Leading Professional Society for Computational Biology and Bioinformatics
Connecting, Training, Empowering, Worldwide

banner

Posters - Schedules

Poster presentations at ISMB/ECCB 2021 will be presented virtually. Authors will pre-record their poster talk (5-7 minutes) and will upload it to the virtual conference platform site along with a PDF of their poster beginning July 19 and no later than July 23. All registered conference participants will have access to the poster and presentation through the conference and content until October 31, 2021. There are Q&A opportunities through a chat function and poster presenters can schedule small group discussions with up to 15 delegates during the conference.

Information on preparing your poster and poster talk are available at: https://www.iscb.org/ismbeccb2021-general/presenterinfo#posters

Ideally authors should be available for interactive chat during the times noted below:

View Posters By Category

Session A: Sunday, July 25 between 15:20 - 16:20 UTC
Session B: Monday, July 26 between 15:20 - 16:20 UTC
Session C: Tuesday, July 27 between 15:20 - 16:20 UTC
Session D: Wednesday, July 28 between 15:20 - 16:20 UTC
Session E: Thursday, July 29 between 15:20 - 16:20 UTC
A model-based data integration pipeline to characterize the multi-level regulation of cell metabolism
COSI: SysMod
  • Marzia Di Filippo, University of Milano-Bicocca, Italy
  • Dario Pescini, University of Milano-Bicocca, Italy
  • Bruno Giovanni Galuzzi, University of Milano-Bicocca, Italy
  • Lilia Alberghina, University of Milano-Bicocca, Italy
  • Marco Vanoni, University of Milano-Bicocca, Italy
  • Giancarlo Mauri, University of Milano-Bicocca, Italy
  • Chiara Damiani, University of Milano-Bicocca, Italy

Short Abstract: The study of metabolism and its regulation is finding increasing application in various fields, including health, wellness, and biotransformations. Complete characterization of regulatory mechanisms controlling metabolism requires knowledge of metabolic fluxes, whose direct determination lags behind other omic technologies, such as metabolomics and transcriptomics. In isolation, these methodologies do not allow accurate characterization of metabolic regulation. Hence, there is a need for integrated methodologies to disassemble the interdependence between different regulatory layers controlling metabolism.
To this aim, we propose a computational pipeline to characterize the landscape of metabolic regulation in different biological samples. The method integrates intracellular and extracellular metabolomics, and transcriptomics, using constraint-based stoichiometric metabolic models as a scaffold. We compute differential reaction expression from transcriptomic data and use constraint-based modeling to predict if the differential expression of metabolic enzymes directly originates differences in metabolic fluxes. In parallel, using metabolomic data, we predict how differences in substrate availability translate into differences in metabolic fluxes. By intersecting these two output datasets, we discriminate fluxes regulated at the metabolic and/or transcriptional level. This information is valuable to better inform targeted action planning in different fields, including personalized prescriptions in multifactorial diseases, such as cancer, and metabolic engineering.

An agent-based model of tumour-associated macrophage differentiation in chronic lymphocytic leukaemia
COSI: SysMod
  • Nina Verstraete, Cancer Research Center of Toulouse INSERM UMR 1037, France
  • Malvina Marku, Cancer Research Center of Toulouse INSERM UMR 1037, France
  • Hélène Arduin, Cancer Research Center of Toulouse INSERM UMR 1037, France
  • Marcin Domagala, Cancer Research Center of Toulouse INSERM UMR 1037, France
  • Jean Jacques Fournié, Cancer Research Center of Toulouse INSERM UMR 1037, France
  • Loic Ysebaert, Cancer Research Center of Toulouse INSERM UMR 1037, France
  • Mary Poupot, Cancer Research Center of Toulouse INSERM UMR 1037, France
  • Vera Pancaldi, Cancer Research Center of Toulouse INSERM UMR 1037, France

Short Abstract: In the tumor microenvironment, tumor-associated macrophages are known to play a critical role in the survival and chemoresistance of cancer cells. In the case of chronic lymphocytic leukemia (CLL), these tumor-associated macrophages are called Nurse-Like Cells (NLCs) and reside mainly in the lymph nodes, where they are able to protect leukemic B cells (B-CLL) from spontaneous apoptosis and contribute to their chemoresistance. NLCs are differentiated from monocytes through cytokines signalling and physical contact with the cancer cells [1], however, the precise mechanisms by which B-CLL cells influence this differentiation are still unknown.

We propose here an agent-based model (ABM) of monocyte-to-macrophage differentiation in an in-vitro co-culture of monocytes and cancer B-CLL cells. This model is a first step to a better understanding of the spatio-temporal dynamics of the tumor microenvironment and of the mechanisms used by cancer cells to influence monocyte differentiation into pro-tumoral macrophages. A more complex model taking into account other immune cell types and integrating Boolean modeling of relevant signaling pathways inside each agent is the next step.

[1] F Boissard et al. Nurse like cells: Chronic lymphocytic leukemia associated macrophages. Leuk. Lymphoma, 56(5):1570- 1572, 2015.

Automated whole-cell modeling from genomic sequence and multi-omics data
COSI: SysMod
  • Kazunari Kaizu, RIKEN Center for Biosystems Dynamics Research, Japan
  • Kozo Nishida, Riken, Japan
  • Koichi Takahashi, RIKEN, Japan

Short Abstract: Recent rapid advances in genome synthesis have increased the significance of genome design techniques. Whole-cell modeling is one of the effective methods for genome design. However, whole-cell models are often constructed manually, and it is difficult to keep them up-to-date against the huge amount of new knowledge. In addition, it is now required to build models on-demand for single nucleotide-level design. Here, we developed a technique to automatically construct and simulate whole-cell models of a Gammaproteobacteria represented by Escherichia coli, based on a genome sequence and multiple omics data as its input. This workflow includes predictions of annotations, such as CDSs, operons, and replication initiation sites, on the given sequence, kinetic modeling of metabolic pathways, and parameter determination against multi-omics data (transcriptome, proteome, and metabolome). The workflow is constructed on Snakemake and confers the reproducibility of modeling. The whole-cell model consists of gene expression and replication system described in an agent-based manner with more than 1 million agents, and a kinetic metabolic model governed by ordinary differential equations of hundreds of variables. The automated modeling can facilitate the construction and maintenance of large complex models and allow us to design a genome from scratch toward an era of synthetic cells.

Boolean Network Inference at Different Levels of Logical Complexity
COSI: SysMod
  • Eline S. van Mantgem, Heinrich Heine Universität Düsseldorf, Germany
  • Gunnar W. Klau, Heinrich Heine Universität Düsseldorf, Germany

Short Abstract: The underlying dynamics of biological signalling networks is often unknown. Previous work on unravelling these complex networks includes heuristic and exact methods that aim to uncover the Boolean functions behind each interaction in an annotated network by explaining experimental observations given as stimuli/response sets.

Our main goal is to infer the logical topology of a network, without the need for a prior annotated network. We consider a generalized framework for inferring Boolean functions of increasing logical complexity, including rudimentary Boolean functions, threshold gates and completely unrestricted truth tables. As a secondary objective, we compare integer linear programming (ILP) to satisfiability solving (SAT) solutions. For each complexity level, we provide two alternative formulations, one based on ILP, which extends previous work to the generalized setting, and a novel formulation based on SAT. Preliminary results of experiments on a well-known EGFR dataset as well as on synthetic data indicate that the SAT implementation significantly outperforms the ILP implementation in terms of running time and can infer an optimal model in networks of up to 205 nodes and 204 edges given 18 experiments in less than 5 minutes on a standard laptop computer.

BpForms and BcForms: a toolkit for concretely describing non-canonical polymers and complexes to facilitate global biochemical networks
COSI: SysMod
  • Paul Lang, University of Oxford, Icahn School of Medicine at Mount Sinai, United Kingdom
  • Yassmine Chebaro, Icahn School of Medicine at Mount Sinai, United States
  • Xiaoyue Zheng, Icahn School of Medicine at Mount Sinai, United States
  • John A. P. Sekar, Icahn School of Medicine at Mount Sinai, United States
  • Bilal Shaikh, Georgetown University Medical Center, United States
  • Darren A. Natale, Georgetown University Medical Center, United States
  • Jonathan R. Karr, Icahn School of Medicine at Mount Sinai, United States

Short Abstract: A central goal in systems biology is to understand how all of the molecules and processes in cells interact to generate behavior. While small molecules can be concretely described by molecular formats such as SMILES, macromolecules are frequently described as sequences of canonical residues. However, non-canonical residues, caps, crosslinks, and nicks are important to many functions of DNAs, RNAs, proteins, and complexes. One barrier towards models that explain how networks of such non-canonical macromolecules perform complex functions is our limited formats for concretely describing them. To overcome this barrier, we develop BpForms and BcForms, a toolkit for unambiguously representing the primary structure of macromolecules as combinations of residues, caps, crosslinks, and nicks. The toolkit can help omics researchers perform quality control and exchange information about macromolecules, help systems biologists assemble global models of cells that encompass processes such as post-translational modification, and help bioengineers design cells.

Curating and Comparing 114 Strain-Specific Genome-Scale Metabolic Models of Staphylococcus aureus
COSI: SysMod
  • Andreas Dräger, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany
  • Alina Renz, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany

Short Abstract: Staphylococcus aureus is a high-priority pathogen causing severe infections with high morbidity and mortality worldwide. It is one of the most successful and prominent modern pathogens. An effective fight against S. aureus infections requires novel antimicrobial targets. Recent advances in whole-genome sequencing and high-throughput techniques facilitate the generation of genome-scale metabolic models (GEMs). Among the multiple applications of GEMs is drug-targeting in pathogens. Hence, comprehensive and predictive metabolic reconstructions of S. aureus could facilitate the identification of novel targets for antimicrobial therapies.

This work aims at reviewing all 114 available GEMs of multiple S. aureus strains. We updated each model to a current version of SBML and evaluated its scope, including the number of reactions, metabolites, and genes. Furthermore, all models were quality-controlled using MEMOTE. Growth capabilities and model similarities were examined.

This work should lead as a guide for choosing the appropriate GEM for a given research question. With the information about the availability, the format, and the strengths and potentials of each model, one can either choose an existing model or combine several models to create models with even higher predictive values. This facilitates model-driven discoveries of novel antimicrobial targets to fight multi-drug resistant S. aureus strains.

Data-driven learning of continuum models from stochastic simulations of non-equilibrium biological dynamics
COSI: SysMod
  • Suryanarayana Maddu, Max Planck Institute for Molecular Cell Biology and Genetics, Germany
  • Christian L. Müller, Ludwig Maximilian University of Munich, Germany
  • Ivo F. Sbalzarini, Max Planck Institute for Molecular Cell Biology and Genetics, Germany

Short Abstract: We present a statistical learning framework to infer deterministic mean-field models from stochastic dynamics of mechanically active agents, such as cytoskeletal filaments and motor proteins, cells, or animals in a swarm. Physical theories of self-organized, non-equilibrium active systems exist both at the microscopic scale of individual agents (e.g., Brownian dynamics, Langevin equations), and at the continuum mean-field scale (e.g., active polar gel equtions). Naturally, the question arises \textit{how} interactions between microscopic components lead to mean-field dynamics and \textit{which} continuum description is sufficient to describe a given microscopic collective. Deriving such a coarse-graining by hand using classic methods like homogenization or volume averaging is tedious and difficult, due to the non-equilibrium physics, nonlinearities, and inherent fluctuations of the microscopic system. Data-driven inference of coarse-grained models from microscopic data has therefore emerged as a complementary approach, enabled by advances in machine learning~\cite{rudy2017data}.

Filling mechanistic knowledge gaps in biological models through a hybrid AI-ODE framework
COSI: SysMod
  • Anna Fochesato, Fondazione The Microsoft Research - University of Trento COSBI, Italy
  • Federico Reali, Fondazione The Microsoft Research - University of Trento COSBI, Italy
  • Luca Marchetti, Fondazione The Microsoft Research - University of Trento COSBI, Italy

Short Abstract: Defining differential equations at certain biological levels may be hampered by the lack of mechanistic details. Phenomenological equations are a rule of thumb to fill these knowledge gaps and get a model, but they come at the cost of being arbitrary choices. In this context, deep learning and hierarchical modeling can complement each other to automatically decipher trajectory patterns and variable interplay from data and underlying physics. Such integrated technologies may bridge low-level molecular descriptions and clinical manifestations as needed in quantitative systems pharmacology (QSP) applications.
We developed a PyTorch pipeline that integrates a Long Short-Term Memory (LSTM) neural network and known ODEs to reproduce the dynamics of model variables with missing equations and to forecast future model states. Our approach is designed to generalize outside the training domain and does not require prior intuition on suitable regression classes nor dynamical laws. In our benchmarks, the variable interplay within the AI-ODE system and the adopted learning protocol improved the training procedure, leading to accurate predictions as a result. Further tests on relevant QSP applications and a modeling-oriented extension of the pipeline to account for model rate calibration are currently under investigation.

Genome-scale modeling of Pseudomonas aeruginosa PA14 unveils broad metabolic capabilities and role of metabolism in virulence and drug potentiation
COSI: SysMod
  • Sanjeev Dahal, Queen's University, Canada
  • Laurence Yang, Queen's University, Canada

Short Abstract: A highly curated genome-scale metabolic model of cystic fibrosis (CF) pathogen, Pseudomonas aeruginosa PA14 was developed in this study. During the reconstruction process, a set of substrate utilization and gene-essentiality data was used to improve the predictive abilities of the model. Furthermore, strain-specific processes (e.g., phenazine transport and redox metabolism, cofactor metabolism, carnitine metabolism, oxalate metabolism, etc.) were added to the reconstruction after a thorough literature review. Through this extensive process, a three-compartment, BiGG model, iSD1511 was created which is a significant improvement over the previous modeling effort for this strain. iSD1511 was assessed using another set of gene essentiality and substrate utilization data, and the prediction accuracies as high as 92.7% and 93.5%, respectively, were computed. The model can simulate growth in both aerobic and anaerobic conditions. iSD1511 can also be used to simulate condition-dependent production of phenazines and their effect on the growth of PA14 strain. Finally, the model was used to provide mechanistic explanations for two case studies: a) effect of gene deletion on gluconate production, and b) metabolic influence on drug tolerance of P. aeruginosa. Overall, this work provides a computational framework to aid in the development of effective intervention strategies against P. aeruginosa.

High-quality genome-scale reconstruction of Corynebacterium glutamicum ATCC 13032
COSI: SysMod
  • Elisabeth Zelle, Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Germany
  • Katharina Nöh, Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Germany
  • Wolfgang Wiechert, Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Germany
  • Martina Feierabend, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany
  • Alina Renz, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany
  • Andreas Dräger, Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Germany

Short Abstract: The reconstruction of genome-scale metabolic models (GEMs) is a growing field within systems biology. GEMs allow predicting varying phenotypic states. Thus, they have proven useful to guide metabolic engineering strategies of relevant biotechnological organisms. One microbe of particular biotechnological relevance is the Corynebacterium glutamicum, ideal for producing l-amino acids at an industrial scale. We here present an updated high-quality GEM of the Corynebacterium glutamicum ATCC 13032.
The high reconstruction quality of this GEM is obtained by applying a high level of annotations and cross-references of metabolites, reactions, and genes. The GEM comprises 1,045 metabolites, 1,545 reactions, and 807 genes. Our model reproduces experimentally validated data and had realistic growth rates when tested on different media under aerobic and anaerobic conditions. Moreover, the model produces all canonical amino acids.
This new in-silico model is useful in filling knowledge gaps in C. glutamicum: l-glutamate could still be produced even when the formerly believed relevant enzyme pyruvate carboxylase was knocked out. Our study shows that by integrating high reconstruction standards, GEMs can prove fruitful in consistently reproducing experimentally validated knowledge, filling knowledge gaps, and supporting metabolic engineers.

HiLoop: a toolbox for detection, quantification, modeling, and visualization of high feedback in gene regulatory networks
COSI: SysMod
  • Benjamin Nordick, School of Genome Science and Technology, University of Tennessee Knoxville, United States
  • Tian Hong, Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee Knoxville, United States

Short Abstract: Interconnected feedback loops in gene regulatory networks can give rise to sophisticated dynamics such as stepwise cell lineage commitment or low-rate irreversible differentiation. Despite their importance in biology, there is a lack of tools for extracting and visualizing these interconnected, “high-feedback” subnetworks from complex networks. Though many algorithms can enumerate or detect enrichment of specific motifs in networks, they cannot search for motifs defined by cycle interconnection. We developed a software toolbox HiLoop that can enumerate and intuitively display instances of high-feedback motifs even from large networks. With HiLoop, dynamics of selected subnetworks can be modeled with ordinary differential equations under varied parameter sets to characterize the landscape of possible attractors. HiLoop can compare a network to numerous permutations thereof to quantify the enrichment of high-feedback motifs. We used HiLoop to show that a network of genes involved in epithelial-mesenchymal transition is strongly enriched in a particular interconnection of three positive feedback loops. Modeling output of several selected subnetworks compactly yet clearly communicated the types of multistable and/or oscillatory systems that they are expected to produce. We contribute HiLoop as both a hypothesis generator and quantification method for exploring dynamics of complex feedback-rich regulatory networks.

Hybrid system models for phage regulatory networks: what can we learn?
COSI: SysMod
  • Juris Viksna, Institute of Mathematics and Computer Science, University of Latvia, Latvia
  • Karlis Cerans, Institute of Mathematics and Computer Science, University of Latvia, Latvia
  • Karlis Freivalds, Institute of Mathematics and Computer Science, University of Latvia, Latvia
  • Lelde Lace, Institute of Mathematics and Computer Science, University of Latvia, Latvia
  • Gatis Melkus, Institute of Mathematics and Computer Science, University of Latvia, Latvia
  • Darta Rituma, Institute of Mathematics and Computer Science, University of Latvia, Latvia

Short Abstract: Gene regulatory networks form an attractive avenue for understanding biological systems. Among gene regulatory networks, the lysis/lysogeny switch in bacteriophage lambda has received particular attention as the classical example of a genetic switch. We have previously found success in analyzing bacteriophage lamba using a hybrid system model formalism which reveals that the lytic and lysogenic states form stable attractors in our model's state space. In our current work we further refine our hybrid system based modeling approach by expanding our phage model with additional data, analyzing the model’s overall robustness across a variety of conditions and network layouts. We apply and adapt our insights into the regulatory patterns of phage lambda to additional temperate phages, notably the HK022 lambdoid phage and Mu.

Identification of linked metabolites allows a reliable and quantitative analysis of the finger sweat metabolome
COSI: SysMod
  • Jürgen Zanghellini, Department of Analytical Chemistry, University of Vienna, Austria
  • Mathias Gotsmy, Department of Analytical Chemistry, University of Vienna, Austria
  • Julia Brunmair, Department of Analytical Chemistry, University of Vienna, Austria, Austria
  • Andrea Bileck, Joint Metabolome Facility, University and Medical University of Vienna, Austria, Austria
  • Astrid Slany, Department of Analytical Chemistry, University of Vienna, Austria
  • Samuel Matthias Meier-Menches, Institute of Inorganic Chemistry, University of Vienna, Austria
  • Christopher Gerner, Joint Metabolome Facility, University and Medical University of Vienna, Austria

Short Abstract: Typically, clinical metabolome analysis is performed on blood samples. However, drawing blood is not only a cumbersome procedure for patients but requires qualified personnel which impairs measurement during real life settings. A promising alternative is the analysis of the metabolome from finger sweat, which benefits from simple sampling procedures. However, a major obstacle is the inability to control the amount of sweat produced by the sweat glands on the fingertips at any given time. Not addressing this problem prevents a reliable quantification of metabolites.

Here we present a computational method based on the identification of metabolically linked species in the sweat metabolome that allows us to estimate sweat volumes and enables an individualized, accurate, quantitative finger sweat analysis for clinical applications. In a proof-of-principal application we used short interval sampling of sweat from fingertips to monitor the dynamic response of 41 individuals after caffeine consumption. We not only identified corresponding xenobiotics concentration time-series but extracted individualized kinetic parameters of caffeine metabolisation from sweat and show their long-time stability.

In conclusion, this work highlights the feasibility of individualized and reliable biomonitoring using sweat samples from fingertips which may have far reaching implications for personalised medical diagnostics and biomarker discovery.

Integrative modelling of chondrocyte mechanotransduction and biochemical stimulation
COSI: SysMod
  • Maria Segarra-Queralt, Universitat Pompeu Fabra, Spain
  • Gemma Piella, Universitat Pompeu Fabra, Spain
  • Jérôme Noailly, Universitat Pompeu Fabra, Spain

Short Abstract: Through mechanotransduction (MT) mechanical loads cause changes on chondrocyte (CC) phenotype. Systematic representations of these procedures can inform early strategies for osteoarthritis management, characterized by an increase of catabolic events compared to biosynthetic activity. To this end, a network model of CC activity that incorporates important mechanosensors (MS) and the downstream signalling cascades is proposed. Key mechanoregulatory molecular interactions is mapped in an interactome, based on 72 journal articles. It is translated into a semi-quantitative mathematical model based on a system of differential equations that converges to steady states (SS), which can be extrapolated to systematic descriptions of CC metabolism. An anabolic SS is reached when a MS related to physio-osmotic conditions is activated: TRPV4. A pro-inflammatory stimulus and an injurious load perturbation (PIEZO1/2 activation) can induce a catabolic shift (t-test, α=0.05) compared to the physio-osmotic SS: pro-inflammatory cytokines and proteases have higher expression rates than structural proteins. Remarkably, Sox9 (a healthy marker), is highly expressed in the physio-osmotic SS; but in an injurious environment NFkB, HIF2a or Runx2 increase (related to inflammatory and hypertrophic events). An intracellular network-based model of a CC is developed that could predict expected MT and inflammation effects revealing the potential of exploitation in OA.

Metabolic drug repurposing for autoimmune diseases
COSI: SysMod
  • Matteo Barberis, University of Surrey, United Kingdom
  • Tomáš Helikar, University of Nebraska-Lincoln, United States
  • Bhanwar Lal Puniya, University of Nebraska-Lincoln, United States
  • Rada Amin, University of Nebraska-Lincoln, United States
  • Bailee Lichter, University of Nebraska-Lincoln, United States
  • Robert Moore, University of Nebraska-Lincoln, United States
  • Alex Ciurej, University of Nebraska-Lincoln, United States
  • Sydney Bennett, University of Nebraska-Lincoln, United States
  • Ab Rauf Shah, University of Nebraska-Lincoln, United States
  • Zhongyuan Zhao, University of Nebraska-Lincoln, United States
  • Brandt Bessell, University of Nebraska-Lincoln, United States

Short Abstract: CD4+ T cells provide cell-mediated protection against diseases. When dysregulated, CD4+ T cells are associated with autoimmune and other immune-mediated diseases. Metabolism of CD4+ T cells regulates their function, therefore offer an opportunity to explore as a drug target against autoimmune diseases. In this study, we developed constraint-based models of naive and T helper 1, 2, and 17 subtypes. We mapped existing drugs and compounds and simulated metabolic behaviors under drug-induced inhibitions of metabolic genes. We integrated these metabolic behaviors with gene expression data of three autoimmune diseases, rheumatoid arthritis (RA), multiple sclerosis (MS), and primary biliary cholangitis (PBC). We identified and prioritized drugs and their targets that reversed the directions of differentially expressed genes in diseases. We identified 68 metabolic drug targets for the three studied diseases. We performed in vitro experiments and mined experiments available in the literature to validate results. The experimental results showed that 50% of the drug targets suppressed CD4+ T cell proliferation. In the end, we developed an integrated pipeline to explore metabolic models to identify drug targets and repurposable drugs.

Multi-compartment models (MCMs) reveal echoing of transcriptional response during effector-triggered immunity in Arabidopsis
COSI: SysMod
  • Chad Myers, University of Minnesota, United States
  • Xiaotong Liu, University of Minnesota, United States
  • Fumiaki Katagiri, University of Minnesota, United States

Short Abstract: Effector-Triggered Immunity (ETI), which is induced by some pathogen effectors, is one major mode of plant innate immunity and is associated with massive transcriptional reprogramming. Plant cells directly receiving such effectors (cell population 1) and those indirectly responding (cell population 2) probably have different transcriptome dynamics but cannot be characterized separately with bulk RNA-seq. We analyzed a public time-series transcriptome dataset profiled during ETI in Arabidopsis after inoculation of Pseudomonas syringae strain expressing the effector AvrRpt2. We found that double-peak transcript patterns were prevalent among 2329 ETI-inducible genes. To quantitatively interpret the double-peak patterns, we developed a novel computational approach based on Multi-compartment models (MCMs) and fit the model to the transcript data of each gene. The model enables a linear decomposition of a double-peak pattern into two single-peak patterns and therefore allows us to characterize the peak patterns parametrically, such as the peak time and peak amplitude. We demonstrated that the first- and second-peaks of most genes highly likely represent the responses in cell populations 1 and 2, respectively and the timing and amplitude of the second peak response for each gene echoes the dynamics of the first peak response for the majority of genes.

Network modelling of articular chondrocyte molecular regulation in health and osteoarthritis
COSI: SysMod
  • Maria Segarra-Queralt, Universitat Pompeu Fabra, Spain
  • Michael Neidlin, National Techinical University of Athens, Greece
  • Gemma Piella, Universitat Pompeu Fabra, Spain
  • Laura Tio, IMIM, Spain
  • Leonidas Alexopoulos, National Technical University of Athens, Greece
  • Miguel Ánguel González-Ballester, ICREA, Spain
  • Jérôme Noailly, Universitat Pompeu Fabra, Spain

Short Abstract: Overstimulation of catabolic events compared to biosynthetic activity of cartilage chondrocytes (CC) leads to articular cartilage degradation when osteoarthritis (OA) appears. Mechanical and biochemical signals are involved in this dysregulation. To allow the semi-quantitative interpretation of these proceedings, we propose a network-based model (NBM) at the CC level that incorporates the actions of catabolic and anabolic factors on the expression of structural proteins and proteases. Understanding better such mechanisms might leverage the identification of new therapeutic targets that could stop OA progression and improve current conservative pain-killers’ treatments. Our NBM appears from a combination of knowledge-based and a data-driven approaches. First, an OA specific protein interactome was developed based on reported actions in specialized literature. Then, a targeted network enrichment is developed using STRING. Finally, it is successfully calibrated against experimental data through a genetic algorithm. Independent validation with multiplexed phosphoproteomics measurements shows that the optimized NBM has a relative error of 3.5%. It also captures CC’s reported behaviors with 95% of accuracy, and it correctly predicts the main outcomes of an OA treatment with biologics. Therefore, the proposed methodology allows us to model an optimal NBM that controls CC metabolism. Further research should target the incorporation of mechanical signals.

Predicting Viral Life History Traits from Vaccine Resistance to Endemicity
COSI: SysMod
  • Nash Rochman, The National Institutes of Health, United States
  • Yuri Wolf, The National Institutes of Health, United States
  • Eugene Koonin, The National Institutes of Health, United States

Short Abstract: The prediction of emergent variant characteristics remains extremely challenging. Faced with a new pathogen, it is desirable to be able to forecast the case fatality rate (CFR) into the future. Leveraging a compartment model, we reveal general constraints among human pathogenic respiratory viruses where the variation of multiple parameters in concert leads to decreased virulence and increased pathogen fitness but not independent variation. Highly virulent viruses are constrained by host behavior, whereas moderately virulent viruses are constrained by the relationship between the duration of immunity and CFR. When the immune population is rapidly expanded through vaccination, specific predictions can be made in the face of dramatically altered selective pressures. The potential emergence of vaccine resistance constrains optimal vaccine distribution. Analogous to low-dose antibiotic exposure, recently vaccinated, partially immunized individuals play an outsized role in the emergence of resistance. When an escape variant is modestly less infectious than the originating strain, there exists an optimal rate of vaccine distribution. Exceeding this rate increases the cumulative number of infections due to vaccine escape. Modulating the rate of host-host contact for the recently vaccinated population by less than an order of magnitude can alter the cumulative number of infections by more than 20%.

RCGAToolbox: A real-coded genetic algorithm software for parameter estimation of kinetic models
COSI: SysMod
  • Kazuhiro Maeda, Kyushu Institute of Technology, Japan
  • Fred Boogerd, VU University Amsterdam, Netherlands
  • Hiroyuki Kurata, Kyushu Institute of Technology, Japan

Short Abstract: Kinetic modeling is essential in understanding the dynamic behavior of biochemical networks, such as metabolic and signal transduction pathways. However, parameter estimation remains a major bottleneck in the development of kinetic models.

We present RCGAToolbox, software for real-coded genetic algorithms (RCGAs), which accelerates the parameter estimation of kinetic models. RCGAToolbox provides two RCGAs: the unimodal normal distribution crossover with minimal generation gap (UNDX/MGG) and real-coded ensemble crossover star with just generation gap (REXstar/JGG), using the stochastic ranking method. The RCGAToolbox also provides user-friendly graphical user interfaces.

We tested 33 problems: 27 mathematical benchmarks and six parameter estimations. The performance of RCGAToolbox was comparable to that of an existing parameter estimation tool. We also found that REXstar/JGG outperformed a widely-used algorithm in one of the parameter estimation problems.

RCGAToolbox is available from github.com/kmaeda16/RCGAToolbox under GNU GPLv3, with application examples. RCGAToolbox runs on MATLAB in Windows, Linux, and Mac.

Relating early cellular events to Drug-Induced Liver Injury (DILI) using time-resolved transcriptomic and histopathology data
COSI: SysMod
  • Andreas Bender, University of Cambridge, United Kingdom
  • Anika Liu, University of Cambridge, United Kingdom
  • Namshik Han, University of Cambridge, United Kingdom
  • Jordi Munoz-Muriedas, GSK, United Kingdom

Short Abstract: To enable earlier detection of compound toxicity in drug development, it is necessary to better understand the biological mechanisms. We aimed to identify early changes in pathway and transcription factor (TF) activity relevant for Drug-Induced Liver Injury which take place before adverse histological changes are observed. To do so, we used data from the TG-GATEs database, which comprises time-resolved transcriptomics and histopathology from repeat-dose studies in rats across eight timepoints, ranging from 3 hours to 4 weeks. We were able to recover known events preceding DILI, with some being more frequent, e.g. mitophagy, and others more confident, e.g. bile acid recycling. Furthermore, we prioritized additional pathways and transcription factors (TFs) based on time concordance. Among TFs, we were able to separate induced TFs, such as Cebpa, from post-transcriptionally activated ones, e.g. Srebf2, based on whether differential TF expression is observed before changes in regulon activity. Furthermore, we identified interactions between TFs which are supported by functional interactions as well as time concordance providing hypotheses on toxicity-related gene-regulatory mechanisms, e.g. Hnf4a-dependent Cebpa expression. Overall, we demonstrate how time-resolved transcriptomics can derive mechanistic hypothesis and how this can be combined with other streams of causal evidence.

SimiC enables the inference of complex gene regulatory dynamics across cell phenotypes
COSI: SysMod
  • Idoia Ochoa, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801., United States
  • Mikel Hernaez, Computational Biology Program, CIMA University of Navarra, Spain., Spain
  • Jianhao Peng, Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
  • Guillermo Serrano, Computational Biology Program, CIMA University of Navarra, Spain., Spain
  • Ian M. Traniello, Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801., United States
  • Maria E. Calleja-Cervantes, Hemato-Oncology Program, CIMA University of Navarra, Spain., Spain
  • Ullas V. Chembazhi, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801., United States
  • Sushant Bangru, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801., United States
  • Teresa Ezponda, Hemato-Oncology Program, CIMA University of Navarra, Spain., Spain
  • Juan Roberto Rodriguez-Madoz, Hemato-Oncology Program, CIMA University of Navarra, Spain., Spain
  • Auinash Kalsotra, Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801., United States
  • Felipe Prosper, Hemato-Oncology Program, CIMA University of Navarra, Spain., Spain

Short Abstract: Single-cell RNA-Sequencing has made possible to infer high-resolution gene regulatory networks (GRNs), providing deep biological insights by revealing regulatory interactions at single-cell resolution. Current single-cell GRN inference methods produce a single GRN per input dataset, limiting their capability to uncover complex regulatory relationships across related cell phenotypes and potentially missing relationships between cells from different phenotypes.
We present SimiC, a single-cell GRN inference framework that enables uncovering complex regulatory dynamics at single-cell resolution across a range of systems, both model and non-model alike, that would have otherwise been missed by previously proposed methods. SimiC produces one GRN per phenotype while imposing a similarity constraint that allows for a smooth transition between GRNs, enabling a direct comparison between different states, treatments, or conditions.
We tested SimiC on simulated and real datasets, showing improved performance with respect to previous methods. Specifically, SimiC was able to: i) recapitulate CAR T cell dynamics after tumor recognition; ii) unravel well-known regulatory patterns on a regenerating liver; and iii) implicating glial cells in the generation of distinct behavioral states in honeybees.
Thus, SimiC establishes a new approach to quantitating regulatory architectures between GRNs of distinct cellular phenotypes, with far-reaching implications for systems biology.

Simulating drug effects on whole-cell level simulation
COSI: SysMod
  • Bence Keömley-Horváth, Pázmány Péter Catholic University, Hungary
  • Attila Csikász-Nagy, Pázmány Péter Catholic University, Hungary
  • István Reguly, Pázmány Péter Catholic University, Hungary

Short Abstract: Our group developed a whole-cell protein complex prediction tool called Cytocast. With that tool, we can simulate how the proteins bind and unbind and form different kinds of protein complexes, which can help us to understand how the cell behaves in different conditions. Based on the input data, our model can simulate any cell in any state. The input file describes the cell's geometrical structure, the proteins and complexes, and localization and abundances. The interactions are described on the level of the proteins' binding sites. These binding sites do not have any structure, and they are referred to as point-like objects. The cell is divided into tens of thousands of sub-volumes. Thus the reaction simulation can run separately, and the particles are mixed through the diffusions.
We created a pipeline that provides data from our database and analysis tools to evaluate the simulation results of the different conditions. We can use this to compare treated and not-treated cells for different tissues and predicting the main and side effects of drugs.

The in silico experiments on Antigen-presenting Cell signaling pathways
COSI: SysMod
  • Sara Sadat Aghamiri, University of Nebraska Lincoln, United States
  • Rada Amin-Ali, University of Nebraska Lincoln, United States
  • Tomas Helikar, University of Nebraska Lincoln, United States

Short Abstract: Antigen-presenting cells (APC) are immune cells mediating immune response by processing and presenting antigens to lymphocytes such as T cells. Dendritic cells (DC) are considered critical APCs that play an essential role in immune responses. Developments in Omics-technologies revolutionized the research in biological fields, which significantly increased the demand for integrative approaches to address novel predictions and analysis. One of these approaches is computational modeling that provides an executable, dynamic network revealing system-level behaviors through in silico perturbations. The in silico simulations provide a platform for researchers to generate new or rank existing hypotheses before starting the wet-lab experiments, saving time, energy, and materials.
Our DC model is a manual curated, entirely annotated, and expert validated dynamic network comprising different cellular compartments to facilitate tracking flow of information in the system (e.g. extracellular stimuli, surface markers, signaling pathways, transcription factors, and secreted cytokines). The model is built in the Cell Collective platform utilizing logical formalism and contains more than two hundred nodes and interactions illustrating the APC function in DC.
Taking together, our DC cell-specific model of APC functions will address the links between the innate and adaptive immunity crosstalk.

The natural history of clonal haematopoiesis
COSI: SysMod
  • José Guilherme de Almeida, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK, United Kingdom
  • Margarete Fabre, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK, United Kingdom
  • George Vassiliou, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK, United Kingdom
  • Moritz Gerstung, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK., United Kingdom

Short Abstract: Clonal haematopoiesis (CH) describes the clonal expansion of mutant haematopoietic stem cells in healthy and mostly elderly individuals. Yet CH is also considered an important step in the progression to blood cancers, such as acute myeloid leukaemia and myelodysplastic syndrome. Therefore, understanding CH evolution is important to better understand the progression from seemingly inoffensive somatic mutation to cancer. Here, we use longitudinal deep targeted sequencing data for 385 elderly individuals to estimate clonal dynamics and age at onset for all clones through hierarchical Bayesian modelling. We observe distinct annual growth rates for each gene, with the lowest occurring in DNMT3A (4%) and the highest in U2AF1 (21%). The extrapolated age at onset of clonal growth shows similar patterns for most genes, with clones arising uniformly through life. Against this trend, mutations such as those in U2AF1 or SRSF2-P95H appear exclusively later in life. With single cell colonies for 3 individuals we are able to verify our estimates and observe clonal expansions with no known drivers, suggesting that a portion of CH goes by unnoticed. This work offers a comprehensive view on the dynamics and evolution of CH, disentangling the effects that driver genetics and other effects have on CH progression.

The Systems Biology Simulation Core Library
COSI: SysMod
  • Hemil Panchiwala, Department of Computer Science and Engineering, Indian Institute of Technology, India
  • Shalin Shah, Department of Electrical and Computer Engineering, Duke University and Bloomberg LP, United States
  • Hannes Planatscher, Signatope GmbH, Germany
  • Mykola Zakharchuk, Department of Computer Science, University of Tübingen, Germany
  • Matthias König, Institute for Theoretical Biology, Humboldt University of Berlin, Germany
  • Andreas Dräger, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany

Short Abstract: Summary: Studying biological systems generally relies on computational modeling and simulation, e.g., for model-driven discovery and hypothesis testing. Progress in standardization efforts led to the development of interrelated file formats to exchange and reuse models in systems biology, such as SBML, the Simulation Experiment Description Markup Language (SED-ML), or the Open Modeling EXchange format (OMEX). Conducting simulation experiments based on these formats requires efficient and reusable implementations to make them accessible to the broader scientific community and to ensure the reproducibility of the results. The Systems Biology Simulation Core Library (SBSCL) provides interpreters and solvers for these standards as a versatile open-source API in Java™. The library simulates even complex biomodels and supports deterministic Ordinary Differential Equations (ODEs); Stochastic Differential Equations (SDEs); constraint-based analyses; recent SBML and SED-ML versions; exchange of results, and visualization of in silico experiments; open modeling exchange formats (COMBINE archives); hierarchically structured models; and compatibility with standard testing systems, including the Systems Biology Test Suite and published models from the BioModels and BiGG databases.
Availability: SBSCL is freely available at draeger-lab.github.io/SBSCL/ and via Maven.

Tissue-specific reconstruction of constraint-based metabolic models based on ReconX
COSI: SysMod
  • Nantia Leonidou, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany
  • Alina Renz, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany
  • Reihaneh Mostolizadeh, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany
  • Andreas Dräger, Computational Systems Biology, Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Germany

Short Abstract: COVID-19 has been characterized as one of the deadliest respiratory diseases. In this regard, scientists globally try to understand the host’s immunopathological response, how the novel coronavirus (SARS-CoV-2) adapts, and how it spreads. Currently, great efforts are made to detect effective therapies against the coronaviruses. Identifying potential antiviral targets is of great interest, and one way to detect them is by analyzing metabolic changes in infected cells. In 2012, Wang et al. published mCADRE, aiming to reconstruct tissue-specific models using gene expression data and network topology information. The algorithm is implemented in MATLAB, and its functionality is based solely on the first version of the human model resulting in its limited usability.

We present pymCADRE, a re-implementation of mCADRE in Python 3.8. Its functionality was tested using all three currently available versions of the human metabolic network. Internal optimizations done with fastFVA resulted in context-specific models closer to the ground truth. Additionally, host-virus models were created to help to identify potential antiviral targets against SARS-CoV-2. With those models, the recently identified potential antiviral target enzyme guanylate kinase was further investigated. Further improvements could be done to make it feasible with more complex models, like Recon2.2 and Recon3D.

Understanding the evolution of Apicomplexan metabolism through minimal metabolic model computation
COSI: SysMod
  • Nirvana Nursimulu, University of Toronto, Canada
  • Alan Moses, University of Toronto, Canada
  • John Parkinson, University of Toronto, Canada

Short Abstract: The phylum Apicomplexa comprises various medically-relevant parasites with diverse life cycle strategies, driven at least partly by distinct metabolic capacities. While the malaria-causing Plasmodium falciparum is mosquito-borne and infects liver cells and erythrocytes, Toxoplasma gondii is able to infect any nucleated cell of warm-blooded animals. Through constraint-based reconstruction and analysis, we seek to understand how each parasite has adapted its metabolism to best exploit its corresponding host environment. Our central hypothesis is that each parasite has reduced its metabolic potential in response to its ability to scavenge its hosts’ nutrients. We begin by reconstructing the metabolic models of various Apicomplexan parasites together with their free-living relatives (Vitrella brassicaformis and Chromera velia). We then apply mixed-integer linear programming to predict minimal metabolic models from the free-living models—functional models with the smallest number of reactions. Following in silico knock-out experiments in the free-living models, we identify essential reactions and compute their degree of conservation in the parasitic models. Next, we perform simulations to investigate which metabolic pathways are favoured in minimal models given different profiles of nutrient availabilities (representing different host environments). Overall, our analyses suggest that Apicomplexan metabolism has evolved reductively and so as to efficiently exploit host environments.



International Society for Computational Biology
525-K East Market Street, RM 330
Leesburg, VA, USA 20176

ISCB On the Web

Twitter Facebook Linkedin
Flickr Youtube