199 results on '"genome‐scale modeling"'
Search Results
2. Microbiome modeling: a beginner's guide.
- Author
-
Lange, Emanuel, Kranert, Lena, Krüger, Jacob, Benndorf, Dirk, and Heyer, Robert
- Subjects
BIOLOGICAL systems ,SYSTEMS biology ,COMPUTATIONAL biology ,MICROBIAL ecology ,RESEARCH personnel ,ANIMAL navigation ,BIOMES - Abstract
Microbiomes, comprised of diverse microbial species and viruses, play pivotal roles in human health, environmental processes, and biotechnological applications and interact with each other, their environment, and hosts via ecological interactions. Our understanding of microbiomes is still limited and hampered by their complexity. A concept improving this understanding is systems biology, which focuses on the holistic description of biological systems utilizing experimental and computational methods. An important set of such experimental methods are metaomics methods which analyze microbiomes and output lists of molecular features. These lists of data are integrated, interpreted, and compiled into computational microbiome models, to predict, optimize, and control microbiome behavior. There exists a gap in understanding betweenmicrobiologists andmodelers/bioinformaticians, stemming froma lack of interdisciplinary knowledge. This knowledge gap hinders the establishment of computational models in microbiome analysis. This review aims to bridge this gap and is tailored formicrobiologists, researchers newtomicrobiomemodeling, and bioinformaticians. To achieve this goal, it provides an interdisciplinary overview of microbiome modeling, starting with fundamental knowledge of microbiomes, metaomics methods, common modeling formalisms, and how models facilitate microbiome control. It concludes with guidelines and repositories for modeling. Each section provides entry-level information, example applications, and important references, serving as a valuable resource for comprehending and navigating the complex landscape of microbiome research and modeling. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Current State, Challenges, and Opportunities in Genome-Scale Resource Allocation Models: A Mathematical Perspective.
- Author
-
Schroeder, Wheaton L., Suthers, Patrick F., Willis, Thomas C., Mooney, Eric J., and Maranas, Costas D.
- Subjects
COMPUTATIONAL biology ,METABOLIC models ,SYSTEMS biology ,MATHEMATICAL models ,CELL size - Abstract
Stoichiometric genome-scale metabolic models (generally abbreviated GSM, GSMM, or GEM) have had many applications in exploring phenotypes and guiding metabolic engineering interventions. Nevertheless, these models and predictions thereof can become limited as they do not directly account for protein cost, enzyme kinetics, and cell surface or volume proteome limitations. Lack of such mechanistic detail could lead to overly optimistic predictions and engineered strains. Initial efforts to correct these deficiencies were by the application of precursor tools for GSMs, such as flux balance analysis with molecular crowding. In the past decade, several frameworks have been introduced to incorporate proteome-related limitations using a genome-scale stoichiometric model as the reconstruction basis, which herein are called resource allocation models (RAMs). This review provides a broad overview of representative or commonly used existing RAM frameworks. This review discusses increasingly complex models, beginning with stoichiometric models to precursor to RAM frameworks to existing RAM frameworks. RAM frameworks are broadly divided into two categories: coarse-grained and fine-grained, with different strengths and challenges. Discussion includes pinpointing their utility, data needs, highlighting framework strengths and limitations, and appropriateness to various research endeavors, largely through contrasting their mathematical frameworks. Finally, promising future applications of RAMs are discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Microbiome modeling: a beginner's guide
- Author
-
Emanuel Lange, Lena Kranert, Jacob Krüger, Dirk Benndorf, and Robert Heyer
- Subjects
systems microbiology ,microbial ecology ,omics data integration ,human microbiome ,genome-scale modeling ,constraint-based modeling ,Microbiology ,QR1-502 - Abstract
Microbiomes, comprised of diverse microbial species and viruses, play pivotal roles in human health, environmental processes, and biotechnological applications and interact with each other, their environment, and hosts via ecological interactions. Our understanding of microbiomes is still limited and hampered by their complexity. A concept improving this understanding is systems biology, which focuses on the holistic description of biological systems utilizing experimental and computational methods. An important set of such experimental methods are metaomics methods which analyze microbiomes and output lists of molecular features. These lists of data are integrated, interpreted, and compiled into computational microbiome models, to predict, optimize, and control microbiome behavior. There exists a gap in understanding between microbiologists and modelers/bioinformaticians, stemming from a lack of interdisciplinary knowledge. This knowledge gap hinders the establishment of computational models in microbiome analysis. This review aims to bridge this gap and is tailored for microbiologists, researchers new to microbiome modeling, and bioinformaticians. To achieve this goal, it provides an interdisciplinary overview of microbiome modeling, starting with fundamental knowledge of microbiomes, metaomics methods, common modeling formalisms, and how models facilitate microbiome control. It concludes with guidelines and repositories for modeling. Each section provides entry-level information, example applications, and important references, serving as a valuable resource for comprehending and navigating the complex landscape of microbiome research and modeling.
- Published
- 2024
- Full Text
- View/download PDF
5. A benchmark of optimization solvers for genome-scale metabolic modeling of organisms and communities
- Author
-
Daniel Machado
- Subjects
genome-scale modeling ,metabolism ,optimization methods ,Microbiology ,QR1-502 - Abstract
ABSTRACTGenome-scale metabolic modeling is a powerful framework for predicting metabolic phenotypes of any organism with an annotated genome. For two decades, this framework has been used for the rational design of microbial cell factories. In the last decade, the range of applications has exploded, and new frontiers have emerged, including the study of the gut microbiome and its health implications and the role of microbial communities in global ecosystems. However, all the critical steps in this framework, from model construction to simulation, require the use of powerful linear optimization solvers, with the choice often relying on commercial solvers for their well-known computational efficiency. In this work, I benchmark a total of six solvers (two commercial and four open source) and measure their performance to solve linear and mixed-integer linear problems of increasing complexity. Although commercial solvers are still the fastest, at least two open-source solvers show comparable performance. These results show that genome-scale metabolic modeling does not need to be hindered by commercial licensing schemes and can become a truly open science framework for solving urgent societal challenges.IMPORTANCEModeling the metabolism of organisms and communities allows for computational exploration of their metabolic capabilities and testing their response to genetic and environmental perturbations. This holds the potential to address multiple societal issues related to human health and the environment. One of the current limitations is the use of commercial optimization solvers with restrictive licenses for academic and non-academic use. This work compares the performance of several commercial and open-source solvers to solve some of the most complex problems in the field. Benchmarking results show that, although commercial solvers are indeed faster, some of the open-source options can also efficiently tackle the hardest problems, showing great promise for the development of open science applications.
- Published
- 2024
- Full Text
- View/download PDF
6. Current State, Challenges, and Opportunities in Genome-Scale Resource Allocation Models: A Mathematical Perspective
- Author
-
Wheaton L. Schroeder, Patrick F. Suthers, Thomas C. Willis, Eric J. Mooney, and Costas D. Maranas
- Subjects
systems biology ,computational biology ,genome-scale modeling ,Microbiology ,QR1-502 - Abstract
Stoichiometric genome-scale metabolic models (generally abbreviated GSM, GSMM, or GEM) have had many applications in exploring phenotypes and guiding metabolic engineering interventions. Nevertheless, these models and predictions thereof can become limited as they do not directly account for protein cost, enzyme kinetics, and cell surface or volume proteome limitations. Lack of such mechanistic detail could lead to overly optimistic predictions and engineered strains. Initial efforts to correct these deficiencies were by the application of precursor tools for GSMs, such as flux balance analysis with molecular crowding. In the past decade, several frameworks have been introduced to incorporate proteome-related limitations using a genome-scale stoichiometric model as the reconstruction basis, which herein are called resource allocation models (RAMs). This review provides a broad overview of representative or commonly used existing RAM frameworks. This review discusses increasingly complex models, beginning with stoichiometric models to precursor to RAM frameworks to existing RAM frameworks. RAM frameworks are broadly divided into two categories: coarse-grained and fine-grained, with different strengths and challenges. Discussion includes pinpointing their utility, data needs, highlighting framework strengths and limitations, and appropriateness to various research endeavors, largely through contrasting their mathematical frameworks. Finally, promising future applications of RAMs are discussed.
- Published
- 2024
- Full Text
- View/download PDF
7. Optimizing Strategies for Bio-Based Ethanol Production Using Genome-Scale Metabolic Modeling of the Hyperthermophilic Archaeon, Pyrococcus furiosus.
- Author
-
Vailionis, Jason L., Weishu Zhao, Ke Zhang, Rodionov, Dmitry A., Lipscomb, Gina L., Tanwee, Tania N. N., O'Quinn, Hailey C., Bing, Ryan G., Kelly, Robert M., Adams, Michael W. W., and Ying Zhang
- Subjects
- *
PYROCOCCUS furiosus , *METABOLIC models , *AMINO acid metabolism , *CLIMATE change , *ADENOSINE triphosphatase , *ETHANOL - Abstract
A genome-scale metabolic model, encompassing a total of 623 genes, 727 reactions, and 865 metabolites, was developed for Pyrococcus furiosus, an archaeon that grows optimally at 100°C by carbohydrate and peptide fermentation. The model uses subsystem-based genome annotation, along with extensive manual curation of 237 genereaction associations including those involved in central carbon metabolism, amino acid metabolism, and energy metabolism. The redox and energy balance of P. furiosus was investigated through random sampling of flux distributions in the model during growth on disaccharides. The core energy balance of the model was shown to depend on high acetate production and the coupling of a sodium-dependent ATP synthase and membrane-bound hydrogenase, which generates a sodium gradient in a ferredoxin-dependent manner, aligning with existing understanding of P. furiosus metabolism. The model was utilized to inform genetic engineering designs that favor the production of ethanol over acetate by implementing an NADPH and CO-dependent energy economy. The P. furiosus model is a powerful tool for understanding the relationship between generation of end products and redox/energy balance at a systems-level that will aid in the design of optimal engineering strategies for production of bio-based chemicals and fuels. IMPORTANCE The bio-based production of organic chemicals provides a sustainable alternative to fossil-based production in the face of today's climate challenges. In this work, we present a genome-scale metabolic reconstruction of Pyrococcus furiosus, a wellestablished platform organism that has been engineered to produce a variety of chemicals and fuels. The metabolic model was used to design optimal engineering strategies to produce ethanol. The redox and energy balance of P. furiosus was examined in detail, which provided useful insights that will guide future engineering designs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Enzyme promiscuity shapes adaptation to novel growth substrates.
- Author
-
Guzmán, Gabriela I, Sandberg, Troy E, LaCroix, Ryan A, Nyerges, Ákos, Papp, Henrietta, de Raad, Markus, King, Zachary A, Hefner, Ying, Northen, Trent R, Notebaart, Richard A, Pál, Csaba, Palsson, Bernhard O, Papp, Balázs, and Feist, Adam M
- Subjects
Escherichia coli K12 ,Succinates ,Tartrates ,Enzymes ,Deoxyribose ,Arabinose ,Escherichia coli Proteins ,Adaptation ,Physiological ,Evolution ,Molecular ,Substrate Specificity ,Mutation ,Computer Simulation ,adaptive evolution ,enzyme promiscuity ,genome‐scale modeling ,systems biology ,genome-scale modeling ,Bioinformatics ,Biochemistry and Cell Biology ,Other Biological Sciences - Abstract
Evidence suggests that novel enzyme functions evolved from low-level promiscuous activities in ancestral enzymes. Yet, the evolutionary dynamics and physiological mechanisms of how such side activities contribute to systems-level adaptations are not well characterized. Furthermore, it remains untested whether knowledge of an organism's promiscuous reaction set, or underground metabolism, can aid in forecasting the genetic basis of metabolic adaptations. Here, we employ a computational model of underground metabolism and laboratory evolution experiments to examine the role of enzyme promiscuity in the acquisition and optimization of growth on predicted non-native substrates in Escherichia coli K-12 MG1655. After as few as approximately 20 generations, evolved populations repeatedly acquired the capacity to grow on five predicted non-native substrates-D-lyxose, D-2-deoxyribose, D-arabinose, m-tartrate, and monomethyl succinate. Altered promiscuous activities were shown to be directly involved in establishing high-efficiency pathways. Structural mutations shifted enzyme substrate turnover rates toward the new substrate while retaining a preference for the primary substrate. Finally, genes underlying the phenotypic innovations were accurately predicted by genome-scale model simulations of metabolism with enzyme promiscuity.
- Published
- 2019
9. Predicting the metabolic capabilities of Synechococcus elongatus PCC 7942 adapted to different light regimes.
- Author
-
Broddrick, Jared T, Welkie, David G, Jallet, Denis, Golden, Susan S, Peers, Graham, and Palsson, Bernhard O
- Subjects
Synechococcus ,Oxygen ,Butylene Glycols ,Chlorophyll ,Pigmentation ,Acclimatization ,Photosynthesis ,Energy Metabolism ,Genome ,Light ,Computer Simulation ,Metabolic Engineering ,Metabolic Flux Analysis ,Constraint based modeling ,Cyanobacteria engineering ,Flux balance analysis ,Genome-scale modeling ,Synechococcus elongatus ,Bioengineering ,Biotechnology ,Industrial Biotechnology - Abstract
There is great interest in engineering photoautotrophic metabolism to generate bioproducts of societal importance. Despite the success in employing genome-scale modeling coupled with flux balance analysis to engineer heterotrophic metabolism, the lack of proper constraints necessary to generate biologically realistic predictions has hindered broad application of this methodology to phototrophic metabolism. Here we describe a methodology for constraining genome-scale models of photoautotrophy in the cyanobacteria Synechococcus elongatus PCC 7942. Experimental photophysiology parameters coupled to genome-scale flux balance analysis resulted in accurate predictions of growth rates and metabolic reaction fluxes at low and high light conditions. Additionally, by constraining photon uptake fluxes, we characterized the metabolic cost of excess excitation energy. The predicted energy fluxes were consistent with known light-adapted phenotypes in cyanobacteria. Finally, we leveraged the modeling framework to characterize existing photoautotrophic and photomixtotrophic engineering strategies for 2,3-butanediol production in S. elongatus. This methodology, applicable to genome-scale modeling of all phototrophic microorganisms, can facilitate the use of flux balance analysis in the engineering of light-driven metabolism.
- Published
- 2019
10. Recent advances in culture medium design for enhanced production of monoclonal antibodies in CHO cells: A comparative study of machine learning and systems biology approaches.
- Author
-
Kavoni, Hossein, Savizi, Iman Shahidi Pour, Lewis, Nathan E., and Shojaosadati, Seyed Abbas
- Subjects
- *
CHO cell , *CELL culture , *PHARMACEUTICAL biotechnology industry , *ANTIBODY formation , *CELL growth - Abstract
The production of monoclonal antibodies (mAbs) using Chinese Hamster Ovary (CHO) cells has revolutionized the treatment of numerous diseases, solidifying their position as a cornerstone of the biopharmaceutical industry. However, achieving maximum mAb production while upholding strict product quality standards remains a significant hurdle. Optimizing cell culture media emerges as a critical factor in this endeavor, requiring a nuanced understanding of the complex interplay of nutrients, growth factors, and other components that profoundly influence cellular growth, productivity, and product quality. Significant strides have been made in media optimization, including techniques such as media blending, one factor at a time, and statistical design of experiments approaches. The present review provides a comprehensive analysis of the recent advancements in culture media design strategies, focusing on the comparative application of systems biology (SB) and machine learning (ML) approaches. The applications of SB and ML in optimizing CHO cell culture medium and successful examples of their use are summarized. Finally, we highlight the immense potential of integrating SB and ML, emphasizing the development of hybrid models that leverage the strengths of both approaches for robust, efficient, and scalable optimization of mAb production in CHO cells. This review provides a roadmap for researchers and industry professionals to navigate the complex landscape of mAb production optimization, paving the way for developing next-generation CHO cell culture media that drive significant improvements in yield and productivity. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. Metabolic Robustness to Growth Temperature of a Cold- Adapted Marine Bacterium
- Author
-
Christopher Riccardi, Marzia Calvanese, Veronica Ghini, Tania Alonso-Vásquez, Elena Perrin, Paola Turano, Giorgio Giurato, Alessandro Weisz, Ermenegilda Parrilli, Maria Luisa Tutino, and Marco Fondi
- Subjects
cold-adaptation ,genome-scale modeling ,metabolomics ,transcriptomics ,Microbiology ,QR1-502 - Abstract
ABSTRACT Microbial communities experience continuous environmental changes, with temperature fluctuations being the most impacting. This is particularly important considering the ongoing global warming but also in the “simpler” context of seasonal variability of sea-surface temperature. Understanding how microorganisms react at the cellular level can improve our understanding of their possible adaptations to a changing environment. In this work, we investigated the mechanisms through which metabolic homeostasis is maintained in a cold-adapted marine bacterium during growth at temperatures that differ widely (15 and 0°C). We have quantified its intracellular and extracellular central metabolomes together with changes occurring at the transcriptomic level in the same growth conditions. This information was then used to contextualize a genome-scale metabolic reconstruction, and to provide a systemic understanding of cellular adaptation to growth at 2 different temperatures. Our findings indicate a strong metabolic robustness at the level of the main central metabolites, counteracted by a relatively deep transcriptomic reprogramming that includes changes in gene expression of hundreds of metabolic genes. We interpret this as a transcriptomic buffering of cellular metabolism, able to produce overlapping metabolic phenotypes, despite the wide temperature gap. Moreover, we show that metabolic adaptation seems to be mostly played at the level of few key intermediates (e.g., phosphoenolpyruvate) and in the cross talk between the main central metabolic pathways. Overall, our findings reveal a complex interplay at gene expression level that contributes to the robustness/resilience of core metabolism, also promoting the leveraging of state-of-the-art multi-disciplinary approaches to fully comprehend molecular adaptations to environmental fluctuations. IMPORTANCE This manuscript addresses a central and broad interest topic in environmental microbiology, i.e. the effect of growth temperature on microbial cell physiology. We investigated if and how metabolic homeostasis is maintained in a cold-adapted bacterium during growth at temperatures that differ widely and that match measured changes on the field. Our integrative approach revealed an extraordinary robustness of the central metabolome to growth temperature. However, this was counteracted by deep changes at the transcriptional level, and especially in the metabolic part of the transcriptome. This conflictual scenario was interpreted as a transcriptomic buffering of cellular metabolism, and was investigated using genome-scale metabolic modeling. Overall, our findings reveal a complex interplay at gene expression level that contributes to the robustness/resilience of core metabolism, also promoting the use of state-of-the-art multi-disciplinary approaches to fully comprehend molecular adaptations to environmental fluctuations.
- Published
- 2023
- Full Text
- View/download PDF
12. Integration of physiologically relevant photosynthetic energy flows into whole genome models of light‐driven metabolism.
- Author
-
Broddrick, Jared T., Ware, Maxwell A., Jallet, Denis, Palsson, Bernhard O., and Peers, Graham
- Subjects
- *
PHAEODACTYLUM tricornutum , *METABOLIC models , *CHLOROPHYLL spectra , *ELECTRON transport , *EXCESS electrons , *LIGHT absorption , *BACTERIAL metabolism - Abstract
SUMMARY: Characterizing photosynthetic productivity is necessary to understand the ecological contributions and biotechnology potential of plants, algae, and cyanobacteria. Light capture efficiency and photophysiology have long been characterized by measurements of chlorophyll fluorescence dynamics. However, these investigations typically do not consider the metabolic network downstream of light harvesting. By contrast, genome‐scale metabolic models capture species‐specific metabolic capabilities but have yet to incorporate the rapid regulation of the light harvesting apparatus. Here, we combine chlorophyll fluorescence parameters defining photosynthetic and non‐photosynthetic yield of absorbed light energy with a metabolic model of the pennate diatom Phaeodactylum tricornutum. This integration increases the model predictive accuracy regarding growth rate, intracellular oxygen production and consumption, and metabolic pathway usage. Through the quantification of excess electron transport, we uncover the sequential activation of non‐radiative energy dissipation processes, cross‐compartment electron shuttling, and non‐photochemical quenching as the rapid photoacclimation strategy in P. tricornutum. Interestingly, the photon absorption thresholds that trigger the transition between these mechanisms were consistent at low and high incident photon fluxes. We use this understanding to explore engineering strategies for rerouting cellular resources and excess light energy towards bioproducts in silico. Overall, we present a methodology for incorporating a common, informative data type into computational models of light‐driven metabolism and show its utilization within the design–build–test–learn cycle for engineering of photosynthetic organisms. Significance Statement: Whole genome models of metabolism are improved by the addition of parameters associated with measured photo‐physiology. The approach is demonstrated in the photosynthetic alga, Phaeodactylum tricornutum, but this approach could be applied to any plant system. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. Genome-scale modeling of Chinese hamster ovary cells by hybrid semi-parametric flux balance analysis.
- Author
-
Ramos, João R. C., Oliveira, Gil P., Dumas, Patrick, and Oliveira, Rui
- Abstract
Flux balance analysis (FBA) is currently the standard method to compute metabolic fluxes in genome-scale networks. Several FBA extensions employing diverse objective functions and/or constraints have been published. Here we propose a hybrid semi-parametric FBA extension that combines mechanistic-level constraints (parametric) with empirical constraints (non-parametric) in the same linear program. A CHO dataset with 27 measured exchange fluxes obtained from 21 reactor experiments served to evaluate the method. The mechanistic constraints were deduced from a reduced CHO-K1 genome-scale network with 686 metabolites, 788 reactions and 210 degrees of freedom. The non-parametric constraints were obtained by principal component analysis of the flux dataset. The two types of constraints were integrated in the same linear program showing comparable computational cost to standard FBA. The hybrid FBA is shown to significantly improve the specific growth rate prediction under different constraints scenarios. A metabolically efficient cell growth feed targeting minimal byproducts accumulation was designed by hybrid FBA. It is concluded that integrating parametric and nonparametric constraints in the same linear program may be an efficient approach to reduce the solution space and to improve the predictive power of FBA methods when critical mechanistic information is missing. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Understanding How Context Influences Function Across Biological Scales in Multicellular Mammalian Systems
- Author
-
Baghdassarian, Hratch Matthew
- Subjects
Bioinformatics ,Immunology ,Cellular biology ,cell-cell communication ,genome-scale modeling ,resource allocation ,single-cell RNA sequencing ,systems immunology - Abstract
In mammalian systems, no cell acts in isolation, but rather coordinates to achieve higher-order function. Such cell behaviors are complex and influenced by context. To comprehensively understand them, we must understand how molecular interactions affect cell phenotypes, and analogously, how cell interactions affect higher-order phenotypes. I begin by examining the role of resource allocation in cellular decision-making processes. I underscore the significance of resource constraints and context in shaping cellular phenotypes and enabling population-level behaviors. My research then pivots to a detailed investigation into a rare systemic inflammatory disorder, Disabling Pansclerotic Morphea (DPM). I report on the discovery of novel variants in the STAT4 gene that are linked to DPM. Leveraging these insights, we propose a successful therapeutic approach using the JAK inhibitor, ruxolitinib, thereby demonstrating the importance of a context-informed genetic understanding in disease management. The JAK-STAT pathway is a key signaling pathway mediating immune cell communication. Thus, I shift the focus of my research to intercellular communication. My novel unsupervised method, Tensor-cell2cell, deciphers complex cell-cell communication patterns across multiple contexts (e.g., time points, disease severities, or spatial contexts). Given the generalizability of this approach to use other communication methods’ outputs as its input, I then introduce a protocol integrating two computational tools, LIANA and Tensor-cell2cell. LIANA is similarly generalizable in that it provides a centralized resource to run many methods, thus providing a natural preceding step to Tensor-cell2cell. The protocol enhances robustness and flexibility in identifying cell-cell communication programs across multiple samples. Finally, I present humanME, a computational tool for generating and analyzing human ME-Models from input metabolic models. This approach refines the prediction accuracy of growth rate and offers unique solutions, highlighting the importance of machinery resources in constraining intracellular activities.Collectively, this body of work leverages omics to provide mechanistic insights to how cellular context impacts interactions and functions in mammalian systems across molecular-, cell-, and tissue-scales. The new methods and tools proposed herein pave the way for more nuanced, context-driven research, underpinning future advancements in human health and disease.
- Published
- 2023
15. Pangenome Flux Balance Analysis Toward Panphenomes
- Author
-
Norsigian, Charles J., Fang, Xin, Palsson, Bernhard O., Monk, Jonathan M., Tettelin, Hervé, editor, and Medini, Duccio, editor
- Published
- 2020
- Full Text
- View/download PDF
16. Genome-scale modeling drives 70-fold improvement of intracellular heme production in Saccharomyces cerevisiae.
- Author
-
Ishchuk, Olena P., Domenzain, Iván, Sánchez, Benjamín J., Muñiz-Paredes, Facundo, Martínez, José L., Nielsen, Jens, and Petranovic, Dina
- Subjects
- *
HEME , *SACCHAROMYCES cerevisiae , *HEMOPROTEINS , *FOOD additives , *OXYGEN carriers - Abstract
Heme is an oxygen carrier and a cofactor of both industrial enzymes and food additives. The intracellular level of free heme is low, which limits the synthesis of heme proteins. Therefore, increasing heme synthesis allows an increased production of heme proteins. Using the genome-scale metabolic model (GEM) Yeast8 for the yeast Saccharomyces cerevisiae, we identified fluxes potentially important to heme synthesis. With this model, in silico simulations highlighted 84 gene targets for balancing biomass and increasing heme production. Of those identified, 76 genes were individually deleted or overexpressed in experiments. Empirically, 40 genes individually increased heme production (up to threefold). Heme was increased by modifying target genes, which not only included the genes involved in heme biosynthesis, but also those involved in glycolysis, pyruvate, Fe-S clusters, glycine, and succinyl-coenzyme A (CoA) metabolism. Next, we developed an algorithmic method for predicting an optimal combination of these genes by using the enzyme-constrained extension of the Yeast8 model, ecYeast8. The computationally identified combination for enhanced heme production was evaluated using the heme ligand-binding biosensor (Heme-LBB). The positive targets were combined using CRISPR-Cas9 in the yeast strain (IMX581-HEM15-HEM14-HEM3-Δshm1-HEM2-Δhmx1-FET4-Δgcv2-HEM1-Δgcv1-HEM13), which produces 70-fold-higher levels of intracellular heme. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
17. Modeling and analysis of the rapid aerobic metabolism of Geobacillus sp. LC300
- Author
-
Ljungqvist, Emil E. and Ljungqvist, Emil E.
- Abstract
To mitigate climate change, global greenhouse gas emissions must be halved before 2030. To achieve this goal, alternative routes for fuel and chemical production that do not rely on fossil resources must be explored. Industrial biotechnology has been identified as a key technology in this transition, allowing the sustainable valorization of biomass to biofuels and biochemicals. Geobacillus sp. LC300 is a thermophilic microorganism displaying remarkable growth rates and metabolic capabilities, thus showing promise for development into a microbial cell factory for sustainable production of biochemicals. However, the metabolism of the organism is unexplored, and its metabolic requirements and optimal growth conditions unknown. The aim of this thesis was to investigate the fast metabolism of Geobacillus sp. LC300 and thereby evaluate the potential and facilitate the development of the organism as a microbial cell factory. To explore the metabolic landscape of G. sp. LC300, a homology-based genome-scale metabolic model was constructed. By analyzing the model-predicted metabolic pathways, a prototrophy for all amino acids was predicted, along with an auxotrophy for vitamin B12. Analysis of transporters further predicted growth on several carbon sources, and the model showed accurate predictions of intracellular flux distributions and growth yields on both glucose and xylose. This model serves as a crucial tool for understanding the G. sp. LC300’s metabolism and guiding metabolic engineering efforts to optimize it for industrial use. Growth media previously used for the cultivation of G. sp. LC300 contained complex components, such as yeast extract, and was unable to support growth to high cell densities. This complicated quantitative studies of metabolism where controlled conditions and high cell densities are important for quantification of rates and yields. A minimal medium was developed based on the biomass composition predicted by the genome-scale model. In this devel, För att motverka klimatförändringar måste de globala utsläppen av växthusgaser halveras innan 2030. För att nå detta mål måste nya produktionsprocesser för bränslen och kemikalier utvecklas som är oberoende av fossila resurser. Industriell bioteknik utgör en nyckel-teknik i denna omställning på grund av dess förutsättningar för omvandling av biomassa till biobränslen och biokemikalier. Geobacillus sp. LC300 är en termofil bakterie som uppvisar påfallande höga tillväxthastigheter och metabol förmåga, vilket gör den lovande för att utvecklas till en mikrobiell cellfabrik för biokemikalieproduktion. Bakteriens metabolism är dock outforskad, och dess näringsbehov och optimala tillväxtförhållanden okända. Målet med denna avhandling var att utforska G. sp. LC300s snabba metabolism och därmed utvärdera dess potential och underlätta dess utveckling till en mikrobiell cellfabrik.En homologibaserad genomskalemodell konstruerades för att utforska dess metabolism. Genom att analysera modellens metabola vägar förutspåddes en prototrofi för alla 20 aminosyror, samt en auxotrofi för vitamin B12. Genom analys av transportprotein kunde tillväxtmöjligheter på flera olika kolkällor även förutspås, och modellen estimerade både intra- och extracellulära reaktions-hastigheter på både glukos och xylos med hög noggrannhet. Modellen är ett viktigt verktyg för att utöka förståelsen för G. sp. LC300s metabolism och som guide vid manipulering av metabolismen för att därmed utveckla organismen till en mikrobiell cellfabrik. De odlingsmedia som tidigare använts för odling av G. sp. LC300 innehåller komplexa komponenter, t. ex. jästextrakt, och saknar näringsinnehåll för odlingar med hög celldensitet. Detta komplicerar kvantitativa studier av metabolismen som kräver precis kontroll över odlingsbetingelser och höga celldensiteter för kvantifiering av hastigheter och utbyten. För att undgå detta problem utvecklades ett minimalt medium med definierad sammansättning, baserat på den cellmassakomposit, QC 2024-08-20
- Published
- 2024
18. Multi-omics Quantification of Species Variation of Escherichia coli Links Molecular Features with Strain Phenotypes
- Author
-
Monk, Jonathan M, Koza, Anna, Campodonico, Miguel A, Machado, Daniel, Seoane, Jose Miguel, Palsson, Bernhard O, Herrgård, Markus J, and Feist, Adam M
- Subjects
Biological Sciences ,Industrial Biotechnology ,Human Genome ,Biotechnology ,Genetics ,Escherichia coli ,Escherichia coli Proteins ,Genome ,Bacterial ,Genomics ,Metabolic Engineering ,Metabolic Networks and Pathways ,Phenotype ,genome-scale modeling ,metabolic engineering ,systems biology ,Biochemistry and Cell Biology ,Biochemistry and cell biology - Abstract
Escherichia coli strains are widely used in academic research and biotechnology. New technologies for quantifying strain-specific differences and their underlying contributing factors promise greater understanding of how these differences significantly impact physiology, synthetic biology, metabolic engineering, and process design. Here, we quantified strain-specific differences in seven widely used strains of E. coli (BL21, C, Crooks, DH5a, K-12 MG1655, K-12 W3110, and W) using genomics, phenomics, transcriptomics, and genome-scale modeling. Metabolic physiology and gene expression varied widely with downstream implications for productivity, product yield, and titer. These differences could be linked to differential regulatory structure. Analyzing high-flux reactions and expression of encoding genes resulted in a correlated and quantitative link between these sets, with strain-specific caveats. Integrated modeling revealed that certain strains are better suited to produce given compounds or express desired constructs considering native expression states of pathways that enable high-production phenotypes. This study yields a framework for quantitatively comparing strains in a species with implications for strain selection.
- Published
- 2016
19. Advances in flux balance analysis by integrating machine learning and mechanism-based models
- Author
-
Ankur Sahu, Mary-Ann Blätke, Jędrzej Jakub Szymański, and Nadine Töpfer
- Subjects
Flux balance analysis ,Genome-scale modeling ,Machine learning ,Kinetic models ,Petri-nets ,Multi-scale modeling ,Biotechnology ,TP248.13-248.65 - Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
20. NCMW: A Python Package to Analyze Metabolic Interactions in the Nasal Microbiome
- Author
-
Manuel Glöckler, Andreas Dräger, and Reihaneh Mostolizadeh
- Subjects
microbial communities ,nasal microbiome ,computational biology ,genome-scale modeling ,constraint-based modeling ,Computer applications to medicine. Medical informatics ,R858-859.7 - Abstract
The human upper respiratory tract is the reservoir of a diverse community of commensals and potential pathogens (pathobionts), including Streptococcus pneumoniae (pneumococcus), Haemophilus influenzae, Moraxella catarrhalis, and Staphylococcus aureus, which occasionally turn into pathogens causing infectious diseases, while the contribution of many nasal microorganisms to human health remains undiscovered. To better understand the composition of the nasal microbiome community, we create a workflow of the community model, which mimics the human nasal environment. To address this challenge, constraint-based reconstruction of biochemically accurate genome-scale metabolic models (GEMs) networks of microorganisms is mandatory. Our workflow applies constraint-based modeling (CBM), simulates the metabolism between species in a given microbiome, and facilitates generating novel hypotheses on microbial interactions. Utilizing this workflow, we hope to gain a better understanding of interactions from the metabolic modeling perspective. This article presents nasal community modeling workflow (NCMW)—a python package based on GEMs of species as a starting point for understanding the composition of the nasal microbiome community. The package is constructed as a step-by-step mathematical framework for metabolic modeling and analysis of the nasal microbial community. Using constraint-based models reduces the need for culturing species in vitro, a process that is not convenient in the environment of human noses.Availability: NCMW is freely available on the Python Package Index (PIP) via pip install NCMW. The source code, documentation, and usage examples (Jupyter Notebook and example files) are available at https://github.com/manuelgloeckler/ncmw.
- Published
- 2022
- Full Text
- View/download PDF
21. Systems-Level Modeling for CRISPR-Based Metabolic Engineering.
- Author
-
Cardiff RAL, Carothers JM, Zalatan JG, and Sauro HM
- Subjects
- RNA, Guide, CRISPR-Cas Systems genetics, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Gene Editing methods, Metabolic Engineering methods, CRISPR-Cas Systems genetics
- Abstract
The CRISPR-Cas system has enabled the development of sophisticated, multigene metabolic engineering programs through the use of guide RNA-directed activation or repression of target genes. To optimize biosynthetic pathways in microbial systems, we need improved models to inform design and implementation of transcriptional programs. Recent progress has resulted in new modeling approaches for identifying gene targets and predicting the efficacy of guide RNA targeting. Genome-scale and flux balance models have successfully been applied to identify targets for improving biosynthetic production yields using combinatorial CRISPR-interference (CRISPRi) programs. The advent of new approaches for tunable and dynamic CRISPR activation (CRISPRa) promises to further advance these engineering capabilities. Once appropriate targets are identified, guide RNA prediction models can lead to increased efficacy in gene targeting. Developing improved models and incorporating approaches from machine learning may be able to overcome current limitations and greatly expand the capabilities of CRISPR-Cas9 tools for metabolic engineering.
- Published
- 2024
- Full Text
- View/download PDF
22. Model-driven discovery of underground metabolic functions in Escherichia coli.
- Author
-
Guzmán, Gabriela, Utrilla, José, Nurk, Sergey, Brunk, Elizabeth, Monk, Jonathan, Ebrahim, Ali, Palsson, Bernhard, and Feist, Adam
- Subjects
genome-scale modeling ,isozyme discovery ,substrate promiscuity ,systems biology ,underground metabolism ,Escherichia coli ,Genome ,Bacterial ,Models ,Biological - Abstract
Enzyme promiscuity toward substrates has been discussed in evolutionary terms as providing the flexibility to adapt to novel environments. In the present work, we describe an approach toward exploring such enzyme promiscuity in the space of a metabolic network. This approach leverages genome-scale models, which have been widely used for predicting growth phenotypes in various environments or following a genetic perturbation; however, these predictions occasionally fail. Failed predictions of gene essentiality offer an opportunity for targeting biological discovery, suggesting the presence of unknown underground pathways stemming from enzymatic cross-reactivity. We demonstrate a workflow that couples constraint-based modeling and bioinformatic tools with KO strain analysis and adaptive laboratory evolution for the purpose of predicting promiscuity at the genome scale. Three cases of genes that are incorrectly predicted as essential in Escherichia coli--aspC, argD, and gltA--are examined, and isozyme functions are uncovered for each to a different extent. Seven isozyme functions based on genetic and transcriptional evidence are suggested between the genes aspC and tyrB, argD and astC, gabT and puuE, and gltA and prpC. This study demonstrates how a targeted model-driven approach to discovery can systematically fill knowledge gaps, characterize underground metabolism, and elucidate regulatory mechanisms of adaptation in response to gene KO perturbations.
- Published
- 2015
23. Optimizing cultivation of Cordyceps militaris for fast growth and cordycepin overproduction using rational design of synthetic media
- Author
-
Nachon Raethong, Hao Wang, Jens Nielsen, and Wanwipa Vongsangnak
- Subjects
Cordycepin ,Cordyceps militaris ,Genome-scale modeling ,Synthetic media design ,Systems biology ,Biotechnology ,TP248.13-248.65 - Abstract
Cordyceps militaris is an entomopathogenic fungus which is often used in Asia as a traditional medicine developed from age-old wisdom. Presently, cordycepin from C. militaris is a great interest in medicinal applications. However, cellular growth of C. militaris and the association with cordycepin production remain poorly understood. To explore the metabolism of C. militaris as potential cell factories in medical and biotechnology applications, this study developed a high-quality genome-scale metabolic model of C. militaris, iNR1329, based on its genomic content and physiological data. The model included a total of 1329 genes, 1821 biochemical reactions, and 1171 metabolites among 4 different cellular compartments. Its in silico growth simulation results agreed well with experimental data on different carbon sources. iNR1329 was further used for optimizing the growth and cordycepin overproduction using a novel approach, POPCORN, for rational design of synthetic media. In addition to the high-quality GEM iNR1329, the presented POPCORN approach was successfully used to rationally design an optimal synthetic medium with C:N ratio of 8:1 for enhancing 3.5-fold increase in cordycepin production. This study thus provides a novel insight into C. militaris physiology and highlights a potential GEM-driven method for synthetic media design and metabolic engineering application. The iNR1329 and the POPCORN approach are available at the GitHub repository: https://github.com/sysbiomics/Cordyceps_militaris-GEM.
- Published
- 2020
- Full Text
- View/download PDF
24. In silico identification of metabolic engineering strategies for improved lipid production in Yarrowia lipolytica by genome-scale metabolic modeling
- Author
-
Minsuk Kim, Beom Gi Park, Eun-Jung Kim, Joonwon Kim, and Byung-Gee Kim
- Subjects
Genome-scale modeling ,Systems biology ,Metabolic engineering ,Yarrowia lipolytica ,eMOMA ,Lipid ,Fuel ,TP315-360 ,Biotechnology ,TP248.13-248.65 - Abstract
Abstract Background Yarrowia lipolytica, an oleaginous yeast, is a promising platform strain for production of biofuels and oleochemicals as it can accumulate a high level of lipids in response to nitrogen limitation. Accordingly, many metabolic engineering efforts have been made to develop engineered strains of Y. lipolytica with higher lipid yields. Genome-scale model of metabolism (GEM) is a powerful tool for identifying novel genetic designs for metabolic engineering. Several GEMs for Y. lipolytica have recently been developed; however, not many applications of the GEMs have been reported for actual metabolic engineering of Y. lipolytica. The major obstacle impeding the application of Y. lipolytica GEMs is the lack of proper methods for predicting phenotypes of the cells in the nitrogen-limited condition, or more specifically in the stationary phase of a batch culture. Results In this study, we showed that environmental version of minimization of metabolic adjustment (eMOMA) can be used for predicting metabolic flux distribution of Y. lipolytica under the nitrogen-limited condition and identifying metabolic engineering strategies to improve lipid production in Y. lipolytica. Several well-characterized overexpression targets, such as diglyceride acyltransferase, acetyl-CoA carboxylase, and stearoyl-CoA desaturase, were successfully rediscovered by our eMOMA-based design method, showing the relevance of prediction results. Interestingly, the eMOMA-based design method also suggested non-intuitive knockout targets, and we experimentally validated the prediction with a mutant lacking YALI0F30745g, one of the predicted targets involved in one-carbon/methionine metabolism. The mutant accumulated 45% more lipids compared to the wild-type. Conclusion This study demonstrated that eMOMA is a powerful computational method for understanding and engineering the metabolism of Y. lipolytica and potentially other oleaginous microorganisms.
- Published
- 2019
- Full Text
- View/download PDF
25. Dynamic genome-scale cell-specific metabolic models reveal novel inter-cellular and intra-cellular metabolic communications during ovarian follicle development
- Author
-
Beatriz Peñalver Bernabé, Ines Thiele, Eugene Galdones, Anaar Siletz, Sriram Chandrasekaran, Teresa K. Woodruff, Linda J. Broadbelt, and Lonnie D. Shea
- Subjects
Ovarian follicle development ,Metabolism ,Metabolic communities ,Secreted metabolites ,Cell-type specific metabolic models ,Genome-scale modeling ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background The maturation of the female germ cell, the oocyte, requires the synthesis and storing of all the necessary metabolites to support multiple divisions after fertilization. Oocyte maturation is only possible in the presence of surrounding, diverse, and changing layers of somatic cells. Our understanding of metabolic interactions between the oocyte and somatic cells has been limited due to dynamic nature of ovarian follicle development, thus warranting a systems approach. Results Here, we developed a genome-scale metabolic model of the mouse ovarian follicle. This model was constructed using an updated mouse general metabolic model (Mouse Recon 2) and contains several key ovarian follicle development metabolic pathways. We used this model to characterize the changes in the metabolism of each follicular cell type (i.e., oocyte, granulosa cells, including cumulus and mural cells), during ovarian follicle development in vivo. Using this model, we predicted major metabolic pathways that are differentially active across multiple follicle stages. We identified a set of possible secreted and consumed metabolites that could potentially serve as biomarkers for monitoring follicle development, as well as metabolites for addition to in vitro culture media that support the growth and maturation of primordial follicles. Conclusions Our systems approach to model follicle metabolism can guide future experimental studies to validate the model results and improve oocyte maturation approaches and support growth of primordial follicles in vitro.
- Published
- 2019
- Full Text
- View/download PDF
26. Enzyme promiscuity shapes adaptation to novel growth substrates
- Author
-
Gabriela I Guzmán, Troy E Sandberg, Ryan A LaCroix, Ákos Nyerges, Henrietta Papp, Markus de Raad, Zachary A King, Ying Hefner, Trent R Northen, Richard A Notebaart, Csaba Pál, Bernhard O Palsson, Balázs Papp, and Adam M Feist
- Subjects
adaptive evolution ,enzyme promiscuity ,genome‐scale modeling ,systems biology ,Biology (General) ,QH301-705.5 ,Medicine (General) ,R5-920 - Abstract
Abstract Evidence suggests that novel enzyme functions evolved from low‐level promiscuous activities in ancestral enzymes. Yet, the evolutionary dynamics and physiological mechanisms of how such side activities contribute to systems‐level adaptations are not well characterized. Furthermore, it remains untested whether knowledge of an organism's promiscuous reaction set, or underground metabolism, can aid in forecasting the genetic basis of metabolic adaptations. Here, we employ a computational model of underground metabolism and laboratory evolution experiments to examine the role of enzyme promiscuity in the acquisition and optimization of growth on predicted non‐native substrates in Escherichia coli K‐12 MG1655. After as few as approximately 20 generations, evolved populations repeatedly acquired the capacity to grow on five predicted non‐native substrates—D‐lyxose, D‐2‐deoxyribose, D‐arabinose, m‐tartrate, and monomethyl succinate. Altered promiscuous activities were shown to be directly involved in establishing high‐efficiency pathways. Structural mutations shifted enzyme substrate turnover rates toward the new substrate while retaining a preference for the primary substrate. Finally, genes underlying the phenotypic innovations were accurately predicted by genome‐scale model simulations of metabolism with enzyme promiscuity.
- Published
- 2019
- Full Text
- View/download PDF
27. Enhancing Metabolic Models with Genome-Scale Experimental Data
- Author
-
Jensen, Kristian, Gudmundsson, Steinn, Herrgård, Markus J., Barciszewski, Jan, Series Editor, Rajewsky, Nikolaus, Series Editor, Erdmann, Volker A., Founding Editor, and Jurga, Stefan, editor
- Published
- 2018
- Full Text
- View/download PDF
28. Enabling rational gut microbiome manipulations by understanding gut ecology through experimentally-evidenced in silico models
- Author
-
Juan P. Molina Ortiz, Dale D. McClure, Erin R. Shanahan, Fariba Dehghani, Andrew J. Holmes, and Mark N. Read
- Subjects
precision medicine ,dietary intervention ,gut microbial ecology ,host–microbiome interactions ,genome-scale modeling ,agent-based modeling ,microbial culturing ,computational microbiology ,systems biology ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
The gut microbiome has emerged as a contributing factor in non-communicable disease, rendering it a target of health-promoting interventions. Yet current understanding of the host-microbiome dynamic is insufficient to predict the variation in intervention outcomes across individuals. We explore the mechanisms that underpin the gut bacterial ecosystem and highlight how a more complete understanding of this ecology will enable improved intervention outcomes. This ecology varies within the gut over space and time. Interventions disrupt these processes, with cascading consequences throughout the ecosystem. In vivo studies cannot isolate and probe these processes at the required spatiotemporal resolutions, and in vitro studies lack the representative complexity required. However, we highlight that, together, both approaches can inform in silico models that integrate cellular-level dynamics, can extrapolate to explain bacterial community outcomes, permit experimentation and observation over ecological processes at high spatiotemporal resolution, and can serve as predictive platforms on which to prototype interventions. Thus, it is a concerted integration of these techniques that will enable rational targeted manipulations of the gut ecosystem.
- Published
- 2021
- Full Text
- View/download PDF
29. Quantitative analysis of amino acid metabolism in liver cancer links glutamate excretion to nucleotide synthesis.
- Author
-
Nilsson, Avlant, Haanstr, Jurgen R., Engqvist, Martin, Gerding, Albert, Bakker, Barbara M., Klingmüller, Ursula, Teusink, Bas, and Nielsen, Jens
- Subjects
- *
NUCLEOTIDE synthesis , *AMINO acid analysis , *AMINO acid metabolism , *LIVER cancer , *GLUTAMIC acid - Abstract
Many cancer cells consume glutamine at high rates; counterintuitively, they simultaneously excrete glutamate, the first intermediate in glutamine metabolism. Glutamine consumption has been linked to replenishment of tricarboxylic acid cycle (TCA) intermediates and synthesis of adenosine triphosphate (ATP), but the reason for glutamate excretion is unclear. Here, we dynamically profile the uptake and excretion fluxes of a liver cancer cell line (HepG2) and use genome-scale metabolic modeling for in-depth analysis. We find that up to 30% of the glutamine is metabolized in the cytosol, primarily for nucleotide synthesis, producing cytosolic glutamate. We hypothesize that excreting glutamate helps the cell to increase the nucleotide synthesis rate to sustain growth. Indeed, we show experimentally that partial inhibition of glutamate excretion reduces cell growth. Our integrative approach thus links glutamine addiction to glutamate excretion in cancer and points toward potential drug targets. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
30. The common message of constraint-based optimization approaches: overflow metabolism is caused by two growth-limiting constraints.
- Author
-
de Groot, Daan H., Lischke, Julia, Muolo, Riccardo, Planqué, Robert, Bruggeman, Frank J., and Teusink, Bas
- Subjects
- *
METABOLISM , *SACCHAROMYCES cerevisiae , *METABOLIC models , *ESCHERICHIA coli , *CANCER cells - Abstract
Living cells can express different metabolic pathways that support growth. The criteria that determine which pathways are selected in which environment remain unclear. One recurrent selection is overflow metabolism: the simultaneous usage of an ATP-efficient and -inefficient pathway, shown for example in Escherichia coli, Saccharomyces cerevisiae and cancer cells. Many models, based on different assumptions, can reproduce this observation. Therefore, they provide no conclusive evidence which mechanism is causing overflow metabolism. We compare the mathematical structure of these models. Although ranging from flux balance analyses to self-fabricating metabolism and expression models, we can rewrite all models into one standard form. We conclude that all models predict overflow metabolism when two, model-specific, growth-limiting constraints are hit. This is consistent with recent theory. Thus, identifying these two constraints is essential for understanding overflow metabolism. We list all imposed constraints by these models, so that they can hopefully be tested in future experiments. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
31. Genome-Scale Metabolic Modeling of Chitin-Degrading Microbial Systems
- Author
-
Kessell, Aimee Kristin
- Subjects
- Chitin degradation, Community modeling, Genome-scale modeling, Metabolic modeling, Microbial communities, Microbiological enginneering
- Abstract
As a major component of fungal cell walls and exoskeletons of invertebrates, chitin is widespread in soils, constituting the second most abundant biopolymer in nature. Composed of N-acetyl-D-glucosamine chains, it serves as a vital source of nutrients, including both carbon and nitrogen, for the growth of microorganisms. A solid understanding of the microbial degradation of chitin is critical for predicting their impacts on biogeochemical cycling in soil ecosystems. Organisms that degrade biopolymers (degraders) produce energetically expensive extracellular enzymes to break down complex organic carbons into simpler labile forms that are sharable with other species, including those that do not contribute directly to the degradation process (cheaters). Therefore, it impacts not only the metabolic and growth efficiencies of the degraders but also fosters diverse interspecies interactions within microbial communities. The level of complexity in this process necessitates the use of mechanistic metabolic models. However, reconstruction of phenotype-consistent genome-scale metabolic networks is still challenging due to the frequent occurrence of false positives (model prediction of biomass production in media where actual organism cannot grow) when gapfilled using typical sequential gapfilling approaches. In this work, I developed a new iterative gapfilling method to address this issue and applied it to build metabolic networks of chitin-degrading communities and their isolates—using a consortium of Cellvibrio japonicus (degrader) and Escherichia coli (non-degrader) as a model system. This new development revealed previously unknown and interesting findings on how bioenergetic cost on chitin degradation affects degrader’s metabolism and its interactions with non-degraders. The model also provided mechanistic interpretations of the predicted changes in metabolism and interactions based on carbon and nitrogen use efficiencies. Both the methods and findings are reproducible, and may be used in other biopolymer-degrading communities.
- Published
- 2024
32. Constrain and conquer: explaining metabolic strategies of microbial life through optimal resource allocation
- Author
-
Grigaitis, Pranas and Grigaitis, Pranas
- Abstract
Optimal allocation of available cellular resources is central for microorganisms to strive and outcompete individuals of the same and other microbial species. Allocation of resources mainly manifests as production of proteins required to sustain growth, and thus shapes the metabolic strategies microbes undertake. In different conditions, different metabolic strategies are superior to others (sustain fastest growth per unit protein), and will be adopted as a result of optimal resource allocation. We have been using computational large-scale, fine-grained resource allocation models to characterize resource allocation strategies in three model microorganisms: Escherichia coli, Saccharomyces cerevisiae, and Schizosaccharomyces pombe. We find that different metabolic strategies are selected in a condition-dependent manner based on both their efficacy per protein and allocation of respective proteins in different cellular compartments. Moreover, we observe that perturbations of these optimal strategies (e.g. forced expression of unneeded protein) come at the cost of decreased growth rate, consistent with existing body of experimental data. Finally, we use these models to test biological hypotheses and thus argue that resource allocation models can be successfully used to identify the metabolic strategies which govern microbial growth.
- Published
- 2023
- Full Text
- View/download PDF
33. The Power of Metabolism for Predicting Microbial Community Dynamics
- Author
-
Jeremy M. Chacón and William R. Harcombe
- Subjects
metabolism ,antibiotics ,bacteriophage ,ecology ,evolution ,genome-scale modeling ,Microbiology ,QR1-502 - Abstract
ABSTRACT Quantitative understanding and prediction of microbial community dynamics are an outstanding challenge. We test the hypothesis that metabolic mechanisms provide a foundation for accurate prediction of dynamics in microbial systems. In our research, metabolic models have been able to accurately predict species interactions, evolutionary trajectories, and response to perturbation in simple synthetic consortia. However, metabolic models have many constraints and often serve best as null models to identify additional processes at play. We anticipate that major advances in metabolic systems biology will involve scaling bottom-up approaches to complex communities and expanding the processes that are incorporated in a metabolic perspective. Ultimately, cellular metabolism will inform predictive ecology that enables precision management of microbial systems.
- Published
- 2019
- Full Text
- View/download PDF
34. Comparative Genome-Scale Metabolic Modeling of Metallo-Beta-Lactamase–Producing Multidrug-Resistant Klebsiella pneumoniae Clinical Isolates
- Author
-
Charles J. Norsigian, Heba Attia, Richard Szubin, Aymen S. Yassin, Bernhard Ø. Palsson, Ramy K. Aziz, and Jonathan M. Monk
- Subjects
multi-drug resistance ,MDR ,Klebsiella pneumoniae ,colistin ,genome-scale modeling ,Microbiology ,QR1-502 - Abstract
The emergence and spread of metallo-beta-lactamase–producing multidrug-resistant (MDR) Klebsiella pneumoniae is a serious public health threat, which is further complicated by the increased prevalence of colistin resistance. The link between antimicrobial resistance acquired by strains of Klebsiella and their unique metabolic capabilities has not been determined. Here, we reconstruct genome-scale metabolic models for 22 K. pneumoniae strains with various resistance profiles to different antibiotics, including two strains exhibiting colistin resistance isolated from Cairo, Egypt. We use the models to predict growth capabilities on 265 different sole carbon, nitrogen, sulfur, and phosphorus sources for all 22 strains. Alternate nitrogen source utilization of glutamate, arginine, histidine, and ethanolamine among others provided discriminatory power for identifying resistance to amikacin, tetracycline, and gentamicin. Thus, genome-scale model based predictions of growth capabilities on alternative substrates may lead to construction of classification trees that are indicative of antibiotic resistance in Klebsiella isolates.
- Published
- 2019
- Full Text
- View/download PDF
35. A benchmark of optimization solvers for genome-scale metabolic modeling of organisms and communities.
- Author
-
Machado D
- Subjects
- Humans, Ecosystem, Algorithms, Genome, Benchmarking, Biochemical Phenomena
- Abstract
Genome-scale metabolic modeling is a powerful framework for predicting metabolic phenotypes of any organism with an annotated genome. For two decades, this framework has been used for the rational design of microbial cell factories. In the last decade, the range of applications has exploded, and new frontiers have emerged, including the study of the gut microbiome and its health implications and the role of microbial communities in global ecosystems. However, all the critical steps in this framework, from model construction to simulation, require the use of powerful linear optimization solvers, with the choice often relying on commercial solvers for their well-known computational efficiency. In this work, I benchmark a total of six solvers (two commercial and four open source) and measure their performance to solve linear and mixed-integer linear problems of increasing complexity. Although commercial solvers are still the fastest, at least two open-source solvers show comparable performance. These results show that genome-scale metabolic modeling does not need to be hindered by commercial licensing schemes and can become a truly open science framework for solving urgent societal challenges.IMPORTANCEModeling the metabolism of organisms and communities allows for computational exploration of their metabolic capabilities and testing their response to genetic and environmental perturbations. This holds the potential to address multiple societal issues related to human health and the environment. One of the current limitations is the use of commercial optimization solvers with restrictive licenses for academic and non-academic use. This work compares the performance of several commercial and open-source solvers to solve some of the most complex problems in the field. Benchmarking results show that, although commercial solvers are indeed faster, some of the open-source options can also efficiently tackle the hardest problems, showing great promise for the development of open science applications., Competing Interests: The author declares no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
36. A Framework Linking Glycolytic Metabolic Capabilities and Tumor Dynamics.
- Author
-
Tzamali, Eleftheria, Tzedakis, Georgios, and Sakkalis, Vangelis
- Subjects
TUMOR growth ,TUMORS ,TUMOR microenvironment ,DENSITY matrices ,CELL tumors - Abstract
Metabolic reprogramming is a hallmark of cancer. The main aim of this paper is to integrate a genome-scale metabolic description of tumor cells into a tumor growth model that accounts for the spatiotemporally heterogeneous tumor microenvironment, in order to study the effects of microscopic characteristics on tumor evolution. A lactate maximization metabolic strategy that allows near-optimal growth solution, while maximizing lactate secretion, is assumed. The proposed sub-cellular metabolic model is then incorporated into a hybrid discrete-continuous model of tumor growth. We produced several phenotypes by applying different constraints and optimization criteria in the metabolic model and explored the tumor evolution of the various phenotypes in different vasculature conditions and extracellular matrix densities. At first, we showed that the metabolic capabilities of phenotypes depending on resource availability can vary in a counter-intuitive manner. We then showed that: first, tumor population, morphology, and spread are affected differently in different conditions, allowing thus phenotypes to be superior than others in different conditions; and second, polyclonal tumors consisting of different phenotypes can exploit their different metabolic capabilities to enhance further tumor evolution. The proposed framework comprises a proof-of-concept demonstration showing the importance of considering the metabolic capabilities of phenotypes on predicting tumor evolution. The proposed framework allows the incorporation of context-specific and patient-specific data for the study of personalized tumor evolution and therapy efficacy, linking genome to metabolic capabilities and tumor dynamics. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
37. Dynamic genome-scale cell-specific metabolic models reveal novel inter-cellular and intra-cellular metabolic communications during ovarian follicle development.
- Author
-
Peñalver Bernabé, Beatriz, Thiele, Ines, Galdones, Eugene, Siletz, Anaar, Chandrasekaran, Sriram, Woodruff, Teresa K., Broadbelt, Linda J., and Shea, Lonnie D.
- Subjects
- *
OVARIAN follicle , *METABOLIC models , *GRANULOSA cells , *GERM cells , *SOMATIC cells , *OVUM - Abstract
Background: The maturation of the female germ cell, the oocyte, requires the synthesis and storing of all the necessary metabolites to support multiple divisions after fertilization. Oocyte maturation is only possible in the presence of surrounding, diverse, and changing layers of somatic cells. Our understanding of metabolic interactions between the oocyte and somatic cells has been limited due to dynamic nature of ovarian follicle development, thus warranting a systems approach. Results: Here, we developed a genome-scale metabolic model of the mouse ovarian follicle. This model was constructed using an updated mouse general metabolic model (Mouse Recon 2) and contains several key ovarian follicle development metabolic pathways. We used this model to characterize the changes in the metabolism of each follicular cell type (i.e., oocyte, granulosa cells, including cumulus and mural cells), during ovarian follicle development in vivo. Using this model, we predicted major metabolic pathways that are differentially active across multiple follicle stages. We identified a set of possible secreted and consumed metabolites that could potentially serve as biomarkers for monitoring follicle development, as well as metabolites for addition to in vitro culture media that support the growth and maturation of primordial follicles. Conclusions: Our systems approach to model follicle metabolism can guide future experimental studies to validate the model results and improve oocyte maturation approaches and support growth of primordial follicles in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
38. Computational inference of the transcriptional regulatory network of Candida glabrata.
- Author
-
Xu, Nan and Liu, Liming
- Abstract
Candida glabrata is a major cause of candidiasis and the second most frequent opportunistic yeast pathogen. Its infectious and antifungal mechanisms are globally regulated by the transcription systems of pathogenic fungi. In this study, we reconstructed the genome-scale transcriptional regulatory network (TRN) of C. glabrata, consisting of 6634 interactive relationships between 145 transcription factors and 3230 target genes, based on genomic and transcriptomic data. The C. glabrata TRN was found to have a typical topological structure and significant network cohesiveness. Moreover, this network could be functionally divided into several sub-networks, including networks involving carbon, nitrogen, growth-associated metabolic profiles, stress response to acidity, hyperosmosis, peroxidation, hypoxia and virulence. Furthermore, by integrating the genome-scale metabolic model of C. glabrata, six essential metabolites and eight related enzymes were systematically selected as drug targets. Overall, elucidation of the genome-scale TRN of C. glabrata has expanded our knowledge of the contents and structures of microbial regulatory networks and improved our understanding of the regulatory behaviors of growth, metabolism and gene expression programs in response to environmental stimuli. Reconstruction of genome-scale transcriptional regulatory network for non-model microbe Candida glabrata and integration with metabolic model for the in silico drug targets. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
39. Comparative Genome-Scale Metabolic Modeling of Metallo-Beta-Lactamase–Producing Multidrug-Resistant Klebsiella pneumoniae Clinical Isolates.
- Author
-
Norsigian, Charles J., Attia, Heba, Szubin, Richard, Yassin, Aymen S., Palsson, Bernhard Ø., Aziz, Ramy K., and Monk, Jonathan M.
- Subjects
KLEBSIELLA pneumoniae ,COLISTIN ,METABOLIC models ,ANTIBIOTICS ,TETRACYCLINES ,KLEBSIELLA - Abstract
The emergence and spread of metallo-beta-lactamase–producing multidrug-resistant (MDR) Klebsiella pneumoniae is a serious public health threat, which is further complicated by the increased prevalence of colistin resistance. The link between antimicrobial resistance acquired by strains of Klebsiella and their unique metabolic capabilities has not been determined. Here, we reconstruct genome-scale metabolic models for 22 K. pneumoniae strains with various resistance profiles to different antibiotics, including two strains exhibiting colistin resistance isolated from Cairo, Egypt. We use the models to predict growth capabilities on 265 different sole carbon, nitrogen, sulfur, and phosphorus sources for all 22 strains. Alternate nitrogen source utilization of glutamate, arginine, histidine, and ethanolamine among others provided discriminatory power for identifying resistance to amikacin, tetracycline, and gentamicin. Thus, genome-scale model based predictions of growth capabilities on alternative substrates may lead to construction of classification trees that are indicative of antibiotic resistance in Klebsiella isolates. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
40. Cross‐compartment metabolic coupling enables flexible photoprotective mechanisms in the diatom Phaeodactylum tricornutum.
- Author
-
Broddrick, Jared T., Du, Niu, Smith, Sarah R., Tsuji, Yoshinori, Jallet, Denis, Ware, Maxwell A., Peers, Graham, Matsuda, Yusuke, Dupont, Chris L., Mitchell, B. Greg, Palsson, Bernhard O., and Allen, Andrew E.
- Subjects
- *
PHAEODACTYLUM tricornutum , *OXYGEN evolution reactions , *AMINO acids , *PLANT photorespiration , *GLUTAMINE - Abstract
Summary: Photoacclimation consists of short‐ and long‐term strategies used by photosynthetic organisms to adapt to dynamic light environments. Observable photophysiology changes resulting from these strategies have been used in coarse‐grained models to predict light‐dependent growth and photosynthetic rates. However, the contribution of the broader metabolic network, relevant to species‐specific strategies and fitness, is not accounted for in these simple models.We incorporated photophysiology experimental data with genome‐scale modeling to characterize organism‐level, light‐dependent metabolic changes in the model diatom Phaeodactylum tricornutum. Oxygen evolution and photon absorption rates were combined with condition‐specific biomass compositions to predict metabolic pathway usage for cells acclimated to four different light intensities.Photorespiration, an ornithine‐glutamine shunt, and branched‐chain amino acid metabolism were hypothesized as the primary intercompartment reductant shuttles for mediating excess light energy dissipation. Additionally, simulations suggested that carbon shunted through photorespiration is recycled back to the chloroplast as pyruvate, a mechanism distinct from known strategies in photosynthetic organisms.Our results suggest a flexible metabolic network in P. tricornutum that tunes intercompartment metabolism to optimize energy transport between the organelles, consuming excess energy as needed. Characterization of these intercompartment reductant shuttles broadens our understanding of energy partitioning strategies in this clade of ecologically important primary producers. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
41. Predicting the metabolic capabilities of Synechococcus elongatus PCC 7942 adapted to different light regimes.
- Author
-
Broddrick, Jared T., Welkie, David G., Jallet, Denis, Golden, Susan S., Peers, Graham, and Palsson, Bernhard O.
- Subjects
- *
SYNECHOCOCCUS elongatus , *CYANOBACTERIA , *PHOTOSYNTHESIS , *PHOTONS , *METABOLISM - Abstract
Abstract There is great interest in engineering photoautotrophic metabolism to generate bioproducts of societal importance. Despite the success in employing genome-scale modeling coupled with flux balance analysis to engineer heterotrophic metabolism, the lack of proper constraints necessary to generate biologically realistic predictions has hindered broad application of this methodology to phototrophic metabolism. Here we describe a methodology for constraining genome-scale models of photoautotrophy in the cyanobacteria Synechococcus elongatus PCC 7942. Experimental photophysiology parameters coupled to genome-scale flux balance analysis resulted in accurate predictions of growth rates and metabolic reaction fluxes at low and high light conditions. Additionally, by constraining photon uptake fluxes, we characterized the metabolic cost of excess excitation energy. The predicted energy fluxes were consistent with known light-adapted phenotypes in cyanobacteria. Finally, we leveraged the modeling framework to characterize existing photoautotrophic and photomixtotrophic engineering strategies for 2,3-butanediol production in S. elongatus. This methodology, applicable to genome-scale modeling of all phototrophic microorganisms, can facilitate the use of flux balance analysis in the engineering of light-driven metabolism. Highlights • Light uptake and oxygen evolution accurately constrain phototrophic metabolism. • Metabolic fluxes predicted by these constraints recapitulate experimental results. • Alternative electron transport and photodamage repair rates are quantified. • Photo- and photomixtotrophic 2,3-butanediol engineering designs are characterized. • The constraints can be extended to genome-scale models of any phototroph. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
42. Understanding FBA Solutions under Multiple Nutrient Limitations
- Author
-
Eunice van Pelt-KleinJan, Daan H. de Groot, and Bas Teusink
- Subjects
flux balance analysis ,elementary flux modes ,elementary conversion modes ,genome-scale modeling ,stoichiometric modeling ,phenotype phase plane analysis ,Microbiology ,QR1-502 - Abstract
Genome-scale stoichiometric modeling methods, in particular Flux Balance Analysis (FBA) and variations thereof, are widely used to investigate cell metabolism and to optimize biotechnological processes. Given (1) a metabolic network, which can be reconstructed from an organism’s genome sequence, and (2) constraints on reaction rates, which may be based on measured nutrient uptake rates, FBA predicts which reactions maximize an objective flux, usually the production of cell components. Although FBA solutions may accurately predict the metabolic behavior of a cell, the actual flux predictions are often hard to interpret. This is especially the case for conditions with many constraints, such as for organisms growing in rich nutrient environments: it remains unclear why a certain solution was optimal. Here, we rationalize FBA solutions by explaining for which properties the optimal combination of metabolic strategies is selected. We provide a graphical formalism in which the selection of solutions can be visualized; we illustrate how this perspective provides a glimpse of the logic that underlies genome-scale modeling by applying our formalism to models of various sizes.
- Published
- 2021
- Full Text
- View/download PDF
43. GEM Modeling Tools for Microbial Consortia Assessment
- Author
-
William T Scott Jr. and Sara Benito-Vaquerizo
- Subjects
Systeem en Synthetische Biologie ,Genome-scale modeling ,Microbial Communities ,Constaint-based modeling ,Unlock ,Systems and Synthetic Biology ,VLAG - Abstract
This is a repository of the scripts used for a manuscript where we evaluated genome-scale modeling tools for microbial communities.
- Published
- 2023
- Full Text
- View/download PDF
44. Flux Balance Analysis of Mammalian Cell Systems.
- Author
-
Morrissey J, Strain B, and Kontoravdi C
- Subjects
- Animals, Cell Line, Mammals
- Abstract
Flux balance analysis (FBA) is a computational methodology to model and analyze the metabolic behavior of cells. In this chapter, we break down the key steps for formulating an FBA model and other FBA-derived methodologies in the context of mammalian cell biology, including strain design, developing cell line-specific models, and conducting flux sampling. We provide annotated COBRApy code for each step to show how it would work in practice., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
45. Toolboxes for cyanobacteria: Recent advances and future direction.
- Author
-
Sun, Tao, Li, Shubin, Song, Xinyu, Diao, Jinjin, Chen, Lei, and Zhang, Weiwen
- Subjects
- *
PHOTOSYNTHETIC cyanobacteria , *MICROBIAL cells , *CARBON dioxide , *SACCHAROMYCES cerevisiae , *GENETIC engineering - Abstract
Photosynthetic cyanobacteria are important primary producers and model organisms for studying photosynthesis and elements cycling on earth. Due to the ability to absorb sunlight and utilize carbon dioxide, cyanobacteria have also been proposed as renewable chassis for carbon-neutral “microbial cell factories”. Recent progresses on cyanobacterial synthetic biology have led to the successful production of more than two dozen of fuels and fine chemicals directly from CO 2 , demonstrating their potential for scale-up application in the future. However, compared with popular heterotrophic chassis like Escherichia coli and Saccharomyces cerevisiae , where abundant genetic tools are available for manipulations at levels from single gene, pathway to whole genome, limited genetic tools are accessible to cyanobacteria. Consequently, this significant technical hurdle restricts both the basic biological researches and further development and application of these renewable systems. Though still lagging the heterotrophic chassis, the vital roles of genetic tools in tuning of gene expression, carbon flux re-direction as well as genome-wide manipulations have been increasingly recognized in cyanobacteria. In recent years, significant progresses on developing and introducing new and efficient genetic tools have been made for cyanobacteria, including promoters, riboswitches, ribosome binding site engineering, clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease (CRISPR/Cas) systems, small RNA regulatory tools and genome-scale modeling strategies. In this review, we critically summarize recent advances on development and applications as well as technical limitations and future directions of the genetic tools in cyanobacteria. In addition, toolboxes feasible for using in large-scale cultivation are also briefly discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
46. Unlocking Human Brain Metabolism by Genome-Scale and Multiomics Metabolic Models: Relevance for Neurology Research, Health, and Disease.
- Author
-
Sertbas, Mustafa and Ulgen, Kutlu O.
- Subjects
- *
METABOLIC models , *NEUROLOGY , *ASTROCYTES , *NEURONS , *PUBLIC health , *NEUROLOGICAL disorders ,BRAIN metabolism - Abstract
Neurology research and clinical practice are transforming toward postgenomics integrative biology. One such example is the study of human brain metabolism that is highly sophisticated due to reactions occurring in and between the astrocytes and neurons. Because of the inherent difficulty of performing experimental studies in human brain, metabolic network modeling has grown in importance to decipher the contribution of brain metabolite kinetics to human health and disease. Multiomics system science-driven metabolic models, using genome-scale and transcriptomics Big Data, offer the promise of new insights on metabolic networks in human brain. Added to this, the availability of omics technologies in both developed and developing world, neurology research, and clinical practice ought to be repositioned with a view to systems medicine. In this expert analysis, we present a critical and in-depth overview of the basic tenets of human brain metabolism, together with the most recent metabolic modeling strategies and computational studies of brain in health and neurological diseases. Human genome-scale metabolic models developed in a both global and brain-specific manner and multiomics synthesis of knowledge are highlighted in particular. We conclude by underscoring the value of multiomics modeling for metabolic diseases and computational investigations of the brain networks, with a view to unlocking the pathophysiology of Alzheimer's disease, Parkinson's disease, migraine, stroke, epilepsy, and multiple sclerosis, among other neurological disorders of importance for global health. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
47. Genome-scale biological models for industrial microbial systems.
- Author
-
Xu, Nan, Ye, Chao, and Liu, Liming
- Subjects
- *
MICROORGANISMS , *FERMENTATION , *CELL growth , *BIOLOGICAL models , *MICROBIOLOGY - Abstract
The primary aims and challenges associated with microbial fermentation include achieving faster cell growth, higher productivity, and more robust production processes. Genome-scale biological models, predicting the formation of an interaction among genetic materials, enzymes, and metabolites, constitute a systematic and comprehensive platform to analyze and optimize the microbial growth and production of biological products. Genome-scale biological models can help optimize microbial growth-associated traits by simulating biomass formation, predicting growth rates, and identifying the requirements for cell growth. With regard to microbial product biosynthesis, genome-scale biological models can be used to design product biosynthetic pathways, accelerate production efficiency, and reduce metabolic side effects, leading to improved production performance. The present review discusses the development of microbial genome-scale biological models since their emergence and emphasizes their pertinent application in improving industrial microbial fermentation of biological products. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
48. Computational approaches to metabolic engineering utilizing systems biology and synthetic biology
- Author
-
Stephen S. Fong
- Subjects
Metabolic engineering ,Genome-scale modeling ,Synthetic biology ,Computational design ,Biotechnology ,TP248.13-248.65 - Abstract
Metabolic engineering modifies cellular function to address various biochemical applications. Underlying metabolic engineering efforts are a host of tools and knowledge that are integrated to enable successful outcomes. Concurrent development of computational and experimental tools has enabled different approaches to metabolic engineering. One approach is to leverage knowledge and computational tools to prospectively predict designs to achieve the desired outcome. An alternative approach is to utilize combinatorial experimental tools to empirically explore the range of cellular function and to screen for desired traits. This mini-review focuses on computational systems biology and synthetic biology tools that can be used in combination for prospective in silico strain design.
- Published
- 2014
- Full Text
- View/download PDF
49. Employing Systems Biology for Discovery and Engineering in Phototrophic Microorganisms
- Author
-
Broddrick, Jared Thomas
- Subjects
Systematic biology ,Cellular biology ,Biochemistry ,Genome-scale modeling ,Microalgae ,Photosynthesis ,Systems biology - Abstract
Global respiratory balance is maintained by photosynthetic organisms, yet the importance of this contribution is not commensurate with our understanding of light-driven metabolic processes. Meanwhile, there has been a increase in the desire to engineer phototrophic microorganisms. Excessive demands by modern society have been depleting nature's resources over the past centuries. Exploring and developing new sustainable resources to counter increasing consumption has therefore been the focus of research efforts in the academic and private sectors. The emphasis has partly been on using phototrophic organisms that fix carbon dioxide by utilizing light energy to produce energy-dense products. Recent efforts to characterize metabolic capabilities of photosynthetic species as well as engineer attractive candidates require a framework for discovery, data analysis and reconfiguring of existing metabolic networks. The systems biology approach of constraint-based reconstruction and analysis coupled with flux balance analysis has a proven record of contextualizing organism specific information and characterizing cellular metabolism. However, a persistent challenge in the modeling of photoautotrophy has been a mechanistic incorporation of light uptake. As the light environment dictates cell physiology, such as growth rate, biomass composition and metabolic pathway usage, constraining photon flux is a prerequisite for biologically accurate results. Here we describe efforts to address this challenge and the resulting insights into photoautotrphic biology. First, a proper accounting of light uptake and shading coupled with a high-quality genome-scale model of the cyanobacteriaSynechococcus elongatus sp. PCC7942 resulted in accurate growth and flux predictions. Additionally,we concluded despite an incomplete TCA cycle in the organism studied, there was no impact to fitness due to the metabolic network configuration. Next, in an attempt to extend the methodology to eukaryotic microalgae, we generated a genome-scale model of the diatom Phaeodactylum tricornutum. The systems biology perspective elucidated metabolic capabilities in this organism conferred by its unique phylogeny. Applying an improved set of photophysiology constraints, we simulated circadian dynamics and characterized photoprotective mechanisms resulting from the previously elucidated metabolic capabilities; highlighting the importance of the broader metabolic network in dissipating excess light energy. Finally, photophysiology constraints coupled to chlorophyll fluorescence measurements and genome-scale modeling enabled a comparative analysis of light environment acclimation across the phototrophic clades cyanobacteria, green algae and diatoms and quantified the fraction of excess light energy absorbed by the cells and its metabolic fate. Overall, coupling mechanistic constraints on photophysiology with genome-scale modeling accurately characterized the cellular response to the light environment. This work is relevant to understanding species-specific metabolic capabilities and adaptations of interest to biological and bioengineering communities.
- Published
- 2018
50. State-of-the-Art Genetic Modalities to Engineer Cyanobacteria for Sustainable Biosynthesis of Biofuel and Fine-Chemicals to Meet Bio–Economy Challenges
- Author
-
Aqib Zafar Khan, Muhammad Bilal, Shahid Mehmood, Ashutosh Sharma, and Hafiz M. N. Iqbal
- Subjects
cyanobacteria ,metabolic engineering ,commodity chemicals ,genome-scale modeling ,metabolic flux analysis ,CRISPR/cas system ,Science - Abstract
In recent years, metabolic engineering of microorganisms has attained much research interest to produce biofuels and industrially pertinent chemicals. Owing to the relatively fast growth rate, genetic malleability, and carbon neutral production process, cyanobacteria has been recognized as a specialized microorganism with a significant biotechnological perspective. Metabolically engineering cyanobacterial strains have shown great potential for the photosynthetic production of an array of valuable native or non-native chemicals and metabolites with profound agricultural and pharmaceutical significance using CO2 as a building block. In recent years, substantial improvements in developing and introducing novel and efficient genetic tools such as genome-scale modeling, high throughput omics analyses, synthetic/system biology tools, metabolic flux analysis and clustered regularly interspaced short palindromic repeats (CRISPR)-associated nuclease (CRISPR/cas) systems have been made for engineering cyanobacterial strains. Use of these tools and technologies has led to a greater understanding of the host metabolism, as well as endogenous and heterologous carbon regulation mechanisms which consequently results in the expansion of maximum productive ability and biochemical diversity. This review summarizes recent advances in engineering cyanobacteria to produce biofuel and industrially relevant fine chemicals of high interest. Moreover, the development and applications of cutting-edge toolboxes such as the CRISPR-cas9 system, synthetic biology, high-throughput “omics”, and metabolic flux analysis to engineer cyanobacteria for large-scale cultivation are also discussed.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.