TY - JOUR T1 - LASSIM - a network inference toolbox for genome-wide mechanistic modeling JF - bioRxiv DO - 10.1101/115477 SP - 115477 AU - Rasmus Magnusson AU - Guido Pio Mariotti AU - Mattias Köpsén AU - William Lövfors AU - Danuta R Gawel AU - Rebecka Jörnsten AU - Jörg Linde AU - Torbjörn Nordling AU - Elin Nyman AU - Sylvie Schulze AU - Colm E Nestor AU - Huan Zhang AU - Gunnar Cedersund AU - Mikael Benson AU - Andreas Tjärnberg AU - Mika Gustafsson Y1 - 2017/01/01 UR - http://biorxiv.org/content/early/2017/03/09/115477.abstract N2 - Recent technological advancements have made time-resolved, quantitative, multi-omics data available for many model systems, which could be integrated for systems pharmacokinetic use. Here, we present large-scale simulation modeling (LASSIM), which is the first general mathematical tool for performing large-scale inference using mechanistically defined ordinary differential equations (ODE) for gene regulatory networks (GRNs). LASSIM integrates structural knowledge about regulatory interactions and non-linear equations with multiple steady states and dynamic response expression datasets. The rationale behind LASSIM is that biological GRNs can be simplified using a limited subset of core genes that are assumed to regulate all other gene transcription events in the network. LASSIM models are built in two steps, where each step can integrate multiple data-types, and the method is implemented as a general-purpose toolbox using the PyGMo Python package to make the most of multicore computers and high performance clusters, and is available at https://gitlab.com/Gustafsson-lab/lassim. As a method, LASSIM first infers a non-linear ODE system of the pre-specified core genes. Second, LASSIM optimizes the parameters that models the regulation of peripheral genes by core-system genes in parallel. We showed the usefulness of this method by applying LASSIM to infer a large-scale nonlinear model of naïve Th2 differentiation, made possible by integrating Th2 specific bindings, time-series and six public and six novel siRNA-mediated knock-down experiments. ChIP-seq showed significant overlap for all tested transcription factors. Next, we performed novel time-series measurements of total T-cells during differentiation towards Th2 and verified that our LASSIM model could monitor those data significantly better than comparable models that used the same Th2 bindings. In summary, the LASSIM toolbox opens the door to a new type of model-based data analysis that combines the strengths of reliable mechanistic models with truly systems-level data. We exemplified the advantage by inferring the first mechanistically motivated genome-wide model of the Th2 transcription regulatory system, which plays an important role in the progression of immune related diseases.Author summary There are excellent methods to mathematically model time-resolved biological data on a small scale using accurate mechanistic models. Despite the rapidly increasing availability of such data, mechanistic models have not been applied on a genome-wide level due to excessive runtimes and the non-identifiability of model parameters. However, genome-wide, mechanistic models could potentially answer key clinical questions, such as finding the best drug combinations to induce an expression change from a disease to a healthy state.We present LASSIM, which is a toolbox built to infer parameters within mechanistic models on a genomic scale. This is made possible due to a property shared across biological systems, namely the existence of a subset of master regulators, here denoted the core system. The introduction of a core system of genes simplifies the inference into small solvable subproblems, and implies that all main regulatory actions on peripheral genes come from a small set of regulator genes. This separation allows substantial parts of computations to be solved in parallel, i.e. permitting the use of a computer cluster, which substantially reduces the time required for the computation to finish. ER -