ArticlePDF Available

MUFINS: Multi-formalism interaction network simulator

Authors:

Abstract and Figures

Systems Biology has established numerous approaches for mechanistic modelling of molecular networks in the cell and a legacy of models. The current frontier is the integration of models expressed in different formalisms to address the multi-scale biological system organisation challenge. We present MUFINS software, implementing a unique set of approaches for multiformalism simulation of interaction networks. We extend the constraint-based modelling (CBM) framework by incorporation of linear inhibition constraints, enabling for the first time linear modelling of networks simultaneously describing gene regulation, signalling and whole-cell metabolism at steady state. We present a use case where a logical hypergraph model of a regulatory network is expressed by linear constraints and integrated with a Genome Scale Metabolic Network (GSMN) of mouse macrophage. We experimentally validate predictions, demonstrating application of our software in an iterative cycle of hypothesis generation, validation and model refinement. MUFINS incorporates an extended version of our Quasi Steady State Petri Net approach to integrate dynamic models with CBM, which we demonstrate through a dynamic model of cortisol signalling integrated with the human Recon2 GSMN and a model of nutrient dynamics in physiological compartments. Finally, we implement a number of methods for deriving metabolic states from ~omics data, including our new variant of the iMAT congruency approach. We compare our approach with iMAT through analysis of 262 individual tumour transcriptomes, recovering features of metabolic reprogramming in cancer. The software provides graphics user interface with network visualisation, which facilitates use by researchers who are not experienced in coding and mathematical modelling environments.
Mechanistic interpretation of experimental data on perturbation of whole-cell metabolic function by signaling network input and inhibitor. MUFINS was used to integrate a genome-scale metabolic model of the mouse macrophage (RAW264.7) with a large-scale regulatory network. Perturbation of whole-cell metabolism was simulated through activation and inhibition of the signaling network with external production of nitric oxide set as the objective function. Predicted data was then compared with the experimental data. (a) The left panel shows a screenshot of the JyMet interface, demonstrating on screen visualization of the reconstruction, created by automatic hierarchical layout with manual adjustment. Hatched lines are used to indicate regulatory signals, representing inhibition (circle end) or stimulation (arrow head). The right panel is a manually created image representing the pathway examined through JyMet; arrows represent signal flux, while open and filled circles represent inhibition and stimulation, respectively. The visualization depicts where signaling pathways converge on the iNOS gene, which is required for nitric oxide (NO) production in the whole-cell stoichiometric model. Flux rates for an example FBA solution are displayed on the network diagram; on the right panel only flux rates for each transitions are presented for clarity, while the left panel also shows the contribution of each substance to the flux. (b) The original reconstruction was able to predict the increase in NO production following stimulation with LPS, but not the impact of a MEK inhibitor, when compared with the experimental data of nitrate levels in RAW264.7 cell-conditioned medium (c). Nitrate can only be produced by non-enzymatic conversion of NO produced by RAW264.7 cells, and as there is no nitrate consumption in the medium, nitrate concentrations are proportional to nitric oxide production flux. (d) Refinement of the signaling network led to agreement between in silico prediction and in vitro measurement. The refinement was based upon two mechanistic hypotheses: (i) ERK1/2 is a more potent transcriptional activator of the iNOS gene than JNK and HIF1, and (ii) MEK1 is a more potent ERK1/2 kinase than PKC. FBA, Flux Balance Analysis; iNOS, inducible nitric oxide synthase; LPS, lipopolysaccharide; MUFINS, MUlti-Formalism Interaction Network Simulator; QSSPN, Quasi-Steady State Petri Net.
… 
Multi-formalism simulation integrating cortisol signaling with the human Recon2 GSMN reveals a drug interaction with estradiol clearance. (a) The Petri Net diagram of network connectivity created in the Snoopy editor, with overlaid comments for clarity. Color and symbol size has been manually set to match SBGN molecule types and transition types specific to QSSPN. The PN connectivity to implement a timer for administering a network perturbation (cortisol burst), is contained within a coarse transition and shown as an insert. (b) Simulation of glucose and lactate dynamics in the blood physiological compartment, demonstrating a convergence to physiologically realistic steady states. Perturbation of the system through a simulated cortisol infusion starting after 500 min elicits a dynamic alteration in the signaling network, resulting in (c) a predicted increase in CYP34A protein levels, which is confirmed in primary human hepatocytes. The increased expression of CYP3A4 protein is predicted to increase flux through reactions catalyzed by this enzyme, leading to: (d) degradation of excess cortisol and establishment of new steady state; (e) a drug–drug interaction for a second CYP3A4 substrate (estradiol), contained within the GSMN, leading to a decrease in it's steady-state level. The predicted increase in CYP3A4 activity following cortisol exposure is confirmed in primary human hepatocytes (f), as is the enhanced rate of estradiol clearance (g). FBA, Flux Balance Analysis; GSMN, Genome-Scale Metabolic Network; mRNA, messenger RNA; QSSPN, Quasi-Steady State Petri Net. **P o0.01, ***P o0.001.
… 
Content may be subject to copyright.
!
1!
MUFINS: Multi-Formalism Interaction Network Simulator 1!
Huihai Wu1, Axel von Kamp2, Vytautas Leoncikas1, Wataru Mori3, Nilgun Sahin4, Albert 2!
Gevorgyan5, Catherine Linley1, Marek Grabowski6, Ahmad A. Mannan1, Nicholas Stoy1, Graham 3!
R. Stewart1, Lara T. Ward7, David J.M. Lewis1, Jacek Sroka6, Hiroshi Matsuno3, Steffen Klamt2, 4!
Hans V. Westerhoff4,8,9, Johnjoe McFadden1, Nicholas J. Plant1*, Andrzej M. Kierzek1,10,* 5!
6!
*corresponding authors, a.kierzek@surrey.ac.uk, n.plant@surrey.ac.uk 7!
8!
E-mail addresses: Huihai Wu (h.wu@surrey.ac.uk); Axel von Kamp (vonkamp@mpi-9!
magdeburg.mpg.de); Vytautas Leoncikas (v.leoncikas@surrey.ac.uk); Wataru Mori 10!
(porepole8@gmail.com); Nilgun Sahin (nilguenyilmaz@gmail.com); Albert Gevorgyan 11!
(gevorgyana@MedImmune.com); Catherine Linley (c.linley@surrey.ac.uk); Marek Grabowski 12!
(grmarek@gmail.com); Ahmad A. Mannan (a.mannan@abdn.ac.uk); Nicholas Stoy 13!
(n.stoy@surrey.ac.uk); Graham R. Stewart (g.stewart@surrey.ac.uk); Lara T. Ward 14!
(lara.ward@AstraZeneca.com); David J. M. Lewis (d.j.lewis@surrey.ac.uk); Jacek Sroka 15!
(j.sroka@mimuw.edu.pl); Hiroshi Matsuno (matsuno@sci.yamaguchi-u.ac.jp); Steffen Klamt 16!
(klamt@mpi-magdeburg.mpg.de); Hans V. Westerhoff (Hans.Westerhoff@manchester.ac.uk; 17!
H.V.Westerhoff@uva.nl); Johnjoe McFadden (j.mcfadden@surrey.ac.uk); Nicholas J. Plant 18!
(n.plant@surrey.ac.uk); Andrzej M. Kierzek (a.kierzek@surrey.ac.uk); 19!
20!
Institutional addresses: 1) School of Biosciences and Medicine, Faculty of Health and Medical 21!
Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom. 2) Max Planck Institute for 22!
Dynamics of Complex Technical Systems, Magdeburg, Germany. 3) Graduate School of Science 23!
and Engineering & Faculty of Science, Yamaguchi University, Yoshida, Yamaguchi 753-8512, 24!
Japan 4) Molecular Cell Physiology, VU University Amsterdam, Amsterdam, The Netherlands 5) 25!
MedImmune Cambridge, CB21 6GH, United Kingdom 6) Institute of Informatics, University of 26!
!
2!
Warsaw, Warsaw, Poland 7) Oncology DMPK, AstraZeneca, Alderley Park, Cheshire, SK10 4TG, 27!
United Kingdom 8) Manchester Centre for Integrative Systems Biology, University of Manchester, 28!
Manchester, United Kingdom, 9) Synthetic Systems Biology, Netherlands Institute for Systems 29!
Biology, University of Amsterdam, Amsterdam, The Netherlands. 10) Simcyp Limited (a Certara 30!
Company), Sheffield, UK 31!
32!
33!
!
3!
ABSTRACT 34!
Systems Biology has established numerous approaches for mechanistic modelling of molecular 35!
networks in the cell and a legacy of models. The current frontier is the integration of models 36!
expressed in different formalisms to address the multi-scale biological system organisation 37!
challenge. We present MUFINS software, implementing a unique set of approaches for multi-38!
formalism simulation of interaction networks. We extend the constraint-based modelling (CBM) 39!
framework by incorporation of linear inhibition constraints, enabling for the first time linear 40!
modelling of networks simultaneously describing gene regulation, signalling and whole-cell 41!
metabolism at steady state. We present a use case where a logical hypergraph model of a regulatory 42!
network is expressed by linear constraints and integrated with a Genome Scale Metabolic Network 43!
(GSMN) of mouse macrophage. We experimentally validate predictions, demonstrating application 44!
of our software in an iterative cycle of hypothesis generation, validation and model refinement. 45!
MUFINS incorporates an extended version of our Quasi Steady State Petri Net approach to 46!
integrate dynamic models with CBM, which we demonstrate through a dynamic model of cortisol 47!
signalling integrated with the human Recon2 GSMN and a model of nutrient dynamics in 48!
physiological compartments. Finally, we implement a number of methods for deriving metabolic 49!
states from ~omics data, including our new variant of the iMAT congruency approach. We compare 50!
our approach with iMAT through analysis of 262 individual tumour transcriptomes, recovering 51!
features of metabolic reprogramming in cancer. The software provides graphics user interface with 52!
network visualisation, which facilitates use by researchers who are not experienced in coding and 53!
mathematical modelling environments. 54!
55!
56!
57!
58!
59!
!
4!
Introduction 60!
During the last two decades, Systems Biology has established numerous approaches to represent 61!
molecular biology knowledge in the form of mechanistic molecular interaction network models. 62!
This is accompanied by a legacy of thousands of experimentally validated models. Stochastic 63!
kinetic simulations provide the most detailed quantitative description, where individual reactive 64!
collisions occurring at exact times in single cells are simulated by the Gillespie algorithm (1). The 65!
Ordinary Differential Equation (ODE) formalism applied to study the time evolution of average 66!
molecular concentration in cellular population is a workhorse of quantitative modelling (2). While 67!
quantitative biology is developing rapidly, it is still not possible to parameterise quantitative 68!
dynamic models of whole-cell scale networks and simulate genotype-phenotype relationship. 69!
Rather, Constrained Based Modelling (CBM) has achieved spectacular success in modelling 70!
metabolism at the full genome scale (3, 4). The metabolic network can be modelled at quasi-steady 71!
state due to time-scale separation from gene regulation. This enables exploration of metabolic flux 72!
distributions consistent with stoichiometric and thermodynamic constraints as well as flux 73!
measurements and constraints formulated according to ~omics data on enzymatic gene expression 74!
(5). Modelling of large-scale gene regulatory and signalling networks is much more challenging and 75!
a number of qualitative simulation approaches have been formulated, such as analysis of steady 76!
states in logical hypergraphs (6), enumeration of states in dynamic Boolean models (7), Monte 77!
Carlo exploration of the alternative molecular transition sequences constrained by network 78!
connectivity expressed in Petri Net formalism (8, 9). Application of these methods has led to a 79!
legacy of models describing different levels of cellular organisations in different modelling 80!
frameworks. A large proportion of these models are already expressed in Systems Biology Markup 81!
Language (SBML) (10) and over 1000 literature-based models are available in the most recent 82!
version of BioModels (11). Notably, BioModels is attracting interest from the Physiologically 83!
Based Pharmacokinetic (PBPK) modelling field (12), where ODE models of substance 84!
!
5!
concentrations in physiological compartments are routinely used to inform drug development in 85!
pharmaceutical industry. 86!
Given this state of the art and the multi-scale nature of biological systems, the current 87!
challenge is integration of models expressed in different formalisms towards a multi-formalism 88!
simulation covering all scales of biological organisation. The model of Mycoplasma is currently the 89!
most complete in silico cell (13), and demonstrates that mechanistic modeling of the genotype-90!
phenotype relationship requires the integration of sub-system models describing different spatial 91!
and temporal scales constructed in different formalisms. The application of the multi-formalism 92!
approach toward modeling the relationship between genotype and human physiology is an 93!
emerging field and an important component of the personalized medicine challenge. For example, 94!
integration of a PBPK model with human liver-specific GSMN has allowed robust prediction of 95!
therapeutic response in humans (14). In our recent work we integrated a liver-specific GSMN with 96!
a qualitative model of a large-scale regulatory network (9), demonstrating how integration of gene 97!
regulation and metabolism in the context of physiological modeling can provide novel insights into 98!
toxicology, non-alcoholic liver disease and metabolic syndrome. This was achieved by application 99!
of our novel Quasi-Steady State Petri Net (QSSPN) (9) approach integrating CBM and qualitative, 100!
Monte Carlo simulation of a regulatory network represented as a Signaling Petri Net (15). 101!
Numerous alternate methods have been proposed to integrate GSMNs with ODE (16) and logical 102!
(17) dynamic models as well as hybrid algorithms, bridging the gap between exact stochastic and
103!
ODE regimes in fully parameterized dynamic models (18). To fully realize the potential of
104!
computational modelling, it is now imperative to develop software packages that allow the
105!
development and simulation of multi-formalism models in a user interface that is approachable for 106!
experimental scientists. 107!
Here, we present MUFINS (Multi Formalism Interaction Network Simulator) software and 108!
argue that it is the first general software with Graphics User Interface (GUI) capable of integrating 109!
models developed in all major modeling frameworks of Computational Systems Biology. This is 110!
!
6!
demonstrated through Use Cases; first, a model of the mammalian macrophage where linear 111!
inhibitory constraints are for the first time used to integrate a logical model of cellular signaling 112!
with the GSMN for the mammalian cell; second, a model of the human hepatocyte, where an 113!
extended version of our QSSPN method is used to integrate a human GSMN, a detailed kinetic
114!
model of cortisol signaling and a PBPK model; third, the analysis of clinical transcriptome data in
115!
the context of human GSMN using a novel variant of ~omics data integration approach. The use
116!
cases involve laboratory experiments to demonstrate how experimental biologists can utilise the 117!
MUFINS GUI in the iterative cycle of model development, hypothesis generation, experimental 118!
validation and model refinement. Moreover, a comprehensive comparison with other software 119!
shows that MUFINS implements the largest number of CBM methods under a GUI with interactive 120!
network visualization. Thus, MUFINS is uniquely suited for the development and simulation of 121!
multi-formalism models by a wide user community including experimental scientists with 122!
no/limited experience with programmatic interfaces and mathematical modeling environments.
123!
124!
Software overview. 125!
Figure 1 provides an overview of the MUFINS architecture. All simulations are performed in sfba 126!
and qsspn, two computational engines written in C++, which are run either through a GUI or in 127!
command line mode. The sfba code originates from our SurreyFBA software (19) and implements a 128!
comprehensive set of CBM approaches. The major new multi-formalism simulation feature added is
129!
linear inhibitor and activator constraints, which are described and validated in Use Case 1. In
130!
addition to the basic CBM methods available in SurreyFBA, the sfba engine of MUFINS 131!
implements a large number of ~omics data integration algorithms such as iMAT (20), GIMME 132!
(20), GIM3E (21), GNI (22) and our DPA (24). The GNI and DPA features of sfba have been 133!
already used to study M. tuberculosis metabolism (24,25)(23). Furthermore, we include Fast iMAT 134!
a new variant of iMAT approach demonstrated in Use Case 3 below. sfba uses the GLPK library for 135!
Linear Programming (LP) and Mixed Linear Integer Programming (MILP) calculations. However, 136!
!
7!
since MILP implementation in GLPK is inefficient, a version of sfba ready for use with Gurobi 137!
library is provided to facilitate application of MILP-based protocols (e.g. iMAT) on GSMN models. 138!
The qsspn is a computational engine for integration of dynamic and CBM models. It 139!
implements QSSPN approach (9), where a dynamic model constructed in Petri Net (PN) formalism
140!
(24) is connected to a steady state Flux Balance Analysis (FBA; (4)) through PN places setting FBA
141!
bounds and requesting evaluation of objective functions. Previously, we integrated a qualitative PN
142!
model with a hepatocyte GSMN and explored its qualitative dynamic behaviours through Monte 143!
Carlo simulation (15). Here we present a new version of the engine, providing full support for 144!
continuous Petri Nets, implementing ODE models and stochastic Petri Nets representing exact 145!
stochastic simulations. Use Case 2 demonstrates the first integration of quantitative models of gene 146!
regulation with human Recon2 GSMN coupled to an ODE model of nutrients in physiological 147!
compartments. The qualitative simulation features are also further extended by implementation of 148!
QSSPNclient, a version of qsspn which speeds up sampling of alternative qualitative trajectories by
149!
executing FBA on a server that tabulates repeated evaluations of GSMN objective functions for re-150!
occurring sets of bounds. The new version of the QSSPN algorithm, qsspn solver and QSSPNclient 151!
are described in detail as Supplementary Information (page 32). 152!
Connectivity of a Petri Net representing the dynamical part of the QSSPN model and its 153!
interactions with GSMN can be graphically defined using the Petri Net editor Snoopy (25): while 154!
Snoopy provides PN simulation, it does not implement QSSPN and thus is used here exclusively as
155!
an external editor of PN connectivity. Parameters specific to qsspn simulation can be provided
156!
either through the Comments section of place and transition objects, or added later through the 157!
MUFINS GUI. The Snoopy XML file is parsed by the spept2qsspn Python script, which generates 158!
the qsspn input file in qsspn’s native, human readable, text-file format. To facilitate future 159!
integration with other interfaces, a JSON-based input file format is also available. 160!
JyMet is the GUI of MUFINS, and provides an interface for multi-formalism simulations to 161!
users who are not familiar with programming, mathematical modeling environments or working in 162!
!
8!
command line. JyMet code is written in Jython (Java/Python), and originates from SurreyFBA (19). 163!
Here, it has been significantly extended through addition of QSSPN simulation and improved 164!
visualization of metabolic networks. JyMet integrates all elements of the MUFINS environment 165!
(Figure 1). It loads a Snoopy file defining PN connectivity and provides a table interface for
166!
definition of QSSPN specific parameters: place and transition types, lookup tables linking dynamic
167!
and GSMN variables, and arithmetic formulas describing complex rate laws. We note that users can
168!
build dynamic model within the QSSPN spreadsheet interface of JyMet without using Snoopy, but 169!
this is unlikely to be a preferable solution, due to the advantages of PN graphical modeling. Both 170!
sfba and qsspn can be run from JyMet and results are loaded to spreadsheets and plotting functions. 171!
Each table in JyMet can be exported in tab-separated format for further analysis with other 172!
software. Full features of JyMet are described in detail in software documentation and tutorials 173!
(MUFINS1.0_Doc/). Use Case 1 shows application of network visualization for exploration of 174!
alternative model solutions during the iterative cycle of network reconstruction, simulation,
175!
experimental validation and refinement. 176!
The sfba, qsspn and spept2qsspn tools can be run as stand-alone command line tools without 177!
external dependencies. Thus MUFINS is ideal for integration with web and desktop interfaces as 178!
well as computational pipelines. Use Case 3 shows integration of sfba engine with computational 179!
pipeline for analysis of clinical transcriptome data. Previous version of sfba engine (19) has been 180!
already used in web interfaces supporting development and publication of bacterial pathogen
181!
GSMN models (26, 27) and as one of computational engines in METEXPLORE web environment
182!
(28). 183!
MUFINS is open source software, distributed under the GNU GPL license. It can be run on 184!
OS X, Windows and Linux sand the majority of calculations can be run without dependencies. The 185!
methods applying MILP to genome scale models are likely to be very computationally expensive 186!
unless the Gurobi library is installed. 187!
188!
!
9!
Use Cases 189!
Table 1 summarizes Use Cases illustrating the multi-formalism simulation abilities within 190!
MUFINS, including previously published works. 191!
192!
Table 1. Summary of simulation formalisms applied in Use Cases.
193!
Use Case
Formalisms
Reference
Use Case 1: linear inhibitory
constraints
CBM, logical hypergraph, inhibitor and
activator constraints.
This work
Use Case 2: integration of
regulatory networks and GSMNs
CBM, ODE, Gillespie, physiological
compartment models.
This work and (9)
Use Case 3: Prediction of
metabolic landscapes
CBM, congruency approach to analysis
of ~omics data in the context of GSMNs.
This work and (29)
194!
Use case 1: Whole-cell metabolic reprogramming by signaling and gene regulatory networks in the 195!
mammalian macrophage. 196!
An important innovation in MUFINS is the ability to include stimulation and inhibition reactions 197!
within the genome-scale metabolic network. To demonstrate the utility of this approach to derive 198!
biological insights we present a use case entailing integration of gene and signaling regulatory 199!
networks with genome-scale metabolism for the mammalian macrophage. 200!
We applied MUFINS to integrate a logical hypergraph (6) model of the large-scale
201!
regulatory network responsible for the pathogen response of mammalian macrophage with the 202!
published GSMN of the mouse RAW264.7 macrophage cell line (30). A signal transduction 203!
network of 286 interactions and 205 species was reconstructed in CellNetAnalyzer using logical 204!
hypergraph formalism (6): A manually created graph image is shown in Figure 2, with full 205!
description of model construction described in Supplementary Information, and a detailed 206!
description of species, logical formulas and literature references in Supplementary Table 1. Briefly,
207!
!
10!
species within the regulatory network represent protein kinases, transcription factors, genes, 208!
antigens, cytokines and cellular behaviours (e.g. apoptosis) involved in the response of 209!
macrophages to bacterial pathogens. 210!
To integrate signaling and metabolic networks, we have translated the logical hypergraph to
211!
a stoichiometric model with inhibitory constraints. Representation of inhibition in the CBM
212!
framework has always been a challenge, with proposed solutions generally being computationally
213!
expensive methods based on MILP (31). We have extended the approach of Vardi and colleagues 214!
(32) and represented inhibition by linear constraints, enforcing a reciprocal relation between 215!
inhibitor production and inhibited flux. Differences between the original formulation and our 216!
extended implementation are presented as Supplementary Information. An example of the logical 217!
hypergraph conversion to MUFINS reaction formulas is shown in Figure 2. At the software level 218!
this is achieved by exporting CellNetAnalyzer logical rules as a text format and then using the text-219!
replace function in Excel to change the formula format and create a MUFINS reaction table that can
220!
be opened by the JyMet GUI. We note these steps do not require programming experience. 221!
Subsequently, the model was edited in JyMet to define input fluxes. Flux Variability Analysis was 222!
undertaken to identify spurious activations in the model with all input fluxes constrained to 0. 223!
Details of these steps are given in Supplementary Information (page 7), where we also compare our 224!
model pre-processing steps with the much more complex, MILP based protocol used by Vardi et al 225!
(32).
226!
A unique feature of MUFINS is reconstruction of models that combine GSMNs and
227!
regulatory networks with linear inhibitory constraints. To demonstrate this capability we integrated 228!
the signal transduction network model described above with the published GSMN of the 229!
RAW264.7 macrophage cell line (30). We have focused on nitric oxide production, a major 230!
metabolic function of macrophages interacting with bacterial pathogens. Our regulatory network 231!
model describes the regulation of the inducible nitric oxide synthase (iNOS) gene, which we have 232!
added as an activator for the NO synthase reaction in the GSMN (reaction id: R_NOS2). We used a 233!
!
11!
linear activator constraint, as described in Supplementary Information (page 31), to ensure that 234!
stoichiometry of the R_NOS2 reaction is not affected. Figure 3A shows a portion of the total 235!
regulatory network, specifically the immediate signaling pathways regulating iNOS gene 236!
expression. We used this integrated model to simulate nitric oxide production in response to
237!
lipopolysaccharide (LPS). Following Bordbar and colleagues (30) we constrained biomass reaction
238!
flux to 0.0281/h, which reproduces experimentally measured growth rates. We calculated the
239!
maximal extracellular nitric oxide production when LPS input flux to the regulatory network was 240!
opened or closed and when phosphorylation of ERK by MEK1 was inhibited or not. Results 241!
obtained from these four simulations are shown on Figure 3B, and demonstrate that nitric oxide is 242!
produced only when LPS activates the regulatory network, while the inhibitor does not influence 243!
results. The maximal flux through R_NOS agrees with the value of 0.0399 mmol/gDW/h reported 244!
in original publication, thus verifying SBML import of the GSMN model to JyMet. We note that 245!
while in this use case the regulatory network regulates only one enzyme, this is an example of
246!
major global metabolic reprogramming. The production of large amounts of nitric oxide in response 247!
to pathogen requires both precursors and energy, and the GSMN model accounts for stoichiometry 248!
of all reactions linking medium nutrients to metabolic output. Moreover, the GSMN assures that the 249!
cell satisfies other metabolic demands, such as the demand for biomass production whereby the 250!
GSMN model accounts for global stoichiometry of providing cellular components and maintaining 251!
energy during induced nitric oxide production.
252!
To compare the model predictions with experimental data we treated RAW264.7
253!
macrophages with LPS and a MEK inhibitor and measured nitrate concentration in the medium. 254!
Since nitrate can be produced only by the non-enzymatic conversion of nitric oxide from cells, and 255!
there is no nitrate consumption in the medium, concentrations are proportional to the nitric oxide 256!
production flux. Figure 3C shows that the model correctly predicts that LPS is obligate for the 257!
production of nitric oxide in RAW264.7 macrophages. However, the model did not predict the 258!
decrease in nitric oxide production caused by MEK inhibition. To explore this inconsistency, we 259!
!
12!
used the interactive network visualization available in JyMet (Figure 3A) to examine example FBA 260!
solutions. Multiple pathways lead to iNOS activation, some of which are not dependent on MEK. In 261!
the model, the “iNOS substance representing activity of the iNOS gene is produced by three 262!
reactions representing the activity of ERK1/2-, HIF1- and JNK-dependent regulation of iNOS gene
263!
expression. Each of these regulators is, activated by different upstream signaling cascades. We used
264!
JyMet to interactively simulate and visualize different scenarios and concluded that the
265!
experimental results can be replicated if the following assumptions are made: i) ERK induces a 266!
more potent activation of the iNOS gene than JNK and HIF1 ii) MEK1 is a more potent ERK kinase 267!
than PKC. These assumptions were introduced to the model by setting flux bounds of (0, 0.005) for 268!
the transitions “JNK -> iNOS”, “HIF1->iNOS” and “PKC_a_b = ERK1/2” (Figure 3A, right panel). 269!
The upper bound is arbitrary, and selected to ensure that the flux towards iNOS” via “HIF1” or 270!
JNK” reaches only a fraction of value required for maximal activation of R_NOS, with the 271!
remaining activation occurring via ERK1/2 regulation. This refined model is now able to reproduce
272!
the decreased, but not complete inhibition, of nitric oxide production by a MEK inhibitor (Figure 273!
3D). A full description of this cycle of prediction, experimental testing, and model refinement is 274!
presented as Supplementary Information (page 11), detailing how MUFINS and JyMet aid the 275!
iterative refinement cycle required during model development. This data supports the assumptions 276!
above being one possible mechanistic solution to reproduce the observed biological phenotype. 277!
However, we note that further experimental confirmation is required to confirm the predicted
278!
biological insight. In a full iterative cycle of prediction, experiment and model refinement, multiple
279!
molecular targets would be subject to independent experiment verification before the model was 280!
validated. Here, we show one full cycle of simulation and experiment to demonstrate how the 281!
JyMet interface is used in this iterative model development process. 282!
To summarize, we present the first linear model for steady state simulation of networks 283!
integrating signaling, gene regulation and whole-cell metabolism in a mammalian cell. Moreover, 284!
we present the first simulation of perturbation of a global metabolic output by a signaling network 285!
!
13!
inhibitor, and demonstrate this is consistent with experimental data. The ability to formulate 286!
hypotheses in terms of continuous “regulatory strength” is demonstrated. This offers significant 287!
advantages over MILP based approaches such as SR-FBA (33), where the regulatory network is 288!
used exclusively to formulate Boolean, on/off constraints. Finally, the graphics user interface JyMet
289!
allows an interactive exploration of combined signaling and metabolic flux distributions that is
290!
easily approachable by non-specialists. Together, these tools provide an ideal platform for non-
291!
specialists to generate mechanistic hypotheses based upon the interaction of gene and signal 292!
regulatory networks with genome-scale metabolism. These hypotheses can then drive experimental 293!
testing, enhancing our ability to identify novel biological insights. 294!
295!
Use case 2: Kinetic model of cortisol signaling integrated with dFBA simulation of human GSMN. 296!
An important challenge in computational biology is the generation of large-scale models that are 297!
able to reproduce diverse biological functions. One approach to achieve this aim is the integration
298!
of validated small models (or modules) to form larger networks. A major consideration in such 299!
integration is the ability to combine models across different modelling formalisms and biological 300!
scales. In this use case we demonstrate the utility of MUFINS for the generation and simulation of 301!
such multi-formalism, multi-scale models. 302!
Cortisol acts as an important signaling molecule within the body, with roles in circadian biology 303!
and the response to stress episodes. The level of cortisol in the body is interpreted at the cellular
304!
level through interaction with three nuclear receptors: the glucocorticoid receptor; the
305!
mineralocorticoid receptor; the pregnane-X receptor. These signals are integrated and produce a 306!
global metabolic shift corresponding to the current cortisol level. To reproduce such a complex 307!
biological phenomenon it is necessary to combine multiple signaling cascades with genome-scale 308!
reconstructions of metabolism. Here, we demonstrate the application of MUFINS for such a multi-309!
formalism simulation, integrating a detailed kinetic model of cortisol signaling in the liver, a 310!
genome-scale model of liver metabolism, and an ODE model of glucose and lactate dynamics in the 311!
!
14!
blood (Figure 4). This is the first simulation integrating a human GSMN, physiological level ODE 312!
model and detailed kinetic model of an intracellular regulatory network. Thus, this use case 313!
demonstrates that MUFINS provides a unique tool for the integration of PBPK models with the 314!
mechanistic models of molecular networks operating in mammalian tissues.
315!
The model is depicted in Figure 4A. We represent our previously published kinetic model of
316!
cortisol signaling in liver (34, 35) as a Petri net (PN) using Snoopy software (25). Size and colour
317!
of place/transition symbols was used to mirror Systems Biology Graphical Notation (36) molecule 318!
types as well as QSSPN specific place/transition types. The PN transition rates were defined using 319!
the ODE terms of the kinetic model. PN places represent molecular concentrations. This dynamic 320!
model of cortisol signaling was linked to the human GSMN Recon2, with the CYP3A4 enzyme 321!
used as the QSSPN constraint place. Details of the cortisol signaling model and its coupling to 322!
GSMN are available Supplementary Information (page 16). 323!
To model whole-cell metabolism of hepatocytes we used the community-based Recon2
324!
GSMN (37). This model incorporates liver specific reactions from the HepatoNet1 (38), but is a 325!
much more extensive reconstruction of cellular metabolism. Exchange fluxes were constrained 326!
using the HepatoNet1 Physiological Import and Export set (PIPES). The objective function was set 327!
to glucose regeneration from lactate, a major physiological function of the liver, where blood 328!
glucose and lactate concentrations are ODE variables. The dFBA simulation is implemented using 329!
place and transition within the QSSPN, rather than coded as a separate approach. We have further
330!
capitalized on the flexibility of the PN representation to create a timer administering a cortisol
331!
infusion after 500 minutes. This demonstrates how multi-formalism simulations in MUFINS can 332!
include complex, time-dependent perturbations to the model such as boluses or cell division events. 333!
For clarity, the timer is contained within a coarse transition, with the sub-level shown as an inset to 334!
figure 4A. A detailed description of QSSPN place and transition types is given as Supplementary 335!
Information (page 27), along with a detailed description of model construction (page 12). 336!
!
15!
Simulations of the systems response to cortisol infusion, plus experimental confirmation are 337!
shown in Figure 4B-G. As shown in figure 4B, glucose and lactate concentrations converge to their 338!
physiological levels of 4.45 mM and 1.48 mM respectively (39) and are maintained during cortisol 339!
infusion from 500 minutes onwards. Cortisol infusion from 500 minutes produces a number of
340!
effects, which are mediated through activation of the two cognate nuclear receptors for cortisol
341!
within the regulatory network, PXR and GR (40, 41). The cortisol-mediated activation of PXR
342!
results in a predicted increase in expression of the CYP3A4 enzyme (Figure 4C), which is 343!
experimentally confirmed at the transcript, protein and activity levels in vitro using primary human 344!
hepatocytes (Figure 4D). CYP3A4 is one of the main enzymes responsible for the metabolic 345!
clearance of cortisol; thus, the combination of a constant infusion of cortisol, followed by increased 346!
metabolism, results in an elevated blood cortisol level (Figure 4E). We note that the transition to 347!
this new blood cortisol level is not instant, demonstrating a concentration spike consistent with the 348!
time delay caused by the de novo production (i.e. transcription and translation) of CYP3A4 protein.
349!
Finally, in Figure 4F we demonstrate that the cortisol infusion propagates through the signaling and 350!
metabolic networks, leading to predicted changes in blood concentrations for other chemicals. 351!
Estradiol is an endogenous hormone important in a range of biological functions, including 352!
development of the secondary sexual organs in both sexes and proliferation during the menstrual 353!
cycle and pregnancy in women (42, 43). It has considerable clinical application, most notably as a 354!
contraceptive, either as the native compound or synthetic derivatives (44). As shown in figure 4E,
355!
following the cortisol infusion, predicted levels of blood estradiol drop rapidly, decreasing
356!
approximately by one-halve within 500mins. This lower level is maintained throughout the period 357!
where CYP3A4 protein levels are elevated. To confirm this effect, we have measured the clearance 358!
rate for estradiol in naïve primary human hepatocytes and compared it to hepatocytes pre-exposed 359!
to 1µM cortisol, demonstrating enhanced clearance in cortisol-exposed hepatocytes (Figure 4G). In 360!
addition, we note that activation of PXR has previously been linked with a number of drug-drug 361!
interactions with estrogens, demonstrating the extrapolation of these predictions to the clinical 362!
!
16!
setting (41, 45). It is important to note that estradiol concentration was not a variable of the detailed 363!
kinetic model, and was rather identified as a variable of interest by examination of perturbed 364!
GSMN fluxes. This demonstrates how integration of detailed kinetic models with GSMNs can lead 365!
to identification of interactions of biological interest. As such, this approach has much potential for
366!
the prediction of clinically relevant drug-induced disruption of homeostasis, and drug-drug
367!
interactions (41, 45).
368!
In summary, this use case demonstrates the utility of MUFINS to combine legacy models 369!
developed in different formalisms and link molecular network knowledge to quantitative data on 370!
substance concentration at physiological level. As such, MUFINS represents the first software to 371!
allow such multi-scale, multi-formalism simulations through a GUI approachable to the non-372!
specialist. 373!
374!
Use case 3: Analysis of a clinical transcriptome data to understand in vivo tumour metabolism
375!
An important branch of CBM methodology (5) is dedicated to using ~omics data to create tissue 376!
and/or condition specific GSMNs. MUFINS is equipped with state-of-the art CBM methods in this 377!
area; in addition, we have developed Fast iMAT, a new variant of the iMAT approach that is 378!
applicable to large ~omics sample numbers, where iMAT becomes impractical. We recently 379!
reported a preliminary version of Fast iMAT (29), dedicated to the analysis of expression data 380!
discretized to two states (absent or present transcript). Personalized GSMNs for 2000 breast
381!
tumours were generated, identifying a low prognosis cluster with active serotonin production an
382!
important biological insight (29). The first distribution version of MUFINS provides a mature 383!
version of the Fast iMAT algorithm. To demonstrate its utility, we analyze 262 previously 384!
unexamined paired clinical transcriptome samples (46). We demonstrate the significant up-385!
regulation of kyneurenine synthesis in tumour compared to normal breast tissue, and important pro-386!
survival phenotype (47, 48). 387!
388!
!
17!
Comparison of MUFINS with existing tools. 389!
A comparison of MUFINS with existing tools is presented as Supplementary Information. We 390!
conclude that MUFINS is currently the only software supporting integration of i) exact stochastic, 391!
ii) ODE, iii) qualitative dynamic, iv) logical steady state and v) CBM models in a general software
392!
platform with GUI. The only alternative to achieve integration of this range of formalisms is coding
393!
of the model in mathematical modeling environment. While this strategy has achieved success (13),
394!
it is not a plausible proposition for non-specialists who lack programming skills. Moreover, multi-395!
formalism modeling in a mathematical language involves the implementation of a simulation 396!
algorithm dedicated to each model. In MUFINS, each model is run in the same QSSPN simulation 397!
algorithm, with multi-formalism functionality emerging from the interactions between the different 398!
types of Petri Net places and transitions that can be graphically assembled, leading to a 399!
combinatorial diversity of types of models that can be simulated. Using one algorithm and a few 400!
well-defined place and transition types provides clearer control and description of model
401!
assumptions than coding a different main simulation loop for each model. Also, the algorithms 402!
available in MUFINS are validated and optimized against the legacy of previous applications, while 403!
formulation, validation and description of a simulation algorithm dedicated to a particular model 404!
will take additional time. It is our experience that even scientists who can program will find it easier 405!
to implement complex multi formalism models by connecting QSSPN places and transitions to off-406!
the-shelf GSMN models imported to JyMet, rather then by development of dedicated mathematical
407!
modeling code. Moreover, MUFINS provides a wide range of CBM methods that can be used to
408!
model GSMNs before their integration with dynamic models. As Supplementary Information we 409!
perform the largest review of CBM methods conducted to date (165 methods and 30 software 410!
packages), demonstrating that MUFINS is the second most general CBM software after COBRA 411!
toolbox (49) in terms of the number of methods implemented. However, it provides the largest 412!
number of CBM methods under GUI with interactive network visualization. Finally, all CBM 413!
methods can now be applied to models formulated with inhibitor and activator constraints, which 414!
!
18!
again enables execution of new CBM protocols without the need of coding (e.g. iMAT applied to 415!
models involving steady-state regulatory network). 416!
417!
Future directions.
418!
We will continue to develop MUFINS towards improved interoperability with other tools and
419!
model databases, a key for model integration. While currently QSSPN can be simulated only in
420!
MUFINS, definition of this multi-formalism framework in SBML will motivate development of 421!
alternative tools. As shown on Figure 1, QSSPN models can currently be exported into two separate 422!
SBML files representing the CBM and PN parts of the model. We intend to represent QSSPN 423!
lookup tables, reset transitions and flux monitors with existing SBML objects, or to develop a 424!
bespoke SBML package. Furthermore, we will work towards improving integration of SBML files 425!
imported from public repositories into multi-formalism models in the JyMet GUI. This will involve 426!
further work on network visualization in JyMet, providing a graph editor dedicated to connecting
427!
different mechanistic models by common variables. We also plan to develop interoperability 428!
between MUFINS and Garuda (http://www.garuda-alliance.org) to make full use of our multi-429!
formalism simulation tool within this established alliance of systems biology software. This will be 430!
facilitated by design of our software (Figure 1) providing stand-alone simulation engines ideal for 431!
embedding in different interfaces. 432!
433!
Conclusions.
434!
The multi-scale nature of complex biological systems is currently the major challenge preventing 435!
their computational understanding. A number of theoretical frameworks have achieved spectacular 436!
successes in mechanistically modelling different levels of cellular organisation such as metabolic, 437!
signaling and gene regulatory networks. However, in a real cell all these processes proceed 438!
simultaneously, and without multi-scale simulation the insight and predictive power provided by 439!
models will be limited. We present MUFINS, the first general software addressing this multi-440!
!
19!
formalism simulation challenge. Novel algorithms available in MUFINS provide solutions for three 441!
major technological challenges: i) integration of CBM and hybrid stochastic/deterministic dynamic 442!
simulation ii) CBM of integrated signaling/metabolic models iii) analysis of large clinical 443!
transcriptome studies in the context of GSMN. This is demonstrated through three Use Cases,
444!
where we simulate models of mammalian systems composed of: GSMNs, logical hypergraph
445!
models of signalling, kinetic models of gene regulation and PBPK models. We experimentally
446!
validate model predictions and show how our software can aid experimental scientists through an 447!
iterative cycle of hypothesis generation, experimentation and model refinement. Because the need 448!
for a multi-scale, multi-formalism approach is currently most recognised in the context of 449!
Personalised Medicine and Quantitative Systems Pharmacology, we focused our Use Cases on 450!
mammalian cells. However, mechanistic simulation is a major tool in Synthetic Biology, where 451!
MUFINS will be ideal to integrate detailed models of genetic circuits with GSMNs and further 452!
extend molecular cell factory models to include bioreactor mass transfer. Therefore, we believe that
453!
multiformalism simulation with MUFINS will find broad application in mechanistic modelling of 454!
biological systems. 455!
456!
Software Availability: 457!
MUFINS is free, open source software available under GNU GPL license from: 458!
MUFINS home page: http://sysbio3.fhms.surrey.ac.uk/mufins/
459!
GitHub repository: https://github.com/kierzek/MUFINS
460!
461!
Acknowledgements 462!
Development of MUFINS was funded by BBSRC TRDF grant BB/K015974/1 to AMK. AvK, SK, 463!
GRS and AMK were supported by EraSysBio+/BBSRC TB-HOST-NET grant BB/I00453X/1. CL, 464!
NS and DL were supported by The research leading to results obtained by CL, NS and DL has 465!
received support from the Innovative Medicines Initiative Joint Undertaking under grant agreement 466!
!
20!
n 115308, resources of which are composed of financial contribution from the European Union's 467!
Seventh Framework Programme (FP7/2007-2013) and EFPIA companies in kind contribution. MG 468!
and JS were funded by National Science Centre (Poland) grant DEC-2012/07/D/ST6/02492. VL 469!
was funded by the BBSRC grant (BB/K501694/1).
470!
471!
Author contributions.
472!
HW implemented most of the software features and formulated Fast iMAT approach. AvK and SK 473!
developed logical hypergraph model in Use Case 1. VL, LTW and NJP contributed Use Case 3. 474!
WM and HM designed and prototyped network visualization in JyMet. NS, NJP and HVW 475!
conducted study in Use Case 2. AG contributed to development of sfba and JyMet. CL, GRS and 476!
DJML provided experimental part of Use Case 1. MG and JS designed and implemented 477!
QSSPNclient. AAM contributed to development and testing of ~omics data integration approaches. 478!
NS and DJMW contributed to model integration and simulation in Use Case 1. JMcF contributed to
479!
JyMet design and network visualization. HVW contributed to manuscript design. AMK and NJP 480!
wrote manuscript. AMK formulated qsspn algorithm and contributed to qsspn solver 481!
implementation. 482!
483!
References. 484!
1.!Gillespie,!D.T.,!Exact&stochastic&simulation&of&coupled&chemical-reactions.!Journal!of!
485!
Physical!Chemistry,!1977.!81(25):!p.!2340-2361.!
486!
2.!Tyson,!J.J.,!K.!Chen,!and!B.!Novak,!Network&dynamics&and&cell&physiology.!Nature!487!
Reviews!Molecular!Cell!Biology,!2001.!2(12):!p.!908-916.!488!
3.! Bordbar,!A.,!et!al.,!Constraint-based&models&predict&metabolic&and&associated&cellular&489!
functions.!Nature!Reviews!Genetics,!2014.!15(2):!p.!107-120.!490!
4.!Orth,!J.D.,!I.!Thiele,!and!B.O.!Palsson,!What&is&flux&balance&analysis?!Nature!491!
Biotechnology,!2010.!28(3):!p.!245-248.!492!
5.!Lewis,!N.E.,!H.!Nagarajan,!and!B.O.!Palsson,!Constraining&the&metabolic&genotype-493!
phenotype&relationship&using&a&phylogeny&of&in&silico&methods.!Nature!Reviews!494!
Microbiology,!2012.!10(4):!p.!291-305.!495!
6.! Klamt,!S.,!et!al.,!A&methodology&for&the&structural&and&functional&analysis&of&signaling&and&496!
regulatory&networks.!Bmc!Bioinformatics,!2006.!7.!497!
!
21!
7.! Remy,!E.,!et!al.,!A&description&of&dynamical&graphs&associated&to&elementary&regulatory&498!
circuits.!Bioinformatics,!2003.!19:!p.!II172-II178.!499!
8.!Ruths,!D.,!et!al.,!The&Signaling&Petri&Net-Based&Simulator:&A&Non-Parametric&Strategy&for&500!
Characterizing&the&Dynamics&of&Cell-Specific&Signaling&Networks.!Plos!Computational!
501!
Biology,!2008.!4(2).!
502!
9.!Fisher,!C.P.,!et!al.,!QSSPN:&Dynamic&Simulation&of&Molecular&Interaction&Networks&
503!
Describing&Gene&Regulation,&Signalling&and&Whole-Cell&Metabolism&in&Human&Cells.!504!
Bioinformatics,!2013.!29(24):!p.!3181-3190.!505!
10.!Hucka,!M.,!et!al.,!The&systems&biology&markup&language&(SBML):&a&medium&for&506!
representation&and&exchange&of&biochemical&network&models.!Bioinformatics,!2003.!507!
19(4):!p.!524-531.!508!
11.!Chelliah,!V.,!et!al.,!BioModels:&ten-year&anniversary.!Nucleic!Acids!Research,!2015.!509!
43(D1):!p.!D542-D548.!
510!
12.!Jones,!H.M.!and!K.!Rowland-Yeo,!Basic&concepts&in&physiologically&based&511!
pharmacokinetic&modeling&in&drug&discovery&and&development.!CPT:!pharmacometrics!&!512!
systems!pharmacology,!2013.!2:!p.!e63-e63.!513!
13.!Karr,!J.R.,!et!al.,!A&Whole-Cell&Computational&Model&Predicts&Phenotype&from&Genotype.!514!
Cell,!2012.!150(2):!p.!389-401.!515!
14.!Krauss,!M.,!et!al.,!Integrating&Cellular&Metabolism&into&a&Multiscale&Whole-Body&Model.!
516!
PloS!Computational!Biology,!2012.!8(10).!
517!
15.! Ruths,!D.,!L.!Nakhleh,!and!P.T.!Ram,!Rapidly&exploring&structural&and&dynamic&properties&518!
of&signaling&networks&using&PathwayOracle.!Bmc!Systems!Biology,!2008.!2.!519!
16.!Covert,!M.W.,!et!al.,!Integrating&metabolic,&transcriptional&regulatory&and&signal&520!
transduction&models&in&Escherichia&coli.!Bioinformatics,!2008.!24(18):!p.!2044-2050.!521!
17.!Covert,!M.W.,!C.H.!Schilling,!and!B.!Palsson,!Regulation&of&gene&expression&in&flux&balance&522!
models&of&metabolism.!Journal!of!Theoretical!Biology,!2001.!213(1):!p.!73-88.!523!
18.! Puchalka,!J.!and!A.M.!Kierzek,!Bridging&the&gap&between&stochastic&and&deterministic&524!
regimes&in&the&kinetic&simulations&of&the&biochemical&reaction&networks.!Biophysical!525!
Journal,!2004.!86(3):!p.!1357-1372.!526!
19.! Gevorgyan,!A.,!et!al.,!SurreyFBA:&a&command&line&tool&and&graphics&user&interface&for&527!
constraint-based&modeling&of&genome-scale&metabolic&reaction&networks.!Bioinformatics,!528!
2011.!27(3):!p.!433-434.!529!
20.! Becker,!S.A.!and!B.O.!Palsson,!Context-specific&metabolic&networks&are&consistent&with&530!
experiments.!Plos!Computational!Biology,!2008.!4(5).!531!
21.! Schmidt,!B.J.,!et!al.,!GIM(3)E:&condition-specific&models&of&cellular&metabolism&developed&532!
from&metabolomics&and&expression&data.!Bioinformatics,!2013.!29(22):!p.!2900-2908.!533!
22.! Diamant,!I.,!et!al.,!A&network-based&method&for&predicting&gene-nutrient&interactions&and&534!
its&application&to&yeast&amino-acid&metabolism.!Molecular!Biosystems,!2009.!5(12):!p.!535!
1732-1739.!536!
23.! Mendum,!T.A.,!et!al.,!Lipid&metabolism&and&Type&VII&secretion&systems&dominate&the&537!
genome&scale&virulence&profile&of&Mycobacterium&tuberculosis&in&human&dendritic&cells.!538!
Bmc!Genomics,!2015.!16.!539!
24.!Breitling,!R.,!et!al.,!A&structured&approach&for&the&engineering&of&biochemical&network&540!
models,&illustrated&for&signalling&pathways.!Briefings!in!Bioinformatics,!2008.!9(5):!p.!541!
404-421.!542!
!
22!
25.!Rohr,!C.,!W.!Marwan,!and!M.!Heiner,!Snoopy-a&unifying&Petri&net&framework&to&543!
investigate&biomolecular&networks.!Bioinformatics,!2010.!26(7):!p.!974-975.!544!
26.! Mendum,!T.A.,!et!al.,!Interrogation&of&global&mutagenesis&data&with&a&genome&scale&545!
model&of&Neisseria&meningitidis&to&assess&gene&fitness&in&vitro&and&in&sera.!Genome!
546!
Biology,!2011.!12(12).!
547!
27.!Beste,!D.J.V.,!et!al.,!GSMN-TB:&a&web-based&genome&scale&network&model&of&
548!
Mycobacterium&tuberculosis&metabolism.!Genome!Biology,!2007.!8(5).!549!
28.!Cottret,!L.,!et!al.,!MetExplore:&a&web&server&to&link&metabolomic&experiments&and&genome-550!
scale&metabolic&networks.!Nucleic!Acids!Research,!2010.!38:!p.!W132-W137.!551!
29.!Leoncikas,!V.,!et!al.,!Generation&of&2,000&breast&cancer&metabolic&landscapes&reveals&a&552!
poor&prognosis&group&with&active&serotonin&production.!Scientific!Reports,!2016.!6:!p.!553!
19771.!554!
30.! Bordbar,!A.,!et!al.,!Model-driven&multi-omic&data&analysis&elucidates&metabolic&
555!
immunomodulators&of&macrophage&activation.!Molecular!Systems!Biology,!2012.!8.!556!
31.! Dasika,!M.S.,!A.!Burgard,!and!C.D.!Maranas,!A&computational&framework&for&the&557!
topological&analysis&and&targeted&disruption&of&signal&transduction&networks.!Biophysical!558!
Journal,!2006.!91(1):!p.!382-398.!559!
32.!Vardi,!L.,!E.!Ruppin,!and!R.!Sharan,!A&Linearized&Constraint-Based&Approach&for&560!
Modeling&Signaling&Networks.!Journal!of!Computational!Biology,!2012.!19(2):!p.!232-
561!
240.!
562!
33.! Shlomi,!T.,!et!al.,!A&genome-scale&computational&study&of&the&interplay&between&563!
transcriptional&regulation&and&metabolism.!Molecular!Systems!Biology,!2007.!3.!564!
34.! Kolodkin,!A.,!et!al.,!Optimization&of&stress&response&through&the&nuclear&receptor-565!
mediated&cortisol&signalling&network.!Nature!Communications,!2013.!4:!p.!1972.!566!
35.! Kolodkin,!A.N.,!et!al.,!Design&principles&of&nuclear&receptor&signaling:&how&complex&567!
networking&improves&signal&transduction.!Molecular!Systems!Biology,!2010.!6:!p.!446.!568!
36.!Le!Novere,!N.,!et!al.,!The&Systems&Biology&Graphical&Notation.!Nature!Biotechnology,!569!
2009.!27(8):!p.!735-741.!570!
37.!Thiele,!I.,!et!al.,!A&community-driven&global&reconstruction&of&human&metabolism.!Nature!571!
Biotechnology,!2013.!31(5):!p.!419.!572!
38.!Gille,!C.,!et!al.,!HepatoNet1:&a&comprehensive&metabolic&reconstruction&of&the&human&573!
hepatocyte&for&the&analysis&of&liver&physiology.!Molecular!Systems!Biology,!2010.!6:!p.!574!
411.!575!
39.!Kolker,!E.,!et!al.,!MOPED:&Model&Organism&Protein&Expression&Database.!Nucleic!Acids!576!
Research,!2012.!40(D1):!p.!D1093-D1099.!577!
40.!El-Sankary,!W.,!N.!Plant,!and!G.!Gibson,!Regulation&of&the&CYP3A4&gene&by&578!
hydrocortisone&and&xenobiotics:&role&of&the&glucocorticoid&and&pregnane&X&receptors.!579!
Drug!Metabolism!and!Disposition,!2000.!28(5):!p.!493-496.!580!
41.!Plant,!N.,!The&human&cytochrome&P450&3A&sub-family:&transcriptional&regulation,&inter-581!
individual&variation&and&interaction&networks.!Biochimica!et!Biophysica!Acta-General!582!
Subjects,!2007.!1770(3):!p.!478-488.!.!583!
42.!Losordo,!D.W.!and!J.M.!Isner,!Estrogen&and&angiogenesis&-&A&review.!Arteriosclerosis!584!
Thrombosis!and!Vascular!Biology,!2001.!21(1):!p.!6-12.!585!
!
23!
43.!Zheng,!J.,!et!al.,!Estrogen&and&progesterone&receptors,&cell&proliferation,&and&c-fos&586!
expression&in&the&ovine&uterus&during&early&pregnancy.!Endocrinology,!1996.!137(1):!p.!587!
340-348.!588!
44.!Kuhl,!H.,!Pharmacology&of&estrogens&and&progestogens:&influence&of&different&routes&of&
589!
administration.!Climacteric,!2005.!8:!p.!3-63.!
590!
45.!Plant,!N.!and!G.G.!Gibson,!Evaluation&of&the&toxicological&relevance&of&CYP3A4&induction.!
591!
Current!Opinion!in!Drug!Discovery!and!Development,!2003.!6(1):!p.!50-56.!592!
46.!Curtis,!C.,!et!al.,!The&genomic&and&transcriptomic&architecture&of&2,000&breast&tumours&593!
reveals&novel&subgroups.!Nature,!2012.!486(7403):!p.!346-352.!594!
47.!DiNatale,!B.C.,!et!al.,!Kynurenic&Acid&Is&a&Potent&Endogenous&Aryl&Hydrocarbon&Receptor&595!
Ligand&that&Synergistically&Induces&Interleukin-6&in&the&Presence&of&Inflammatory&596!
Signaling.!Toxicological!Sciences,!2010.!115(1):!p.!89-97.!597!
48.! Opitz,!C.A.,!et!al.,!An&endogenous&tumour-promoting&ligand&of&the&human&aryl&
598!
hydrocarbon&receptor.!Nature,!2011.!478(7368):!p.!197-203.!599!
49.!Hoops,!S.,!et!al.,!COPASI&—&a&COmplex&PAthway&SImulator.!Bioinformatics,!2006.!22:!p.!600!
3067-3074.!601!
!602!
!603!
!
604!
! !
605!
!
24!
+606!
Figure+1.+Overview+of+MUFINS.!All!calculations!are!performed!by!two!computational!607!
engines,!which!can!be!also!run!as!stand-alone!command!line!tools.!The!sfba!implements!CBM!608!
methods!and!qsspn!performs!QSSPN!simulations.!JyMet!is!a!graphic!interface!to!all!methods!
609!
providing!spreadsheet!representation!of!models!and!results!as!well!as!metabolic!network!
610!
visualization!and!plots.!JyMet!writes!input!files!for!computational!engines,!starts!calculations,!
611!
imports!output!files!and!displays!results.!In!the!case!of!QSSPN!simulations,!Petri!Net!612!
connectivity!can!be!graphically!edited!by!Snoopy!software,!a!standard!Petri!Net!tool,!which!613!
we!use!as!external!editor.!JyMet!imports!Snoopy!files!and!provides!spreadsheet!interface!614!
allowing!editing!of!QSSPN!parameters!or!independent!creation!of!entire!QSSPN!model.!615!
Conversion!of!Snoopy!files!directly!to!qsspn!engine!is!also!possible!with!command!line!python!616!
script!spept2qsspn. Both!JyMet!and!Snoopy!import!and!export!SBML!file!providing!617!
connectivity!to!other!SBML-compliant!tools.!The!file!formats!used!for!software!component!
618!
communication!are!indicated!by!their!default!extensions!and!described!in!Supplementary!File!619!
Formats.!620!
!
25!
!621!
Figure+2.+The+model+of+cell+signaling,+gene+regulation+and+whole-cell+metabolism+in+622!
RAW264.7+macrophage.+A+signaling!and!gene!regulatory!network!of!286!interactions!623!
between!205!species,!created!in!logical!hypergraph!formalism!is!shown.!This!network!was!
624!
subsequently!converted!to!FBA!formalism!with!linear!inhibitory!constraints!and!coupled!to!
625!
the!RAW264.7!GSMN!through!regulation!of!the!iNOS!gene.!Nitric!oxide!synthesis,!a!major!
626!
metabolic!flux!in!RAW264.7!macrophages!responding!to!a!pathogen,!was!then!simulated!627!
using!constraints!derived!from!both!stoichiometry!of!whole-cell!metabolism!and!logical!rules!628!
within!a!large-scale!regulatory!network.!The!inset!shows!the!conversion!of!logical!hyperedges!629!
determining!the!fate!of!ifn_ab!to!reaction!formulas!with!linear!inhibitor!constraint:!For!all!630!
reactions!producing!ifn_ab,!the!molecule!irf2!is!added,!preceded!by!the!“~”!sign!to!indicate!an!631!
inhibitor.!This!is!parsed!by!MUFINS!to!mean!that!the!reaction!flux!is!inhibited!(i.e.!0)!if!ifr2!is!632!
present.!!
633!
!
26!
!634!
Figure+3.+Mechanistic+interpretation+of+experimental+data+on+perturbation+of+whole-cell+635!
metabolic+function+by+signaling+network+input+and+inhibitor.!MUFINS!was!used!to!636!
integrate!a!genome-scale!metabolic!model!of!the!mouse!macrophage!(RAW264.7)!with!a!
637!
large-scale!regulatory!network.!Perturbation!of!whole!cell!metabolism!was!simulated!through!
638!
activation!and!inhibition!of!the!signaling!network!with!external!production!of!nitric!oxide!set!
639!
as!the!objective!function.!Predicted!data!was!then!compared!to!experimental!data.!A)!The!left!640!
panel!shows!a!screenshot!of!the!JyMet!interface,!demonstrating!on!screen!visualization!of!the!641!
reconstruction,!created!by!automatic!hierarchical!layout!with!manual!adjustment.!Hatched!642!
lines!are!used!to!indicate!regulatory!signals,!representing!inhibition!(circle!end)!or!643!
stimulation!(arrow!head).!The!right!panel!is!a!manually!created!image!representing!the!644!
pathway!examined!through!JyMet;!arrows!represent!signal!flux,!while!open!and!filled!circles!645!
!
27!
represent!inhibition!and!stimulation,!respectively.!The!visualization!depicts!where!signaling!646!
pathways!converge!on!the!iNOS!gene,!which!is!required!for!nitric!oxide!(NO)!production!in!the!647!
whole-cell!stoichiometric!model.!Flux!rates!for!an!example!FBA!solution!are!displayed!on!the!648!
network!diagram;!on!the!right!panel!only!flux!rates!for!each!transitions!are!presented!for!
649!
clarity,!while!the!left!panel!also!shows!the!contribution!of!each!substance!to!the!flux.!B)!The!
650!
original!reconstruction!was!able!to!predict!the!increase!in!NO!production!following!
651!
stimulation!with!LPS,!but!not!the!impact!of!a!MEK!inhibitor,!when!compared!to!experimental!652!
data!of!nitrate!levels!in!RAW264.7!cell-conditioned!medium!(C).!Nitrate!can!only!be!produced!653!
by!non-enzymatic!conversion!of!NO!produced!by!RAW264.7!cells,!and!as!there!is!no!nitrate!654!
consumption!in!the!medium,!nitrate!concentrations!are!proportional!to!nitric!oxide!655!
production!flux.!D)!Refinement!of!the!signaling!network!led!to!agreement!between!in!silico!656!
prediction!and!in!vitro!measurement.!The!refinement!was!based!upon!three!mechanistic!657!
hypotheses:!i)!ERK1/2!is!a!more!potent!transcriptional!activator!of!the!iNOS!gene!than!JNK!
658!
and!HIF1,!and!ii)!MEK1!is!a!more!potent!ERK1/2!kinase!than!PKC.!659!
! !660!
!
28!
!661!
Figure+4.+Multi-formalism+simulation+integrating+cortisol+signaling+with+the+human+662!
Recon2+GSMN+reveals+a+drug+interaction+with+estradiol+clearance.+A)!The!Petri!Net!663!
diagram!of!network!connectivity!created!in!the!Snoopy!editor,!with!overlaid!comments!for!
664!
clarity.!Color!and!symbol!size!has!been!manually!set!to!match!SBGN!molecule!types!and!
665!
transition!types!specific!to!QSSPN.!The!PN!connectivity!to!implement!a!timer!for!
666!
administering!a!network!perturbation!(cortisol!burst),!is!contained!within!a!coarse!transition!667!
!
29!
and!shown!as!an!insert.!!B)!Simulation!of!glucose!and!lactate!dynamics!in!the!blood!668!
physiological!compartment,!demonstrating!a!convergence!to!physiologically!realistic!steady!669!
states.!Perturbation!of!the!system!through!a!simulated!cortisol!infusion!starting!after!500mins!670!
elicits!a!dynamic!alteration!in!the!signaling!network,!resulting!in!(C)!a!predicted!increase!in!
671!
CYP34A!protein!levels,!which!is!confirmed!in!primary!human!hepatocytes.!The!increased!
672!
expression!of!CYP3A4!protein!is!predicted!to!increase!flux!through!reactions!catalyzed!by!this!
673!
enzyme,!leading!to:!(D)!degradation!of!excess!cortisol!and!establishment!of!new!steady!state;!674!
(E)!a!drug-drug!interaction!for!a!second!CYP3A4!substrate!(estradiol),!contained!within!the!675!
GSMN,!leading!to!a!decrease!in!it’s!steady!state!level.!The!predicted!increase!in!CYP3A4!676!
activity!following!cortisol!exposure!is!confirmed!in!primary!human!hepatocytes!(F),!as!is!the!677!
enhanced!rate!of!estradiol!clearance!(G)!!678!
!679!
!
680!
!681!
!682!

Supplementary resources (12)

... Interaction Network Simulator (MUFINS) (Wu et al., 2016). ...
... This results in alterations in the encoded protein level (CYP3A4), which in turn informs the upper and lower bounds within the GSMN for any reactions catalysed by that protein. Fluxes are extracted from FBA sample solution and employed to update the status of the regulatory network, effecting the cycle of stimulus -response-return to baseline (Wu et al., 2016). ...
... As discussed in the previous section, a variety of theoretical techniques have obtained significant results in mechanistically modelling various levels of cellular systems, such as metabolic signalling, including gene regulatory networks (Wu et al., 2016). In a real cell, all these methods progress concurrently, and without multi-scale simulation, the awareness and predictive strength delivered by models will be restricted (Wu et al., 2016). ...
Article
Background: Drug-induced liver injury (DILI) is rare but potentially lethal, and it can cause liver disease attributable to all types of drugs. The adverse impact of most of the DILI incidents is mild, and it recovers after the removal of drugs. The harmful agents should be identified and removed as early as possible to avoid the development of chronic liver damage. Troglitazone (TGZ) was a derivative of a thiazolidinedione drug produced for the treatment of type 2 diabetes, in the late 1990s. However, it was withdrawn from the market due to reported cases of liver toxicities. Several molecular mechanisms have been proposed to underlie TGZ-induced liver toxicity. Understanding the interactions between these mechanisms could aid drug developers in predicting DILI more vigorously. Aim: This thesis is aimed at using a combination of in silico and in vitro approaches to evaluate the interaction of TGZ with multiple biological systems and predict the emergent biological pathways in TGZ-induced liver toxicity. Method: To evaluate TGZ toxicity pathways, a model was constructed using Petri net software termed "SNOOPY" to reconstruct the putative cellular effects of TGZ, including activation of PPARy, interaction with mitochondria, and activation of apoptosis. The model was imported into the MUFINS software suite and simulated. Activation of apoptosis was validated against the published systems biology markup language (SBML) model downloaded from BIOMODELS, upon which the model was created. The effects of TGZ on cellular activities were determined through an in vitro approach. Results: The model created in SNOOPY and simulated in MUFINS could reproduce the behaviour of the original submission of BIOMODELS simulated in COPASI, validating the reconstruction. Other possible TGZ toxicity pathways have been predicted. TGZ-induced apoptosis is done through the activation of caspase 3/7 and 9, in a concentration-dependent manner. Also, a dose-dependent decrease in cellular processes has been recorded. However, caspase-8 activation in TGZ-treated cells has not been recorded. Conclusion: These data support the activation of apoptosis via the intrinsic route. The in sillico model reproduces the original model, and it can therefore be used to predict TGZ induced-liver toxicity. The in vitro assays were useful tools to elucidate TGZ-induced toxicity, making it a suitable model for this study.
... Thus, these models are heavily parameterized and require extensive sets of kinetic information. Several toolboxes for modeling PBPK models which already contain collections of the necessary physiological parameters exist such as MUFINS [105] and the commercial tools PKSim Ò and MoBi Ò (part of the Computational Systems Biology Software Suite of Bayer Technology Services GmbH (Leverkusen, Germany)). On the contrary, FBA models rely only on the reaction stoichiometries of the underlying metabolic network and allow simulating metabolism at a large scale. ...
... Furthermore, examining the simulated trajectories identified the coupling of the chenodiol and cholate branch of the bile acid synthesis pathway through the regulatory network. The QSSPN algorithm is part of the SurreyFBA software [145], which has been integrated with MUFINS, a multi-formalism interaction network simulator [105]. ...
Article
Full-text available
The availability of multi-omics data sets and genome-scale metabolic models for various organisms provide a platform for modeling and analyzing genotype-to-phenotype relationships. Flux balance analysis is the main tool for predicting flux distributions in genome-scale metabolic models and various data-integrative approaches enable modeling context-specific network behavior. Due to its linear nature, this optimization framework is readily scalable to multi-tissue or -organ and even multi-organism models. However, both data and model size can hamper a straightforward biological interpretation of the estimated fluxes. Moreover, flux balance analysis simulates metabolism at steady-state and thus, in its most basic form, does not consider kinetics or regulatory events. The integration of flux balance analysis with complementary data analysis and modeling techniques offers the potential to overcome these challenges. In particular machine learning approaches have emerged as the tool of choice for data reduction and selection of most important variables in big data sets. Kinetic models and formal languages can be used to simulate dynamic behavior. This review article provides an overview of integrative studies that combine flux balance analysis with machine learning approaches, kinetic models, such as physiology-based pharmacokinetic models, and formal graphical modeling languages, such as Petri nets. We discuss the mathematical aspects and biological applications of these integrated approaches and outline challenges and future perspectives.
... Later, MUFINS [74] was presented extending the QSSPN method and adding a GUI, which facilitates the construction and analysis of hybrid FBA models. MUFINS supports the integration of stochastic simulation, deterministic ODE simulation, parameter-free simulation [75] and FBA in a single software platform with GUI, offering multiformalism functionality for modelling multiscale biological systems. ...
Article
Full-text available
Integrated modelling of biological systems is becoming a necessity for constructing models containing the major biochemical processes of such systems in order to obtain a holistic understanding of their dynamics and to elucidate emergent behaviours. Hybrid modelling methods are crucial to achieve integrated modelling of biological systems. This paper reviews currently popular hybrid modelling methods, developed for systems biology, mainly revealing why they are proposed, how they are formed from single modelling formalisms and how to simulate them. By doing this, we identify future research requirements regarding hybrid approaches for further promoting integrated modelling of biological systems.
... • Restricciones de regulación: estas restricciones están ligadas con la expresión génica que depende principalmente del entorno de crecimiento (condiciones medioambientales) del microrganismo. La regulación de la expresión génica podría desencadenar en la expresión o represión de síntesis de enzimas, lo cual conlleva a la activación o inactivación de una reacción en la red metabólica (26) . ...
Article
Full-text available
El metabolismo representa el nivel biológico que más se relaciona con los fenotipos de la célula y, las alteraciones o reprogramaciones de éste pueden, entre otras, (i) afectar la producción de metabolitos primarios o secundarios en microorganismos de interés biotecnológico, (ii) favorecer o no la inhibición del crecimiento en organismos patógenos y (iii) desarrollar desórdenes metabólicos como la obesidad o la diabetes. Es por ello, que el estudio del metabolismo, el rediseño, y el redireccionamiento de fluxes metabólicos se ha convertido en un área importante de investigación (también conocida como Ingeniería Metabólica), ya que ha permitido el desarrollo y diseño de procesos biológicos mejorados, la identificación de blancos terapéuticos, el diseño de estrategias terapéuticas para curar desordenes metabólicos y la identificación de biomarcadores en cáncer, entre otros. Actualmente, se han desarrollado metodologías computacionales que permiten estudiar el metabolismo celular a diferentes condiciones medioambientales, dirigiendo la experimentación con las predicciones del modelo. El propósito de esta revisión es resaltar la importancia del análisis de fluxes metabólicos como una metodología general para estudiar la reprogramación metabólica en distintos organismos de interés biotecnológico, médico, y terapéutico. Este trabajo condensa las bases teóricas y los conceptos claves para entender el análisis de fluxes metabólicos, lo cual será un insumo fundamental para aquellos que se están adentrando al mundo de la biología de sistemas o áreas afines.
... Several other more recent platforms have been developed; see, e.g., [6,[10][11][12][13] for a partial list. Some of these can deal only with Boolean networks, such as BoolNet, while others can handle multi-state models, such as GinSim. ...
Article
Full-text available
Background: At the molecular level, nonlinear networks of heterogeneous molecules control many biological processes, so that systems biology provides a valuable approach in this field, building on the integration of experimental biology with mathematical modeling. One of the biggest challenges to making this integration a reality is that many life scientists do not possess the mathematical expertise needed to build and manipulate mathematical models well enough to use them as tools for hypothesis generation. Available modeling software packages often assume some modeling expertise. There is a need for software tools that are easy to use and intuitive for experimentalists. Results: This paper introduces PlantSimLab, a web-based application developed to allow plant biologists to construct dynamic mathematical models of molecular networks, interrogate them in a manner similar to what is done in the laboratory, and use them as a tool for biological hypothesis generation. It is designed to be used by experimentalists, without direct assistance from mathematical modelers. Conclusions: Mathematical modeling techniques are a useful tool for analyzing complex biological systems, and there is a need for accessible, efficient analysis tools within the biological community. PlantSimLab enables users to build, validate, and use intuitive qualitative dynamic computer models, with a graphical user interface that does not require mathematical modeling expertise. It makes analysis of complex models accessible to a larger community, as it is platform-independent and does not require extensive mathematical expertise.
... To predict the activity of metabolic reactions within the human system the 'Fast iMAT' algorithm of MUFINS 56 , a variant of the iMAT algorithm 57,58 , was used. Under a constraint-based modelling framework, iMAT approaches aim to maximise the congruency between functional -omic data and the activity of metabolic reactions in a metabolic model. ...
Article
Full-text available
Studying circadian rhythms in most human tissues is hampered by difficulty in collecting serial samples. Here we reveal circadian rhythms in the transcriptome and metabolic pathways of human white adipose tissue. Subcutaneous adipose tissue was taken from seven healthy males under highly controlled ‘constant routine’ conditions. Five biopsies per participant were taken at six-hourly intervals for microarray analysis and in silico integrative metabolic modelling. We identified 837 transcripts exhibiting circadian expression profiles (2% of 41619 transcript targeting probes on the array), with clear separation of transcripts peaking in the morning (258 probes) and evening (579 probes). There was only partial overlap of our rhythmic transcripts with published animal adipose and human blood transcriptome data. Morning-peaking transcripts associated with regulation of gene expression, nitrogen compound metabolism, and nucleic acid biology; evening-peaking transcripts associated with organic acid metabolism, cofactor metabolism and redox activity. In silico pathway analysis further indicated circadian regulation of lipid and nucleic acid metabolism; it also predicted circadian variation in key metabolic pathways such as the citric acid cycle and branched chain amino acid degradation. In summary, in vivo circadian rhythms exist in multiple adipose metabolic pathways, including those involved in lipid metabolism, and core aspects of cellular biochemistry.
... Several methods have been developed to predict behaviour over time [dynamic FBA (Varma and Palsson, 1994) and dynamic FVA (Maldonado et al., 2018)], integrate regulation [regulatory FBA (Covert et al., 2001)] and integrate other data types [integrated FBA (Covert et al., 2008) and integrated dynamic FBA (Lee et al., 2008)]. Furthermore, other CBM methods and simulators have been developed to expand their applications, e.g., multi-objective function analyses (Costanza et al., 2012;Zakrzewski et al., 2012), whole human cell metabolic analyses (Fisher et al., 2013), integrate multiple simulation formats (Liao et al., 2012;Wu et al., 2016;Heirendt et al., 2017), of which all could be adapted for use in future primary mitochondrial research. ...
Article
Full-text available
Primary mitochondrial diseases form one of the most common and severe groups of genetic disease, with a birth prevalence of at least 1 in 5000. These disorders are multi-genic and multi-phenotypic (even within the same gene defect) and span the entire age range from prenatal to late adult onset. Mitochondrial disease typically affects one or multiple high-energy demanding organs, and is frequently fatal in early life. Unfortunately, to date there are no known curative therapies, mostly owing to the rarity and heterogeneity of individual mitochondrial diseases, leading to diagnostic odysseys and difficulties in clinical trial design. This review aims to discuss recent advances and challenges of systems approaches for the study of primary mitochondrial diseases. Although there has been an explosion in the generation of omics data, few studies have progressed toward the integration of multiple levels of omics. It is evident that the integration of different types of data to create a more complete representation of biology remains challenging, perhaps due to the scarcity of available integrative tools and the complexity inherent in their use. In addition, “bottom-up” systems approaches have been adopted for use in the iterative cycle of systems biology: from data generation to model prediction and validation. Primary mitochondrial diseases, owing to their complex nature, will most likely benefit from a multidisciplinary approach encompassing clinical, molecular and computational studies integrated together by systems biology to elucidate underlying pathomechanisms for better diagnostics and therapeutic discovery. Just as next generation sequencing has rapidly increased diagnostic rates from approximately 5% up to 60% over two decades, more recent advancing technologies are encouraging; the generation of multi-omics, the integration of multiple types of data, and the ability to predict perturbations will, ultimately, be translated into improved patient care.
Article
Biological systems inherently span multiple levels, which can pose challenges in spatial representation for modelers. We present a protocol that utilizes colored Petri nets to construct and analyze biological models of systems, encompassing both unilevel and multilevel scenarios. We detail a modeling workflow exploiting the PetriNuts platform comprising a set of tools linked together via common file formats. We describe steps for modeling preparation, component-level modeling and analysis, followed by system-level modeling and analysis, and model use.
Article
Full-text available
Motivation: While there are software packages that analyze Boolean, ternary, or other multi-state models, none compute the complete state space of function-based models over any finite set. Results: We propose Cyclone, a simple light-weight software package which simulates the complete state space for a finite dynamical system over any finite set. Availability: Source code is freely available at https://github.com/discretedynamics/cyclone under the Apache-2.0 license.
Article
Temperature affects cellular processes at different spatiotemporal scales, and identifying the genetic and molecular mechanisms underlying temperature responses paves the way to develop approaches for mitigating the effects of future climate scenarios. A systems view of the effects of temperature on cellular physiology can be obtained by focusing on metabolism since: (i) its functions depend on transcription and translation and (ii) its outcomes support organisms' development, growth, and reproduction. Here we provide a systematic review of modelling efforts directed at investigating temperature effects on properties of single biochemical reactions, system-level traits, metabolic subsystems, and whole-cell metabolism across different prokaryotes and eukaryotes. We compare and contrast computational approaches and theories that facilitate modelling of temperature effects on key properties of enzymes and their consideration in constraint-based as well as kinetic models of metabolism. In addition, we provide a summary of insights from computational approaches, facilitating integration of omics data from temperature-modulated experiments with models of metabolic networks, and review the resulting biotechnological applications. Lastly, we provide a perspective on how different types of metabolic modelling can profit from developments in machine learning and models of different cellular layers to improve model-driven insights into the effects of temperature relevant for biotechnological applications.
Article
Full-text available
A major roadblock in the effective treatment of cancers is their heterogeneity, whereby multiple molecular landscapes are classified as a single disease. To explore the contribution of cellular metabolism to cancer heterogeneity, we analyse the Metabric dataset, a landmark genomic and transcriptomic study of 2,000 individual breast tumours, in the context of the human genome-scale metabolic network. We create personalized metabolic landscapes for each tumour by exploring sets of active reactions that satisfy constraints derived from human biochemistry and maximize congruency with the Metabric transcriptome data. Classification of the personalized landscapes derived from 997 tumour samples within the Metabric discovery dataset reveals a novel poor prognosis cluster, reproducible in the 995-sample validation dataset. We experimentally follow mechanistic hypotheses resulting from the computational study and establish that active serotonin production is a major metabolic feature of the poor prognosis group. These data support the reconsideration of concomitant serotonin-specific uptake inhibitors treatment during breast cancer chemotherapy.
Article
Full-text available
Background Mycobacterium tuberculosis continues to kill more people than any other bacterium. Although its archetypal host cell is the macrophage, it also enters, and survives within, dendritic cells (DCs). By modulating the behaviour of the DC, M. tuberculosis is able to manipulate the host’s immune response and establish an infection. To identify the M. tuberculosis genes required for survival within DCs we infected primary human DCs with an M. tuberculosis transposon library and identified mutations with a reduced ability to survive. Results Parallel sequencing of the transposon inserts of the surviving mutants identified a large number of genes as being required for optimal intracellular fitness in DCs. Loci whose mutation attenuated intracellular survival included those involved in synthesising cell wall lipids, not only the well-established virulence factors, pDIM and cord factor, but also sulfolipids and PGL, which have not previously been identified as having a direct virulence role in cells. Other attenuated loci included the secretion systems ESX-1, ESX-2 and ESX-4, alongside many PPE genes, implicating a role for ESX-5. In contrast the canonical ESAT-6 family of ESX substrates did not have intra-DC fitness costs suggesting an alternative ESX-1 associated virulence mechanism. With the aid of a gene-nutrient interaction model, metabolic processes such as cholesterol side chain catabolism, nitrate reductase and cysteine-methionine metabolism were also identified as important for survival in DCs. Conclusion We conclude that many of the virulence factors required for survival in DC are shared with macrophages, but that survival in DCs also requires several additional functions, such as cysteine-methionine metabolism, PGLs, sulfolipids, ESX systems and PPE genes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1569-2) contains supplementary material, which is available to authorized users.
Article
Full-text available
BioModels (http://www.ebi.ac.uk/biomodels/) is a repository of mathematical models of biological processes. A large set of models is curated to verify both correspondence to the biological process that the model seeks to represent, and reproducibility of the simulation results as described in the corresponding peer-reviewed publication. Many models submitted to the database are annotated, cross-referencing its components to external resources such as database records, and terms from controlled vocabularies and ontologies. BioModels comprises two main branches: one is composed of models derived from literature, while the second is generated through automated processes. BioModels currently hosts over 1200 models derived directly from the literature, as well as in excess of 140 000 models automatically generated from pathway resources. This represents an approximate 60-fold growth for literature-based model numbers alone, since BioModels’ first release a decade ago. This article describes updates to the resource over this period, which include changes to the user interface, the annotation profiles of models in the curation pipeline, major infrastructure changes, ability to perform online simulations and the availability of model content in Linked Data form. We also outline planned improvements to cope with a diverse array of new challenges.
Article
The elucidation of breast cancer subgroups and their molecular drivers requires integrated views of the genome and transcriptome from representative numbers of patients. We present an integrated analysis of copy number and gene expression in a discovery and validation set of 997 and 995 primary breast tumours, respectively, with long-term clinical follow-up. Inherited variants (copy number variants and single nucleotide polymorphisms) and acquired somatic copy number aberrations (CNAs) were associated with expression in ~40% of genes, with the landscape dominated by cis- and trans-acting CNAs. By delineating expression outlier genes driven in cis by CNAs, we identified putative cancer genes, including deletions in PPP2R2A, MTAP and MAP2K4. Unsupervised analysis of paired DNA--RNA profiles revealed novel subgroups with distinct clinical outcomes, which reproduced in the validation cohort. These include a high-risk, oestrogen-receptor-positive 11q13/14 cis-acting subgroup and a favourable prognosis subgroup devoid of CNAs. Trans-acting aberration hotspots were found to modulate subgroup-specific gene networks, including a TCR deletion-mediated adaptive immune response in the ‘CNA-devoid’ subgroup and a basal-specific chromosome 5 deletion-associated mitotic network. Our results provide a novel molecular stratification of the breast cancer population, derived from the impact of somatic CNAs on the transcriptome.
Article
To evaluate uterine growth during pregnancy and the potential roles of estrogen and progesterone in regulating uterine cell proliferation and c-fos expression, ewes were assigned randomly to slaughter on day 12 after estrus (nonpregnant, NP), and on days 12, 18, 24, or 30 after mating (pregnant, P) in Exp 1 (n = 7 ewes/day) and on days 12 or 14 after estrus and on days 12, 14, 16, 18, 21, or 24 after mating in Exp 2 (n = 3-6 ewes/day). In Exp 1, endometrial expression of c-fos mRNA was evaluated, and labeling index was determined both in vitro (incorporation of 3H-thymidine) and in vivo (iv injection of bromodeoyxuridine [BrdU], a thymidine analog). Endometrial expression of the c-fos proto-oncogene was increased by approximately 10-fold on days 18, 24, and 30 P compared with day 12 NP or P. Labeling index (proportion of cells incorporating 3H-thymidine or BrdU, which provides an index of the rate of cell proliferation) of endometrial caruncular and intercaruncular tissues was low for day 12 NP or P, incr...
Article
Multiple models of human metabolism have been reconstructed, but each represents only a subset of our knowledge. Here we describe Recon 2, a community-driven, consensus 'metabolic reconstruction', which is the most comprehensive representation of human metabolism that is applicable to computational modeling. Compared with its predecessors, the reconstruction has improved topological and functional features, including ~2× more reactions and ~1.7× more unique metabolites. Using Recon 2 we predicted changes in metabolite biomarkers for 49 inborn errors of metabolism with 77% accuracy when compared to experimental data. Mapping metabolomic data and drug information onto Recon 2 demonstrates its potential for integrating and analyzing diverse data types. Using protein expression data, we automatically generated a compendium of 65 cell type–specific models, providing a basis for manual curation or investigation of cell-specific metabolic properties. Recon 2 will facilitate many future biomedical studies and is freely available at http://humanmetabolism.org/.
Article
The prediction of cellular function from a genotype is a fundamental goal in biology. For metabolism, constraint-based modelling methods systematize biochemical, genetic and genomic knowledge into a mathematical framework that enables a mechanistic description of metabolic physiology. The use of constraint-based approaches has evolved over ~30 years, and an increasing number of studies have recently combined models with high-throughput data sets for prospective experimentation. These studies have led to validation of increasingly important and relevant biological predictions. As reviewed here, these recent successes have tangible implications in the fields of microbial evolution, interaction networks, genetic engineering and drug discovery.
Article
The elucidation of breast cancer subgroups and their molecular drivers requires integrated views of the genome and transcriptome from representative numbers of patients. We present an integrated analysis of copy number and gene expression in a discovery and validation set of 997 and 995 primary breast tumours, respectively, with long-term clinical follow-up. Inherited variants (copy number variants and single nucleotide polymorphisms) and acquired somatic copy number aberrations (CNAs) were associated with expression in similar to 40% of genes, with the landscape dominated by cis-and trans-acting CNAs. By delineating expression outlier genes driven in cis by CNAs, we identified putative cancer genes, including deletions in PPP2R2A, MTAP and MAP2K4. Unsupervised analysis of paired DNA-RNA profiles revealed novel subgroups with distinct clinical outcomes, which reproduced in the validation cohort. These include a high-risk, oestrogen-receptor-positive 11q13/14 cis-acting subgroup and a favourable prognosis subgroup devoid of CNAs. Trans-acting aberration hotspots were found to modulate subgroup-specific gene networks, including a TCR deletion-mediated adaptive immune response in the 'CNA-devoid' subgroup and a basal-specific chromosome 5 deletion-associated mitotic network. Our results provide a novel molecular stratification of the breast cancer population, derived from the impact of somatic CNAs on the transcriptome.