly analyze this big data by taking into account individual variability in genes has enabled the creation of precision (personalized) medicine. We reviewed and highlighted the significance of big data analytics for personalized medicine and health care by focusing mostly on machine learning perspectives on personalized medicine, genomic data models with respect to personalized medicine, the application of data mining algorithms for personalized medicine as well as the challenges we are facing right now in big data analytics.
Collapse
Affiliation(s)
- Mubashir Hassan
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore (UOL), Lahore 54590, Pakistan;
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Faryal Mehwish Awan
- Department of Medical Lab Technology, The University of Haripur, Haripur 22620, Pakistan;
| | - Anam Naz
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore (UOL), Lahore 54590, Pakistan;
| | - Enrique J. deAndrés-Galiana
- Group of Inverse Problems, Optimization and Machine Learning, University of Oviedo, 33003 Oviedo, Spain; (E.J.d.-G.); (J.L.F.-M.)
| | - Oscar Alvarez
- DeepBioInsights, 38311 La Florida, Spain; (O.A.); (A.C.); (L.F.-B.)
| | - Ana Cernea
- DeepBioInsights, 38311 La Florida, Spain; (O.A.); (A.C.); (L.F.-B.)
| | | | - Juan Luis Fernández-Martínez
- Group of Inverse Problems, Optimization and Machine Learning, University of Oviedo, 33003 Oviedo, Spain; (E.J.d.-G.); (J.L.F.-M.)
| | - Andrzej Kloczkowski
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| |
Collapse
|
11
|
Montagud A, Béal J, Tobalina L, Traynard P, Subramanian V, Szalai B, Alföldi R, Puskás L, Valencia A, Barillot E, Saez-Rodriguez J, Calzone L. Patient-specific Boolean models of signalling networks guide personalised treatments. eLife 2022; 11:e72626. [PMID: 35164900 PMCID: PMC9018074 DOI: 10.7554/elife.72626] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/27/2022] [Indexed: 11/22/2022] Open
Abstract
Prostate cancer is the second most occurring cancer in men worldwide. To better understand the mechanisms of tumorigenesis and possible treatment responses, we developed a mathematical model of prostate cancer which considers the major signalling pathways known to be deregulated. We personalised this Boolean model to molecular data to reflect the heterogeneity and specific response to perturbations of cancer patients. A total of 488 prostate samples were used to build patient-specific models and compared to available clinical data. Additionally, eight prostate cell line-specific models were built to validate our approach with dose-response data of several drugs. The effects of single and combined drugs were tested in these models under different growth conditions. We identified 15 actionable points of interventions in one cell line-specific model whose inactivation hinders tumorigenesis. To validate these results, we tested nine small molecule inhibitors of five of those putative targets and found a dose-dependent effect on four of them, notably those targeting HSP90 and PI3K. These results highlight the predictive power of our personalised Boolean models and illustrate how they can be used for precision oncology.
Collapse
Affiliation(s)
- Arnau Montagud
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3BarcelonaSpain
| | - Jonas Béal
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Luis Tobalina
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
| | - Pauline Traynard
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Vigneshwari Subramanian
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
| | - Bence Szalai
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
- Semmelweis University, Faculty of Medicine, Department of PhysiologyBudapestHungary
| | | | | | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3BarcelonaSpain
- ICREA, Pg. Lluís Companys 23BarcelonaSpain
| | - Emmanuel Barillot
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg UniversityHeidelbergGermany
| | - Laurence Calzone
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| |
Collapse
|
12
|
Zhang T, Androulakis IP, Bonate P, Cheng L, Helikar T, Parikh J, Rackauckas C, Subramanian K, Cho CR. Two heads are better than one: current landscape of integrating QSP and machine learning : An ISoP QSP SIG white paper by the working group on the integration of quantitative systems pharmacology and machine learning. J Pharmacokinet Pharmacodyn 2022; 49:5-18. [PMID: 35103884 PMCID: PMC8837505 DOI: 10.1007/s10928-022-09805-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/10/2022] [Indexed: 12/02/2022]
Abstract
Quantitative systems pharmacology (QSP) modeling is applied to address essential questions in drug development, such as the mechanism of action of a therapeutic agent and the progression of disease. Meanwhile, machine learning (ML) approaches also contribute to answering these questions via the analysis of multi-layer 'omics' data such as gene expression, proteomics, metabolomics, and high-throughput imaging. Furthermore, ML approaches can also be applied to aspects of QSP modeling. Both approaches are powerful tools and there is considerable interest in integrating QSP modeling and ML. So far, a few successful implementations have been carried out from which we have learned about how each approach can overcome unique limitations of the other. The QSP + ML working group of the International Society of Pharmacometrics QSP Special Interest Group was convened in September, 2019 to identify and begin realizing new opportunities in QSP and ML integration. The working group, which comprises 21 members representing 18 academic and industry organizations, has identified four categories of current research activity which will be described herein together with case studies of applications to drug development decision making. The working group also concluded that the integration of QSP and ML is still in its early stages of moving from evaluating available technical tools to building case studies. This paper reports on this fast-moving field and serves as a foundation for future codification of best practices.
Collapse
Affiliation(s)
- Tongli Zhang
- University of Cincinnati, Cincinnati, OH, 45267, USA.
| | | | | | | | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | - Christopher Rackauckas
- Pumas-AI, Baltimore, MD, USA
- Department of Mathematics, Massachusetts Institute of Technology, Boston, MA, USA
| | | | | |
Collapse
|
13
|
Rocca A, Kholodenko BN. Can Systems Biology Advance Clinical Precision Oncology? Cancers (Basel) 2021; 13:6312. [PMID: 34944932 PMCID: PMC8699328 DOI: 10.3390/cancers13246312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022] Open
Abstract
Precision oncology is perceived as a way forward to treat individual cancer patients. However, knowing particular cancer mutations is not enough for optimal therapeutic treatment, because cancer genotype-phenotype relationships are nonlinear and dynamic. Systems biology studies the biological processes at the systems' level, using an array of techniques, ranging from statistical methods to network reconstruction and analysis, to mathematical modeling. Its goal is to reconstruct the complex and often counterintuitive dynamic behavior of biological systems and quantitatively predict their responses to environmental perturbations. In this paper, we review the impact of systems biology on precision oncology. We show examples of how the analysis of signal transduction networks allows to dissect resistance to targeted therapies and inform the choice of combinations of targeted drugs based on tumor molecular alterations. Patient-specific biomarkers based on dynamical models of signaling networks can have a greater prognostic value than conventional biomarkers. These examples support systems biology models as valuable tools to advance clinical and translational oncological research.
Collapse
Affiliation(s)
- Andrea Rocca
- Hygiene and Public Health, Local Health Unit of Romagna, 47121 Forlì, Italy
| | - Boris N. Kholodenko
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
14
|
Thobe K, Konrath F, Chapuy B, Wolf J. Patient-Specific Modeling of Diffuse Large B-Cell Lymphoma. Biomedicines 2021; 9:biomedicines9111655. [PMID: 34829885 PMCID: PMC8615565 DOI: 10.3390/biomedicines9111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Personalized medicine aims to tailor treatment to patients based on their individual genetic or molecular background. Especially in diseases with a large molecular heterogeneity, such as diffuse large B-cell lymphoma (DLBCL), personalized medicine has the potential to improve outcome and/or to reduce resistance towards treatment. However, integration of patient-specific information into a computational model is challenging and has not been achieved for DLBCL. Here, we developed a computational model describing signaling pathways and expression of critical germinal center markers. The model integrates the regulatory mechanism of the signaling and gene expression network and covers more than 50 components, many carrying genetic lesions common in DLBCL. Using clinical and genomic data of 164 primary DLBCL patients, we implemented mutations, structural variants and copy number alterations as perturbations in the model using the CoLoMoTo notebook. Leveraging patient-specific genotypes and simulation of the expression of marker genes in specific germinal center conditions allows us to predict the consequence of the modeled pathways for each patient. Finally, besides modeling how genetic perturbations alter physiological signaling, we also predicted for each patient model the effect of rational inhibitors, such as Ibrutinib, that are currently discussed as possible DLBCL treatments, showing patient-dependent variations in effectiveness and synergies.
Collapse
Affiliation(s)
- Kirsten Thobe
- Mathematical Modelling of Cellular Processes, Max Delbrück Center for Molecular Medicine, 13125 Berlin-Buch, Germany; (K.T.); (F.K.)
| | - Fabian Konrath
- Mathematical Modelling of Cellular Processes, Max Delbrück Center for Molecular Medicine, 13125 Berlin-Buch, Germany; (K.T.); (F.K.)
| | - Björn Chapuy
- Department of Hematology and Medical Oncology, University of Göttingen, 37075 Göttingen, Germany;
- Department of Hematology, Oncology and Cancer Immunology, Berlin Medical Center Charité, 12203 Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max Delbrück Center for Molecular Medicine, 13125 Berlin-Buch, Germany; (K.T.); (F.K.)
- Department of Mathematics and Computer Science, Free University Berlin, Arnimallee 14, 14195 Berlin, Germany
- Correspondence:
| |
Collapse
|
15
|
Alveolar Regeneration in COVID-19 Patients: A Network Perspective. Int J Mol Sci 2021; 22:ijms222011279. [PMID: 34681944 PMCID: PMC8538208 DOI: 10.3390/ijms222011279] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
A viral infection involves entry and replication of viral nucleic acid in a host organism, subsequently leading to biochemical and structural alterations in the host cell. In the case of SARS-CoV-2 viral infection, over-activation of the host immune system may lead to lung damage. Albeit the regeneration and fibrotic repair processes being the two protective host responses, prolonged injury may lead to excessive fibrosis, a pathological state that can result in lung collapse. In this review, we discuss regeneration and fibrosis processes in response to SARS-CoV-2 and provide our viewpoint on the triggering of alveolar regeneration in coronavirus disease 2019 (COVID-19) patients.
Collapse
|
16
|
Niu J, Nguyen VA, Ghasemi M, Chen T, Mager DE. Cluster Gauss-Newton and CellNOpt Parameter Estimation in a Small Protein Signaling Network of Vorinostat and Bortezomib Pharmacodynamics. AAPS JOURNAL 2021; 23:110. [PMID: 34622346 DOI: 10.1208/s12248-021-00640-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/19/2021] [Indexed: 11/30/2022]
Abstract
Ordinary differential equation (ODE)-based models of signal transduction pathways often contain parameters that are unidentifiable or unmeasurable by experimental data, and calibrating such models to data remains challenging. Here, two efficient parameter estimation methods, cluster Gauss-Newton (CGN) and CellNOpt (CNO), were applied to fit a signaling network model of U266 multiple myeloma cells to the activity dynamics of key proteins in response to vorinostat and/or bortezomib. A logic-based network model was constructed and transformed to 17 ODEs with 79 parameters estimated within broad ranges of biologically plausible values. The top 10% best-fit parameters by both methods had high uncertainties with CV > 50% for the majority of parameters. The root mean square and prediction errors were comparable without statistically significant differences between the two methods. Despite uncertain parameter estimation, protein dynamics after the sequential combination of bortezomib and vorinostat was predicted with reasonable accuracy and precision. Global sensitivity analyses of partial rank correlation coefficients and Sobol sensitivity demonstrated that apoptosis induction was most sensitive to parameters governing the activity of the proteasome-JNK-caspase-8 axis. Simulations revealed that the greatest magnitude of pharmacodynamic drug interactions between bortezomib and vorinostat occurred at caspase-9, AKT, and Bcl-2. Two sequential combinations were explored in silico, and the outcome matched qualitatively with an empirical evaluation of the pharmacodynamic interaction based on cell viability. Overall, the CGN and CNO algorithms performed similarly for this ODE-based network model calibration, and the calibrated model provided meaningful insights into cellular signaling mechanisms in response to pharmacological perturbations.
Collapse
Affiliation(s)
- Jin Niu
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Van Anh Nguyen
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Mohammad Ghasemi
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Ting Chen
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, 431 Pharmacy Building, Buffalo, NY, 14214, USA.
| |
Collapse
|
17
|
Bērziņa S, Harrison A, Taly V, Xiao W. Technological Advances in Tumor-On-Chip Technology: From Bench to Bedside. Cancers (Basel) 2021; 13:cancers13164192. [PMID: 34439345 PMCID: PMC8394443 DOI: 10.3390/cancers13164192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Various 3D in vitro tumor models are rapidly advancing cancer research. Unlike animal models, they can be produced quickly and are amenable to high-throughput studies. Growing tumor spheroids in microfluidic tumor-on-chip platforms has particularly elevated the capabilities of such models. Tumor-on-chip devices can mimic multiple aspects of the dynamic in vivo tumor microenvironment in a precisely controlled manner. Moreover, new technologies for the on- and off-chip analysis of these tumor mimics are continuously emerging. There is thus an urgent need to review the latest developments in this rapidly progressing field. Here, we present an overview of the technological advances in tumor-on-chip technology by reviewing state-of-the-art tools for on-chip analysis. In particular, we evaluate the potential for tumor-on-chip technology to guide personalized cancer therapies. We strive to appeal to cancer researchers and biomedical engineers alike, informing on current progress, while provoking thought on the outstanding developments needed to achieve clinical-stage research. Abstract Tumor-on-chip technology has cemented its importance as an in vitro tumor model for cancer research. Its ability to recapitulate different elements of the in vivo tumor microenvironment makes it promising for translational medicine, with potential application in enabling personalized anti-cancer therapies. Here, we provide an overview of the current technological advances for tumor-on-chip generation. To further elevate the functionalities of the technology, these approaches need to be coupled with effective analysis tools. This aspect of tumor-on-chip technology is often neglected in the current literature. We address this shortcoming by reviewing state-of-the-art on-chip analysis tools for microfluidic tumor models. Lastly, we focus on the current progress in tumor-on-chip devices using patient-derived samples and evaluate their potential for clinical research and personalized medicine applications.
Collapse
|
18
|
Rozum JC, Gómez Tejeda Zañudo J, Gan X, Deritei D, Albert R. Parity and time reversal elucidate both decision-making in empirical models and attractor scaling in critical Boolean networks. SCIENCE ADVANCES 2021; 7:eabf8124. [PMID: 34272246 PMCID: PMC8284893 DOI: 10.1126/sciadv.abf8124] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/03/2021] [Indexed: 05/14/2023]
Abstract
We present new applications of parity inversion and time reversal to the emergence of complex behavior from simple dynamical rules in stochastic discrete models. Our parity-based encoding of causal relationships and time-reversal construction efficiently reveal discrete analogs of stable and unstable manifolds. We demonstrate their predictive power by studying decision-making in systems biology and statistical physics models. These applications underpin a novel attractor identification algorithm implemented for Boolean networks under stochastic dynamics. Its speed enables resolving a long-standing open question of how attractor count in critical random Boolean networks scales with network size and whether the scaling matches biological observations. Via 80-fold improvement in probed network size (N = 16,384), we find the unexpectedly low scaling exponent of 0.12 ± 0.05, approximately one-tenth the analytical upper bound. We demonstrate a general principle: A system's relationship to its time reversal and state-space inversion constrains its repertoire of emergent behaviors.
Collapse
Affiliation(s)
- Jordan C Rozum
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Jorge Gómez Tejeda Zañudo
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Xiao Gan
- Network Science Institute and Department of Physics, Northeastern University, Boston, MA 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dávid Deritei
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Réka Albert
- Department of Physics, The Pennsylvania State University, University Park, PA 16802, USA.
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
19
|
Cesareni G, Sacco F, Perfetto L. Assembling Disease Networks From Causal Interaction Resources. Front Genet 2021; 12:694468. [PMID: 34178043 PMCID: PMC8226215 DOI: 10.3389/fgene.2021.694468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/19/2021] [Indexed: 12/27/2022] Open
Abstract
The development of high-throughput high-content technologies and the increased ease in their application in clinical settings has raised the expectation of an important impact of these technologies on diagnosis and personalized therapy. Patient genomic and expression profiles yield lists of genes that are mutated or whose expression is modulated in specific disease conditions. The challenge remains of extracting from these lists functional information that may help to shed light on the mechanisms that are perturbed in the disease, thus setting a rational framework that may help clinical decisions. Network approaches are playing an increasing role in the organization and interpretation of patients' data. Biological networks are generated by connecting genes or gene products according to experimental evidence that demonstrates their interactions. Till recently most approaches have relied on networks based on physical interactions between proteins. Such networks miss an important piece of information as they lack details on the functional consequences of the interactions. Over the past few years, a number of resources have started collecting causal information of the type protein A activates/inactivates protein B, in a structured format. This information may be represented as signed directed graphs where physiological and pathological signaling can be conveniently inspected. In this review we will (i) present and compare these resources and discuss the different scope in comparison with pathway resources; (ii) compare resources that explicitly capture causality in terms of data content and proteome coverage (iii) review how causal-graphs can be used to extract disease-specific Boolean networks.
Collapse
Affiliation(s)
- Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Livia Perfetto
- Department of Biology, Fondazione Human Technopole, Milan, Italy
| |
Collapse
|
20
|
Han JZR, Hastings JF, Phimmachanh M, Fey D, Kolch W, Croucher DR. Personalized Medicine for Neuroblastoma: Moving from Static Genotypes to Dynamic Simulations of Drug Response. J Pers Med 2021; 11:395. [PMID: 34064704 PMCID: PMC8151552 DOI: 10.3390/jpm11050395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/19/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
High-risk neuroblastoma is an aggressive childhood cancer that is characterized by high rates of chemoresistance and frequent metastatic relapse. A number of studies have characterized the genetic and epigenetic landscape of neuroblastoma, but due to a generally low mutational burden and paucity of actionable mutations, there are few options for applying a comprehensive personalized medicine approach through the use of targeted therapies. Therefore, the use of multi-agent chemotherapy remains the current standard of care for neuroblastoma, which also conceptually limits the opportunities for developing an effective and widely applicable personalized medicine approach for this disease. However, in this review we outline potential approaches for tailoring the use of chemotherapy agents to the specific molecular characteristics of individual tumours by performing patient-specific simulations of drug-induced apoptotic signalling. By incorporating multiple layers of information about tumour-specific aberrations, including expression as well as mutation data, these models have the potential to rationalize the selection of chemotherapeutics contained within multi-agent treatment regimens and ensure the optimum response is achieved for each individual patient.
Collapse
Affiliation(s)
- Jeremy Z. R. Han
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; (J.Z.R.H.); (J.F.H.); (M.P.)
| | - Jordan F. Hastings
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; (J.Z.R.H.); (J.F.H.); (M.P.)
| | - Monica Phimmachanh
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; (J.Z.R.H.); (J.F.H.); (M.P.)
| | - Dirk Fey
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; (D.F.); (W.K.)
- Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland; (D.F.); (W.K.)
- Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - David R. Croucher
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; (J.Z.R.H.); (J.F.H.); (M.P.)
- St Vincent’s Hospital Clinical School, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
21
|
Finding new edges: systems approaches to MTOR signaling. Biochem Soc Trans 2021; 49:41-54. [PMID: 33544134 PMCID: PMC7924996 DOI: 10.1042/bst20190730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 11/17/2022]
Abstract
Cells have evolved highly intertwined kinase networks to finely tune cellular homeostasis to the environment. The network converging on the mechanistic target of rapamycin (MTOR) kinase constitutes a central hub that integrates metabolic signals and adapts cellular metabolism and functions to nutritional changes and stress. Feedforward and feedback loops, crosstalks and a plethora of modulators finely balance MTOR-driven anabolic and catabolic processes. This complexity renders it difficult — if not impossible — to intuitively decipher signaling dynamics and network topology. Over the last two decades, systems approaches have emerged as powerful tools to simulate signaling network dynamics and responses. In this review, we discuss the contribution of systems studies to the discovery of novel edges and modulators in the MTOR network in healthy cells and in disease.
Collapse
|
22
|
Béal J, Pantolini L, Noël V, Barillot E, Calzone L. Personalized logical models to investigate cancer response to BRAF treatments in melanomas and colorectal cancers. PLoS Comput Biol 2021; 17:e1007900. [PMID: 33507915 PMCID: PMC7872233 DOI: 10.1371/journal.pcbi.1007900] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 02/09/2021] [Accepted: 12/21/2020] [Indexed: 11/19/2022] Open
Abstract
The study of response to cancer treatments has benefited greatly from the contribution of different omics data but their interpretation is sometimes difficult. Some mathematical models based on prior biological knowledge of signaling pathways facilitate this interpretation but often require fitting of their parameters using perturbation data. We propose a more qualitative mechanistic approach, based on logical formalism and on the sole mapping and interpretation of omics data, and able to recover differences in sensitivity to gene inhibition without model training. This approach is showcased by the study of BRAF inhibition in patients with melanomas and colorectal cancers who experience significant differences in sensitivity despite similar omics profiles. We first gather information from literature and build a logical model summarizing the regulatory network of the mitogen-activated protein kinase (MAPK) pathway surrounding BRAF, with factors involved in the BRAF inhibition resistance mechanisms. The relevance of this model is verified by automatically assessing that it qualitatively reproduces response or resistance behaviors identified in the literature. Data from over 100 melanoma and colorectal cancer cell lines are then used to validate the model's ability to explain differences in sensitivity. This generic model is transformed into personalized cell line-specific logical models by integrating the omics information of the cell lines as constraints of the model. The use of mutations alone allows personalized models to correlate significantly with experimental sensitivities to BRAF inhibition, both from drug and CRISPR targeting, and even better with the joint use of mutations and RNA, supporting multi-omics mechanistic models. A comparison of these untrained models with learning approaches highlights similarities in interpretation and complementarity depending on the size of the datasets. This parsimonious pipeline, which can easily be extended to other biological questions, makes it possible to explore the mechanistic causes of the response to treatment, on an individualized basis.
Collapse
Affiliation(s)
- Jonas Béal
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Lorenzo Pantolini
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Vincent Noël
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| |
Collapse
|
23
|
Abstract
MOTIVATION Cells regulate themselves via dizzyingly complex biochemical processes called signaling pathways. These are usually depicted as a network, where nodes represent proteins and edges indicate their influence on each other. In order to understand diseases and therapies at the cellular level, it is crucial to have an accurate understanding of the signaling pathways at work. Since signaling pathways can be modified by disease, the ability to infer signaling pathways from condition- or patient-specific data is highly valuable. A variety of techniques exist for inferring signaling pathways. We build on past works that formulate signaling pathway inference as a Dynamic Bayesian Network structure estimation problem on phosphoproteomic time course data. We take a Bayesian approach, using Markov Chain Monte Carlo to estimate a posterior distribution over possible Dynamic Bayesian Network structures. Our primary contributions are (i) a novel proposal distribution that efficiently samples sparse graphs and (ii) the relaxation of common restrictive modeling assumptions. RESULTS We implement our method, named Sparse Signaling Pathway Sampling, in Julia using the Gen probabilistic programming language. Probabilistic programming is a powerful methodology for building statistical models. The resulting code is modular, extensible and legible. The Gen language, in particular, allows us to customize our inference procedure for biological graphs and ensure efficient sampling. We evaluate our algorithm on simulated data and the HPN-DREAM pathway reconstruction challenge, comparing our performance against a variety of baseline methods. Our results demonstrate the vast potential for probabilistic programming, and Gen specifically, for biological network inference. AVAILABILITY AND IMPLEMENTATION Find the full codebase at https://github.com/gitter-lab/ssps. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- David Merrell
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - Anthony Gitter
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, WI 53726, USA
| |
Collapse
|
24
|
Khalilimeybodi A, Paap AM, Christiansen SLM, Saucerman JJ. Context-specific network modeling identifies new crosstalk in β-adrenergic cardiac hypertrophy. PLoS Comput Biol 2020; 16:e1008490. [PMID: 33338038 PMCID: PMC7781532 DOI: 10.1371/journal.pcbi.1008490] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/04/2021] [Accepted: 11/05/2020] [Indexed: 11/25/2022] Open
Abstract
Cardiac hypertrophy is a context-dependent phenomenon wherein a myriad of biochemical and biomechanical factors regulate myocardial growth through a complex large-scale signaling network. Although numerous studies have investigated hypertrophic signaling pathways, less is known about hypertrophy signaling as a whole network and how this network acts in a context-dependent manner. Here, we developed a systematic approach, CLASSED (Context-specific Logic-bASed Signaling nEtwork Development), to revise a large-scale signaling model based on context-specific data and identify main reactions and new crosstalks regulating context-specific response. CLASSED involves four sequential stages with an automated validation module as a core which builds a logic-based ODE model from the interaction graph and outputs the model validation percent. The context-specific model is developed by estimation of default parameters, classified qualitative validation, hybrid Morris-Sobol global sensitivity analysis, and discovery of missing context-dependent crosstalks. Applying this pipeline to our prior-knowledge hypertrophy network with context-specific data revealed key signaling reactions which distinctly regulate cell response to isoproterenol, phenylephrine, angiotensin II and stretch. Furthermore, with CLASSED we developed a context-specific model of β-adrenergic cardiac hypertrophy. The model predicted new crosstalks between calcium/calmodulin-dependent pathways and upstream signaling of Ras in the ISO-specific context. Experiments in cardiomyocytes validated the model’s predictions on the role of CaMKII-Gβγ and CaN-Gβγ interactions in mediating hypertrophic signals in ISO-specific context and revealed a difference in the phosphorylation magnitude and translocation of ERK1/2 between cardiac myocytes and fibroblasts. CLASSED is a systematic approach for developing context-specific large-scale signaling networks, yielding insights into new-found crosstalks in β-adrenergic cardiac hypertrophy. Pathological cardiac hypertrophy is a disease in which the heart grows abnormally in response to different motivators such as high blood pressure or variations in hormones and growth factors. The shape of the heart after its growth depends on the context in which it grows. Since cell signaling in the cardiac cells plays a key role in the determination of heart shape, a thorough understanding of cardiac cells signaling in each context enlightens the mechanisms which control response of cardiac cells. However, cell signaling in cardiac hypertrophy comprises a complex web of pathways with numerous interactions, and predicting how these interactions control the hypertrophic signal in each context is not achievable by only experiments or general computational models. To address this need, we developed an approach to bring together the experimental data of each context with a signaling network curated from literature to identify the main players of cardiac cells response in each context and attain the context-specific models of cardiac hypertrophy. By utilizing our approach, we identified the main regulators of cardiac hypertrophy in four important contexts. We developed a network model of β-adrenergic cardiac hypertrophy, and predicted and validated new interactions that regulate cardiac cells response in this context.
Collapse
Affiliation(s)
- Ali Khalilimeybodi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Alexander M. Paap
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Steven L. M. Christiansen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
25
|
Modelling of Immune Checkpoint Network Explains Synergistic Effects of Combined Immune Checkpoint Inhibitor Therapy and the Impact of Cytokines in Patient Response. Cancers (Basel) 2020; 12:cancers12123600. [PMID: 33276543 PMCID: PMC7761568 DOI: 10.3390/cancers12123600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/23/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The future of cancer immunotherapy relies on a combination of individually targeted therapies. However, a lot of experiments are needed to define the most effective combinations of drugs. A computational and modelling approach could help reduce the number of experiments and suggest optimal treatments to test. This article presents a logical model of T cell activation influenced by immune checkpoints, and explores the effect of these checkpoints, suggests mechanisms that would explain why some treatments might be better suited than others. The model includes not only programmed cell death protein 1 (PD1) and cytotoxic T-lymphocyte-associated protein 4 (CTL4) downstream pathways but also those of other immune checkpoints such as T cell immunoglobulin and ITIM (immunoreceptor tyrosine-based inhibition motif) domain (TIGIT), lymphocyte activation gene 3 (LAG3), T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), cluster of differentiation 226 (CD226), inducible T-cell costimulator (ICOS), and tumour necrosis factor receptors (TNFRs). Abstract After the success of the new generation of immune therapies, immune checkpoint receptors have become one important center of attention of molecular oncologists. The initial success and hopes of anti-programmed cell death protein 1 (anti-PD1) and anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA4) therapies have shown some limitations since a majority of patients have continued to show resistance. Other immune checkpoints have raised some interest and are under investigation, such as T cell immunoglobulin and ITIM (immunoreceptor tyrosine-based inhibition motif) domain (TIGIT), inducible T-cell costimulator (ICOS), and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), which appear as promising targets for immunotherapy. To explore their role and study possible synergetic effects of these different checkpoints, we have built a model of T cell receptor (TCR) regulation including not only PD1 and CTLA4, but also other well studied checkpoints (TIGIT, TIM3, lymphocyte activation gene 3 (LAG3), cluster of differentiation 226 (CD226), ICOS, and tumour necrosis factor receptors (TNFRs)) and simulated different aspects of T cell biology. Our model shows good correspondence with observations from available experimental studies of anti-PD1 and anti-CTLA4 therapies and suggest efficient combinations of immune checkpoint inhibitors (ICI). Among the possible candidates, TIGIT appears to be the most promising drug target in our model. The model predicts that signal transducer and activator of transcription 1 (STAT1)/STAT4-dependent pathways, activated by cytokines such as interleukin 12 (IL12) and interferon gamma (IFNG), could improve the effect of ICI therapy via upregulation of Tbet, suggesting that the effect of the cytokines related to STAT3/STAT1 activity is dependent on the balance between STAT1 and STAT3 downstream signalling.
Collapse
|
26
|
Niarakis A, Helikar T. A practical guide to mechanistic systems modeling in biology using a logic-based approach. Brief Bioinform 2020; 22:5925256. [PMID: 33064138 DOI: 10.1093/bib/bbaa236] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/10/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022] Open
Abstract
Mechanistic computational models enable the study of regulatory mechanisms implicated in various biological processes. These models provide a means to analyze the dynamics of the systems they describe, and to study and interrogate their properties, and provide insights about the emerging behavior of the system in the presence of single or combined perturbations. Aimed at those who are new to computational modeling, we present here a practical hands-on protocol breaking down the process of mechanistic modeling of biological systems in a succession of precise steps. The protocol provides a framework that includes defining the model scope, choosing validation criteria, selecting the appropriate modeling approach, constructing a model and simulating the model. To ensure broad accessibility of the protocol, we use a logical modeling framework, which presents a lower mathematical barrier of entry, and two easy-to-use and popular modeling software tools: Cell Collective and GINsim. The complete modeling workflow is applied to a well-studied and familiar biological process-the lac operon regulatory system. The protocol can be completed by users with little to no prior computational modeling experience approximately within 3 h.
Collapse
Affiliation(s)
- Anna Niarakis
- GenHotel, Univ Evry, University of Paris-Saclay, Genopole, 91025 Evry, France and Lifeware Group, Inria Saclay-île de France, Palaiseau 91120, France
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
27
|
Tsirvouli E, Touré V, Niederdorfer B, Vázquez M, Flobak Å, Kuiper M. A Middle-Out Modeling Strategy to Extend a Colon Cancer Logical Model Improves Drug Synergy Predictions in Epithelial-Derived Cancer Cell Lines. Front Mol Biosci 2020; 7:502573. [PMID: 33195403 PMCID: PMC7581946 DOI: 10.3389/fmolb.2020.502573] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 09/22/2020] [Indexed: 11/23/2022] Open
Abstract
Cancer is a heterogeneous and complex disease and one of the leading causes of death worldwide. The high tumor heterogeneity between individuals affected by the same cancer type is accompanied by distinct molecular and phenotypic tumor profiles and variation in drug treatment response. In silico modeling of cancer as an aberrantly regulated system of interacting signaling molecules provides a basis to enhance our biological understanding of disease progression, and it offers the means to use computer simulations to test and optimize drug therapy designs on particular cancer types and subtypes. This sets the stage for precision medicine: the design of treatments tailored to individuals or groups of patients based on their tumor-specific molecular cancer profiles. Here, we show how a relatively large manually curated logical model can be efficiently enhanced further by including components highlighted by a multi-omics data analysis of data from Consensus Molecular Subtypes covering colorectal cancer. The model expansion was performed in a pathway-centric manner, following a partitioning of the model into functional subsystems, named modules. The resulting approach constitutes a middle-out modeling strategy enabling a data-driven expansion of a model from a generic and intermediate level of molecular detail to a model better covering relevant processes that are affected in specific cancer subtypes, comprising 183 biological entities and 603 interactions between them, partitioned in 25 functional modules of varying size and structure. We tested this model for its ability to correctly predict drug combination synergies, against a dataset of experimentally determined cell growth responses with 18 drugs in all combinations, on eight cancer cell lines. The results indicate that the extended model had an improved accuracy for drug synergy prediction for the majority of the experimentally tested cancer cell lines, although significant improvements of the model's predictive performance are still needed. Our study demonstrates how a tumor-data driven middle-out approach toward refining a logical model of a biological system can further customize a computer model to represent specific cancer cell lines and provide a basis for identifying synergistic effects of drugs targeting specific regulatory proteins. This approach bridges between preclinical cancer model data and clinical patient data and may thereby ultimately be of help to develop patient-specific in silico models that can steer treatment decisions in the clinic.
Collapse
Affiliation(s)
- Eirini Tsirvouli
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Vasundra Touré
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Barbara Niederdorfer
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Miguel Vázquez
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Åsmund Flobak
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- The Cancer Clinic, St. Olav’s University Hospital, Trondheim, Norway
| | - Martin Kuiper
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
28
|
Niederdorfer B, Touré V, Vazquez M, Thommesen L, Kuiper M, Lægreid A, Flobak Å. Strategies to Enhance Logic Modeling-Based Cell Line-Specific Drug Synergy Prediction. Front Physiol 2020; 11:862. [PMID: 32848834 PMCID: PMC7399174 DOI: 10.3389/fphys.2020.00862] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Discrete dynamical modeling shows promise in prioritizing drug combinations for screening efforts by reducing the experimental workload inherent to the vast numbers of possible drug combinations. We have investigated approaches to predict combination responses across different cancer cell lines using logic models generated from one generic prior-knowledge network representing 144 nodes covering major cancer signaling pathways. Cell-line specific models were configured to agree with baseline activity data from each unperturbed cell line. Testing against experimental data demonstrated a high number of true positive and true negative predictions, including also cell-specific responses. We demonstrate the possible enhancement of predictive capability of models by curation of literature knowledge further detailing subtle biologically founded signaling mechanisms in the model topology. In silico model analysis pinpointed a subset of network nodes highly influencing model predictions. Our results indicate that the performance of logic models can be improved by focusing on high-influence node protein activity data for model configuration and that these nodes accommodate high information flow in the regulatory network.
Collapse
Affiliation(s)
- Barbara Niederdorfer
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Vasundra Touré
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Miguel Vazquez
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Barcelona Supercomputing Center, Barcelona, Spain
| | - Liv Thommesen
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Martin Kuiper
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Astrid Lægreid
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Åsmund Flobak
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,The Cancer Clinic, St. Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
29
|
Lapuente-Santana Ó, Eduati F. Toward Systems Biomarkers of Response to Immune Checkpoint Blockers. Front Oncol 2020; 10:1027. [PMID: 32670886 PMCID: PMC7326813 DOI: 10.3389/fonc.2020.01027] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy with checkpoint blockers (ICBs), aimed at unleashing the immune response toward tumor cells, has shown a great improvement in overall patient survival compared to standard therapy, but only in a subset of patients. While a number of recent studies have significantly improved our understanding of mechanisms playing an important role in the tumor microenvironment (TME), we still have an incomplete view of how the TME works as a whole. This hampers our ability to effectively predict the large heterogeneity of patients' response to ICBs. Systems approaches could overcome this limitation by adopting a holistic perspective to analyze the complexity of tumors. In this Mini Review, we focus on how an integrative view of the increasingly available multi-omics experimental data and computational approaches enables the definition of new systems-based predictive biomarkers. In particular, we will focus on three facets of the TME toward the definition of new systems biomarkers. First, we will review how different types of immune cells influence the efficacy of ICBs, not only in terms of their quantification, but also considering their localization and functional state. Second, we will focus on how different cells in the TME interact, analyzing how inter- and intra-cellular networks play an important role in shaping the immune response and are responsible for resistance to immunotherapy. Finally, we will describe the potential of looking at these networks as dynamic systems and how mathematical models can be used to study the rewiring of the complex interactions taking place in the TME.
Collapse
Affiliation(s)
- Óscar Lapuente-Santana
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Federica Eduati
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
30
|
Eduati F, Jaaks P, Wappler J, Cramer T, Merten CA, Garnett MJ, Saez-Rodriguez J. Patient-specific logic models of signaling pathways from screenings on cancer biopsies to prioritize personalized combination therapies. Mol Syst Biol 2020; 16:e9690. [PMID: 33438807 DOI: 10.15252/msb.209690] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
31
|
Hastings JF, O'Donnell YEI, Fey D, Croucher DR. Applications of personalised signalling network models in precision oncology. Pharmacol Ther 2020; 212:107555. [PMID: 32320730 DOI: 10.1016/j.pharmthera.2020.107555] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
As our ability to provide in-depth, patient-specific characterisation of the molecular alterations within tumours rapidly improves, it is becoming apparent that new approaches will be required to leverage the power of this data and derive the full benefit for each individual patient. Systems biology approaches are beginning to emerge within this field as a potential method of incorporating large volumes of network level data and distilling a coherent, clinically-relevant prediction of drug response. However, the initial promise of this developing field is yet to be realised. Here we argue that in order to develop these precise models of individual drug response and tailor treatment accordingly, we will need to develop mathematical models capable of capturing both the dynamic nature of drug-response signalling networks and key patient-specific information such as mutation status or expression profiles. We also review the modelling approaches commonly utilised within this field, and outline recent examples of their use in furthering the application of systems biology for a precision medicine approach to cancer treatment.
Collapse
Affiliation(s)
- Jordan F Hastings
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | | | - Dirk Fey
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - David R Croucher
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia; School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland; St Vincent's Hospital Clinical School, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|