1
|
Carter EL, Constantinidou C, Alam MT. Applications of genome-scale metabolic models to investigate microbial metabolic adaptations in response to genetic or environmental perturbations. Brief Bioinform 2023; 25:bbad439. [PMID: 38048080 PMCID: PMC10694557 DOI: 10.1093/bib/bbad439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/21/2023] [Accepted: 11/08/2023] [Indexed: 12/05/2023] Open
Abstract
Environmental perturbations are encountered by microorganisms regularly and will require metabolic adaptations to ensure an organism can survive in the newly presenting conditions. In order to study the mechanisms of metabolic adaptation in such conditions, various experimental and computational approaches have been used. Genome-scale metabolic models (GEMs) are one of the most powerful approaches to study metabolism, providing a platform to study the systems level adaptations of an organism to different environments which could otherwise be infeasible experimentally. In this review, we are describing the application of GEMs in understanding how microbes reprogram their metabolic system as a result of environmental variation. In particular, we provide the details of metabolic model reconstruction approaches, various algorithms and tools for model simulation, consequences of genetic perturbations, integration of '-omics' datasets for creating context-specific models and their application in studying metabolic adaptation due to the change in environmental conditions.
Collapse
Affiliation(s)
- Elena Lucy Carter
- Warwick Medical School, University of Warwick, Coventry, CV4 7HL, UK
| | | | | |
Collapse
|
2
|
Vikromvarasiri N, Noda S, Shirai T, Kondo A. Investigation of two metabolic engineering approaches for (R,R)-2,3-butanediol production from glycerol in Bacillus subtilis. J Biol Eng 2023; 17:3. [PMID: 36627686 PMCID: PMC9830791 DOI: 10.1186/s13036-022-00320-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Flux Balance Analysis (FBA) is a well-known bioinformatics tool for metabolic engineering design. Previously, we have successfully used single-level FBA to design metabolic fluxes in Bacillus subtilis to enhance (R,R)-2,3-butanediol (2,3-BD) production from glycerol. OptKnock is another powerful technique for devising gene deletion strategies to maximize microbial growth coupling with improved biochemical production. It has never been used in B. subtilis. In this study, we aimed to compare the use of single-level FBA and OptKnock for designing enhanced 2,3-BD production from glycerol in B. subtilis. RESULTS Single-level FBA and OptKnock were used to design metabolic engineering approaches for B. subtilis to enhance 2,3-BD production from glycerol. Single-level FBA indicated that deletion of ackA, pta, lctE, and mmgA would improve the production of 2,3-BD from glycerol, while OptKnock simulation suggested the deletion of ackA, pta, mmgA, and zwf. Consequently, strains LM01 (single-level FBA-based) and MZ02 (OptKnock-based) were constructed, and their capacity to produce 2,3-BD from glycerol was investigated. The deletion of multiple genes did not negatively affect strain growth and glycerol utilization. The highest 2,3-BD production was detected in strain LM01. Strain MZ02 produced 2,3-BD at a similar level as the wild type, indicating that the OptKnock prediction was erroneous. Two-step FBA was performed to examine the reason for the erroneous OptKnock prediction. Interestingly, we newly found that zwf gene deletion in strain MZ02 improved lactate production, which has never been reported to date. The predictions of single-level FBA for strain MZ02 were in line with experimental findings. CONCLUSIONS We showed that single-level FBA is an effective approach for metabolic design and manipulation to enhance 2,3-BD production from glycerol in B. subtilis. Further, while this approach predicted the phenotypes of generated strains with high precision, OptKnock prediction was not accurate. We suggest that OptKnock modelling predictions be evaluated by using single-level FBA to ensure the accuracy of metabolic pathway design. Furthermore, the zwf gene knockout resulted in the change of metabolic fluxes to enhance the lactate productivity.
Collapse
Affiliation(s)
- Nunthaphan Vikromvarasiri
- grid.509461.f0000 0004 1757 8255RIKEN Center for Sustainable Resource Science, 1‑7‑22 Suehiro‑cho, Tsurumi‑ku, Yokohama, Kanagawa 230‑0045 Japan
| | - Shuhei Noda
- grid.509461.f0000 0004 1757 8255RIKEN Center for Sustainable Resource Science, 1‑7‑22 Suehiro‑cho, Tsurumi‑ku, Yokohama, Kanagawa 230‑0045 Japan
| | - Tomokazu Shirai
- grid.509461.f0000 0004 1757 8255RIKEN Center for Sustainable Resource Science, 1‑7‑22 Suehiro‑cho, Tsurumi‑ku, Yokohama, Kanagawa 230‑0045 Japan
| | - Akihiko Kondo
- grid.509461.f0000 0004 1757 8255RIKEN Center for Sustainable Resource Science, 1‑7‑22 Suehiro‑cho, Tsurumi‑ku, Yokohama, Kanagawa 230‑0045 Japan ,grid.31432.370000 0001 1092 3077Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657-8501 Japan
| |
Collapse
|
3
|
Sieow BFL, De Sotto R, Seet ZRD, Hwang IY, Chang MW. Synthetic Biology Meets Machine Learning. Methods Mol Biol 2023; 2553:21-39. [PMID: 36227537 DOI: 10.1007/978-1-0716-2617-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
This chapter outlines the myriad applications of machine learning (ML) in synthetic biology, specifically in engineering cell and protein activity, and metabolic pathways. Though by no means comprehensive, the chapter highlights several prominent computational tools applied in the field and their potential use cases. The examples detailed reinforce how ML algorithms can enhance synthetic biology research by providing data-driven insights into the behavior of living systems, even without detailed knowledge of their underlying mechanisms. By doing so, ML promises to increase the efficiency of research projects by modeling hypotheses in silico that can then be tested through experiments. While challenges related to training dataset generation and computational costs remain, ongoing improvements in ML tools are paving the way for smarter and more streamlined synthetic biology workflows that can be readily employed to address grand challenges across manufacturing, medicine, engineering, agriculture, and beyond.
Collapse
Affiliation(s)
- Brendan Fu-Long Sieow
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering Programme, National University of Singapore, Singapore, Singapore
| | - Ryan De Sotto
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhi Ren Darren Seet
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - In Young Hwang
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Matthew Wook Chang
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore, Singapore.
- Synthetic Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Role of Dissimilative Pathway of Komagataella phaffii (Pichia pastoris): Formaldehyde Toxicity and Energy Metabolism. Microorganisms 2022; 10:microorganisms10071466. [PMID: 35889185 PMCID: PMC9321669 DOI: 10.3390/microorganisms10071466] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/30/2023] Open
Abstract
Komagataella phaffii (aka Pichia pastoris) is a yeast able to grow in methanol as the sole carbon and energy source. This substrate is converted into formaldehyde, a toxic intermediary that can either be assimilated to biomass or dissimilated to CO2 through the enzymes formaldehyde dehydrogenase (FLD) and formate dehydrogenase, also producing energy in the form of NADH. The dissimilative pathway has been described as an energy producing and a detoxifying route, but conclusive evidence has not been provided for this. In order to elucidate this theory, we generated mutants lacking the FLD activity (Δfld1) and used flux analysis to evaluate the metabolic impact of this disrupted pathway. Unexpectedly, we found that the specific growth rate of the Δfld1 strain was only slightly lower (92%) than the control. In contrast, the sensitivity to formaldehyde pulses (up to 8mM) was significantly higher in the Δfld1 mutant strain and was associated with a higher maintenance energy. In addition, the intracellular flux estimation revealed a high metabolic flexibility of K. phaffii in response to the disrupted pathway. Our results suggest that the role of the dissimilative pathway is mainly to protect the cells from the harmful effect of formaldehyde, as they were able to compensate for the energy provided from this pathway when disrupted.
Collapse
|
5
|
Narad P, Naresh G, Sengupta A. Metabolomics and flux balance analysis. Bioinformatics 2022. [DOI: 10.1016/b978-0-323-89775-4.00008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
6
|
Metabolic Modeling with MetaFlux. Methods Mol Biol 2021. [PMID: 34718999 DOI: 10.1007/978-1-0716-1585-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
The MetaFlux software supports creating, executing, and solving quantitative metabolic flux models using flux balance analysis (FBA). MetaFlux offers four modes of operation: (1) solving mode executes an FBA model for an individual organism or for an organism community, (2) gene knockout mode executes an FBA model with one or many gene knockouts, (3) development mode assists the user in creating and improving FBA models, and (4) flux variability analysis mode generates a report of the robustness of an FBA model. MetaFlux also solves dynamic FBA (dFBA) for both individual organisms and communities of organisms. MetaFlux can be used in two different environments: on your local computer, which requires the installation of the Pathway Tools software, or through the web, which does not require installation of Pathway Tools. On your local computer, MetaFlux offers all four modes of operation, whereas the web environment provides only the solving mode.Several visualization tools are available to analyze model solutions. The Cellular Overview tool graphically shows the reaction fluxes on an organism's metabolic map once a model is solved. The Omics Dashboard provides a hierarchical approach to visualizing reaction fluxes, organized by metabolic subsystems. For a community of organisms, plotting of accumulated biomasses and metabolites can be performed using the Gnuplot tool.In this chapter, we present eight methods using MetaFlux. Five solving mode methods illustrate execution of models for individual organisms and for organism communities. One method illustrates the gene knockout mode. Two methods for the development mode illustrate steps for developing new metabolic models.
Collapse
|
7
|
Munro LJ, Kell DB. Intelligent host engineering for metabolic flux optimisation in biotechnology. Biochem J 2021; 478:3685-3721. [PMID: 34673920 PMCID: PMC8589332 DOI: 10.1042/bcj20210535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Optimising the function of a protein of length N amino acids by directed evolution involves navigating a 'search space' of possible sequences of some 20N. Optimising the expression levels of P proteins that materially affect host performance, each of which might also take 20 (logarithmically spaced) values, implies a similar search space of 20P. In this combinatorial sense, then, the problems of directed protein evolution and of host engineering are broadly equivalent. In practice, however, they have different means for avoiding the inevitable difficulties of implementation. The spare capacity exhibited in metabolic networks implies that host engineering may admit substantial increases in flux to targets of interest. Thus, we rehearse the relevant issues for those wishing to understand and exploit those modern genome-wide host engineering tools and thinking that have been designed and developed to optimise fluxes towards desirable products in biotechnological processes, with a focus on microbial systems. The aim throughput is 'making such biology predictable'. Strategies have been aimed at both transcription and translation, especially for regulatory processes that can affect multiple targets. However, because there is a limit on how much protein a cell can produce, increasing kcat in selected targets may be a better strategy than increasing protein expression levels for optimal host engineering.
Collapse
Affiliation(s)
- Lachlan J. Munro
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - Douglas B. Kell
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs. Lyngby, Denmark
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, U.K
- Mellizyme Biotechnology Ltd, IC1, Liverpool Science Park, 131 Mount Pleasant, Liverpool L3 5TF, U.K
| |
Collapse
|
8
|
Nakazawa S, Imaichi O, Kogure T, Kubota T, Toyoda K, Suda M, Inui M, Ito K, Shirai T, Araki M. History-Driven Genetic Modification Design Technique Using a Domain-Specific Lexical Model for the Acceleration of DBTL Cycles for Microbial Cell Factories. ACS Synth Biol 2021; 10:2308-2317. [PMID: 34351735 DOI: 10.1021/acssynbio.1c00234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of microbes for conducting bioprocessing via synthetic biology involves design-build-test-learn (DBTL) cycles. To aid the designing step, we developed a computational technique that suggests next genetic modifications on the basis of relatedness to the user's design history of genetic modifications accumulated through former DBTL cycles conducted by the user. This technique, which comprehensively retrieves well-known designs related to the history, involves searching text for previous literature and then mining genes that frequently co-occur in the literature with those modified genes. We further developed a domain-specific lexical model that weights literature that is more related to the domain of metabolic engineering to emphasize genes modified for bioprocessing. Our technique made a suggestion by using a history of creating a Corynebacterium glutamicum strain producing shikimic acid that had 18 genetic modifications. Inspired by the suggestion, eight genes were considered by biologists for further modification, and modifying four of these genes proved experimentally efficient in increasing the production of shikimic acid. These results indicated that our proposed technique successfully utilized the former cycles to suggest relevant designs that biologists considered worth testing. Comprehensive retrieval of well-tested designs will help less-experienced researchers overcome the entry barrier as well as inspire experienced researchers to formulate design concepts that have been overlooked or suspended. This technique will aid DBTL cycles by feeding histories back to the next genetic design, thereby complementing the designing step.
Collapse
Affiliation(s)
- Shiori Nakazawa
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., 1-280, Higashi-Koigakubo, Kokubunji-shi, Tokyo 185-8601, Japan
| | - Osamu Imaichi
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., 1-280, Higashi-Koigakubo, Kokubunji-shi, Tokyo 185-8601, Japan
| | - Takahisa Kogure
- Research Institute of Innovative Technology for Earth, 9-2, Kizugawadai, Kizugawa-shi, Kyoto 619-0292, Japan
| | - Takeshi Kubota
- Research Institute of Innovative Technology for Earth, 9-2, Kizugawadai, Kizugawa-shi, Kyoto 619-0292, Japan
| | - Koichi Toyoda
- Research Institute of Innovative Technology for Earth, 9-2, Kizugawadai, Kizugawa-shi, Kyoto 619-0292, Japan
| | - Masako Suda
- Research Institute of Innovative Technology for Earth, 9-2, Kizugawadai, Kizugawa-shi, Kyoto 619-0292, Japan
| | - Masayuki Inui
- Research Institute of Innovative Technology for Earth, 9-2, Kizugawadai, Kizugawa-shi, Kyoto 619-0292, Japan
| | - Kiyoto Ito
- Center for Exploratory Research, Research and Development Group, Hitachi, Ltd., 1-280, Higashi-Koigakubo, Kokubunji-shi, Tokyo 185-8601, Japan
| | - Tomokazu Shirai
- Riken, 1-6 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 240-0035, Japan
| | - Michihiro Araki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan
- Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
- National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8638, Japan
| |
Collapse
|
9
|
Gayathri KV, Aishwarya S, Kumar PS, Rajendran UR, Gunasekaran K. Metabolic and molecular modelling of zebrafish gut biome to unravel antimicrobial peptides through metagenomics. Microb Pathog 2021; 154:104862. [PMID: 33781870 DOI: 10.1016/j.micpath.2021.104862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/25/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023]
Abstract
Recently efforts have been taken for unravelling mysteries between host-microbe interactions in gut microbiome studies of model organisms through metagenomics. Co-existence and the co-evolution of the microorganisms is the significant cause of the growing antimicrobial menace. There needs a novel approach to develop potential antimicrobials with capabilities to act directly on the resistant microbes with reduced side effects. One such is to tap them from the natural resources, preferably the gut of the most closely related animal model. In this study, we employed metagenomics approaches to identify the large taxonomic genomes of the zebra fish gut. About 256 antimicrobial peptides were identified using gene ontology predictions from Macrel and Pubseed servers. Upon the property predictions, the top 10 antimicrobial peptides were screened based on their action against many resistant bacterial species, including Klebsiella pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus, E. coli, and Bacillus cereus. Metabolic modelling and flux balance analysis (FBA) were computed to conclude the antibiotic such as tetracycline, cephalosporins, puromycin, neomycin biosynthesis pathways were adopted by the microbiome as protection strategies. Molecular modelling strategies, including molecular docking and dynamics, were performed to estimate the antimicrobial peptides' binding against the target-putative nucleic acid binding lipoprotein and confirm stable binding. One specific antimicrobial peptide with the sequence "MPPYLHEIQPHTASNCQTELVIKL" showed promising results with 53% hydrophobic residues and a net charge +2.5, significant for the development of antimicrobial peptides. The said peptide also showed promising interactions with the target protein and expressed stable binding with docking energy of -429.34 kcal/mol and the average root mean square deviation of 1 A0. The study is a novel approach focusing on tapping out potential antimicrobial peptides to be developed against most resistant bacterial species.
Collapse
Affiliation(s)
- K Veena Gayathri
- Department of Bioinformatics, Stella Maris College (Autonomous), Chennai, 600086, India.
| | - S Aishwarya
- Department of Biotechnology, Stella Maris College (Autonomous), Chennai, 600086, India; CAS in Crystallography and Biophysics, University of Madras, Chennai, 600025, India
| | - P Senthil Kumar
- Department of Chemical Engineering, Sri Sivasubramaniya Nadar College of Engineering, Kalavakkam, Chennai, 603 110, India.
| | - U Rohini Rajendran
- Department of Bioinformatics, Stella Maris College (Autonomous), Chennai, 600086, India
| | - K Gunasekaran
- CAS in Crystallography and Biophysics, University of Madras, Chennai, 600025, India
| |
Collapse
|
10
|
Dusad V, Thiel D, Barahona M, Keun HC, Oyarzún DA. Opportunities at the Interface of Network Science and Metabolic Modeling. Front Bioeng Biotechnol 2021; 8:591049. [PMID: 33569373 PMCID: PMC7868444 DOI: 10.3389/fbioe.2020.591049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022] Open
Abstract
Metabolism plays a central role in cell physiology because it provides the molecular machinery for growth. At the genome-scale, metabolism is made up of thousands of reactions interacting with one another. Untangling this complexity is key to understand how cells respond to genetic, environmental, or therapeutic perturbations. Here we discuss the roles of two complementary strategies for the analysis of genome-scale metabolic models: Flux Balance Analysis (FBA) and network science. While FBA estimates metabolic flux on the basis of an optimization principle, network approaches reveal emergent properties of the global metabolic connectivity. We highlight how the integration of both approaches promises to deliver insights on the structure and function of metabolic systems with wide-ranging implications in discovery science, precision medicine and industrial biotechnology.
Collapse
Affiliation(s)
- Varshit Dusad
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Denise Thiel
- Department of Mathematics, Imperial College London, London, United Kingdom
| | - Mauricio Barahona
- Department of Mathematics, Imperial College London, London, United Kingdom
| | - Hector C. Keun
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Diego A. Oyarzún
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
11
|
Anand S, Mukherjee K, Padmanabhan P. An insight to flux-balance analysis for biochemical networks. Biotechnol Genet Eng Rev 2020; 36:32-55. [PMID: 33292061 DOI: 10.1080/02648725.2020.1847440] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Systems biology is one of the integrated ways to study biological systems and is more favourable than the earlier used approaches. It includes metabolic pathway analysis, modelling, and regulatory as well as signal transduction for getting insights into cellular behaviour. Among the various techniques of modelling, simulation, analysis of networks and pathways, flux-based analysis (FBA) has been recognised because of its extensibility as well as simplicity. It is widely accepted because it is not like a mechanistic simulation which depends on accurate kinetic data. The study of fluxes through the network is informative and can give insights even in the absence of kinetic data. FBA is one of the widely used tools to study biochemical networks and needs information of reaction stoichiometry, growth requirements, specific measurement parameters of the biological system, in particular the reconstruction of the metabolic network for the genome-scale, many of which have already been built previously. It defines the boundaries of flux distributions which are possible and achievable with a defined set of genes. This review article gives an insight into FBA, from the extension of flux balancing to mathematical representation followed by a discussion about the formulation of flux-balance analysis problems, defining constraints for the stoichiometry of the pathways and the tools that can be used in FBA such as FASIMA, COBRA toolbox, and OptFlux. It also includes broader areas in terms of applications which can be covered by FBA as well as the queries which can be addressed through FBA.
Collapse
Affiliation(s)
- Shreya Anand
- Department of Bio-Engineering, Birla Institute of Technology , Ranchi, JH, India
| | - Koel Mukherjee
- Department of Bio-Engineering, Birla Institute of Technology , Ranchi, JH, India
| | - Padmini Padmanabhan
- Department of Bio-Engineering, Birla Institute of Technology , Ranchi, JH, India
| |
Collapse
|
12
|
Hari A, Lobo D. Fluxer: a web application to compute, analyze and visualize genome-scale metabolic flux networks. Nucleic Acids Res 2020; 48:W427-W435. [PMID: 32442279 PMCID: PMC7319574 DOI: 10.1093/nar/gkaa409] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/20/2020] [Accepted: 05/06/2020] [Indexed: 12/19/2022] Open
Abstract
Next-generation sequencing has paved the way for the reconstruction of genome-scale metabolic networks as a powerful tool for understanding metabolic circuits in any organism. However, the visualization and extraction of knowledge from these large networks comprising thousands of reactions and metabolites is a current challenge in need of user-friendly tools. Here we present Fluxer (https://fluxer.umbc.edu), a free and open-access novel web application for the computation and visualization of genome-scale metabolic flux networks. Any genome-scale model based on the Systems Biology Markup Language can be uploaded to the tool, which automatically performs Flux Balance Analysis and computes different flux graphs for visualization and analysis. The major metabolic pathways for biomass growth or for biosynthesis of any metabolite can be interactively knocked-out, analyzed and visualized as a spanning tree, dendrogram or complete graph using different layouts. In addition, Fluxer can compute and visualize the k-shortest metabolic paths between any two metabolites or reactions to identify the main metabolic routes between two compounds of interest. The web application includes >80 whole-genome metabolic reconstructions of diverse organisms from bacteria to human, readily available for exploration. Fluxer enables the efficient analysis and visualization of genome-scale metabolic models toward the discovery of key metabolic pathways.
Collapse
Affiliation(s)
- Archana Hari
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland 21250, USA
| | - Daniel Lobo
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland 21250, USA
| |
Collapse
|
13
|
Eicher T, Kinnebrew G, Patt A, Spencer K, Ying K, Ma Q, Machiraju R, Mathé EA. Metabolomics and Multi-Omics Integration: A Survey of Computational Methods and Resources. Metabolites 2020; 10:E202. [PMID: 32429287 PMCID: PMC7281435 DOI: 10.3390/metabo10050202] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/07/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
As researchers are increasingly able to collect data on a large scale from multiple clinical and omics modalities, multi-omics integration is becoming a critical component of metabolomics research. This introduces a need for increased understanding by the metabolomics researcher of computational and statistical analysis methods relevant to multi-omics studies. In this review, we discuss common types of analyses performed in multi-omics studies and the computational and statistical methods that can be used for each type of analysis. We pinpoint the caveats and considerations for analysis methods, including required parameters, sample size and data distribution requirements, sources of a priori knowledge, and techniques for the evaluation of model accuracy. Finally, for the types of analyses discussed, we provide examples of the applications of corresponding methods to clinical and basic research. We intend that our review may be used as a guide for metabolomics researchers to choose effective techniques for multi-omics analyses relevant to their field of study.
Collapse
Affiliation(s)
- Tara Eicher
- Biomedical Informatics Department, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.E.); (G.K.); (K.S.); (Q.M.); (R.M.)
- Computer Science and Engineering Department, The Ohio State University College of Engineering, Columbus, OH 43210, USA
| | - Garrett Kinnebrew
- Biomedical Informatics Department, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.E.); (G.K.); (K.S.); (Q.M.); (R.M.)
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA;
- Bioinformatics Shared Resource Group, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew Patt
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20892, USA;
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Kyle Spencer
- Biomedical Informatics Department, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.E.); (G.K.); (K.S.); (Q.M.); (R.M.)
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
- Nationwide Children’s Research Hospital, Columbus, OH 43210, USA
| | - Kevin Ying
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA;
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| | - Qin Ma
- Biomedical Informatics Department, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.E.); (G.K.); (K.S.); (Q.M.); (R.M.)
| | - Raghu Machiraju
- Biomedical Informatics Department, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.E.); (G.K.); (K.S.); (Q.M.); (R.M.)
- Computer Science and Engineering Department, The Ohio State University College of Engineering, Columbus, OH 43210, USA
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- Translational Data Analytics Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Ewy A. Mathé
- Biomedical Informatics Department, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.E.); (G.K.); (K.S.); (Q.M.); (R.M.)
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, 9800 Medical Center Dr., Rockville, MD, 20892, USA;
| |
Collapse
|
14
|
Blanco-Míguez A, Fdez-Riverola F, Sánchez B, Lourenço A. Resources and tools for the high-throughput, multi-omic study of intestinal microbiota. Brief Bioinform 2020; 20:1032-1056. [PMID: 29186315 DOI: 10.1093/bib/bbx156] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/23/2017] [Indexed: 12/18/2022] Open
Abstract
The human gut microbiome impacts several aspects of human health and disease, including digestion, drug metabolism and the propensity to develop various inflammatory, autoimmune and metabolic diseases. Many of the molecular processes that play a role in the activity and dynamics of the microbiota go beyond species and genic composition and thus, their understanding requires advanced bioinformatics support. This article aims to provide an up-to-date view of the resources and software tools that are being developed and used in human gut microbiome research, in particular data integration and systems-level analysis efforts. These efforts demonstrate the power of standardized and reproducible computational workflows for integrating and analysing varied omics data and gaining deeper insights into microbe community structure and function as well as host-microbe interactions.
Collapse
Affiliation(s)
| | | | | | - Anália Lourenço
- Dpto. de Informática - Universidade de Vigo, ESEI - Escuela Superior de Ingeniería Informática, Edificio politécnico, Campus Universitario As Lagoas s/n, 32004 Ourense, Spain
| |
Collapse
|
15
|
Koduru L, Kim HY, Lakshmanan M, Mohanty B, Lee YQ, Lee CH, Lee D. Genome-scale metabolic reconstruction and in silico analysis of the rice leaf blight pathogen, Xanthomonas oryzae. MOLECULAR PLANT PATHOLOGY 2020; 21:527-540. [PMID: 32068953 PMCID: PMC7060145 DOI: 10.1111/mpp.12914] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/09/2019] [Accepted: 01/13/2020] [Indexed: 05/29/2023]
Abstract
Xanthomonas oryzae pv. oryzae (Xoo) is a vascular pathogen that causes leaf blight in rice, leading to severe yield losses. Since the usage of chemical control methods has not been very promising for the future disease management, it is of high importance to systematically gain new insights about Xoo virulence and pathogenesis, and devise effective strategies to combat the rice disease. To do this, we reconstructed a genome-scale metabolic model of Xoo (iXOO673) and validated the model predictions using culture experiments. Comparison of the metabolic architecture of Xoo and other plant pathogens indicated that the Entner-Doudoroff pathway is a more common feature in these bacteria than previously thought, while suggesting some of the unique virulence mechanisms related to Xoo metabolism. Subsequent constraint-based flux analysis allowed us to show that Xoo modulates fluxes through gluconeogenesis, glycogen biosynthesis, and degradation pathways, thereby exacerbating the leaf blight in rice exposed to nitrogenous fertilizers, which is remarkably consistent with published experimental literature. Moreover, model-based interrogation of transcriptomic data revealed the metabolic components under the diffusible signal factor regulon that are crucial for virulence and survival in Xoo. Finally, we identified promising antibacterial targets for the control of leaf blight in rice by using gene essentiality analysis.
Collapse
Affiliation(s)
- Lokanand Koduru
- Bioprocessing Technology InstituteAgency for Science, Technology and ResearchSingapore
| | - Hyang Yeon Kim
- Department of Bioscience and BiotechnologyKonkuk UniversitySeoulRepublic of Korea
| | - Meiyappan Lakshmanan
- Bioprocessing Technology InstituteAgency for Science, Technology and ResearchSingapore
| | - Bijayalaxmi Mohanty
- Bioprocessing Technology InstituteAgency for Science, Technology and ResearchSingapore
| | - Yi Qing Lee
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and BiotechnologyKonkuk UniversitySeoulRepublic of Korea
| | - Dong‐Yup Lee
- Bioprocessing Technology InstituteAgency for Science, Technology and ResearchSingapore
- School of Chemical EngineeringSungkyunkwan UniversitySuwonRepublic of Korea
| |
Collapse
|
16
|
Mammalian Systems Biotechnology Reveals Global Cellular Adaptations in a Recombinant CHO Cell Line. Cell Syst 2019; 4:530-542.e6. [PMID: 28544881 DOI: 10.1016/j.cels.2017.04.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/14/2017] [Accepted: 04/26/2017] [Indexed: 01/16/2023]
Abstract
Effective development of host cells for therapeutic protein production is hampered by the poor characterization of cellular transfection. Here, we employed a multi-omics-based systems biotechnology approach to elucidate the genotypic and phenotypic differences between a wild-type and recombinant antibody-producing Chinese hamster ovary (CHO) cell line. At the genomic level, we observed extensive rearrangements in specific targeted loci linked to transgene integration sites. Transcriptional re-wiring of DNA damage repair and cellular metabolism in the antibody producer, via changes in gene copy numbers, was also detected. Subsequent integration of transcriptomic data with a genome-scale metabolic model showed a substantial increase in energy metabolism in the antibody producer. Metabolomics, lipidomics, and glycomics analyses revealed an elevation in long-chain lipid species, potentially associated with protein transport and secretion requirements, and a surprising stability of N-glycosylation profiles between both cell lines. Overall, the proposed knowledge-based systems biotechnology framework can further accelerate mammalian cell-line engineering in a targeted manner.
Collapse
|
17
|
Human Systems Biology and Metabolic Modelling: A Review-From Disease Metabolism to Precision Medicine. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8304260. [PMID: 31281846 PMCID: PMC6590590 DOI: 10.1155/2019/8304260] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/07/2019] [Accepted: 05/20/2019] [Indexed: 01/06/2023]
Abstract
In cell and molecular biology, metabolism is the only system that can be fully simulated at genome scale. Metabolic systems biology offers powerful abstraction tools to simulate all known metabolic reactions in a cell, therefore providing a snapshot that is close to its observable phenotype. In this review, we cover the 15 years of human metabolic modelling. We show that, although the past five years have not experienced large improvements in the size of the gene and metabolite sets in human metabolic models, their accuracy is rapidly increasing. We also describe how condition-, tissue-, and patient-specific metabolic models shed light on cell-specific changes occurring in the metabolic network, therefore predicting biomarkers of disease metabolism. We finally discuss current challenges and future promising directions for this research field, including machine/deep learning and precision medicine. In the omics era, profiling patients and biological processes from a multiomic point of view is becoming more common and less expensive. Starting from multiomic data collected from patients and N-of-1 trials where individual patients constitute different case studies, methods for model-building and data integration are being used to generate patient-specific models. Coupled with state-of-the-art machine learning methods, this will allow characterizing each patient's disease phenotype and delivering precision medicine solutions, therefore leading to preventative medicine, reduced treatment, and in silico clinical trials.
Collapse
|
18
|
Heirendt L, Arreckx S, Pfau T, Mendoza SN, Richelle A, Heinken A, Haraldsdóttir HS, Wachowiak J, Keating SM, Vlasov V, Magnusdóttir S, Ng CY, Preciat G, Žagare A, Chan SHJ, Aurich MK, Clancy CM, Modamio J, Sauls JT, Noronha A, Bordbar A, Cousins B, El Assal DC, Valcarcel LV, Apaolaza I, Ghaderi S, Ahookhosh M, Ben Guebila M, Kostromins A, Sompairac N, Le HM, Ma D, Sun Y, Wang L, Yurkovich JT, Oliveira MAP, Vuong PT, El Assal LP, Kuperstein I, Zinovyev A, Hinton HS, Bryant WA, Aragón Artacho FJ, Planes FJ, Stalidzans E, Maass A, Vempala S, Hucka M, Saunders MA, Maranas CD, Lewis NE, Sauter T, Palsson BØ, Thiele I, Fleming RMT. Creation and analysis of biochemical constraint-based models using the COBRA Toolbox v.3.0. Nat Protoc 2019; 14:639-702. [PMID: 30787451 PMCID: PMC6635304 DOI: 10.1038/s41596-018-0098-2] [Citation(s) in RCA: 622] [Impact Index Per Article: 124.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Constraint-based reconstruction and analysis (COBRA) provides a molecular mechanistic framework for integrative analysis of experimental molecular systems biology data and quantitative prediction of physicochemically and biochemically feasible phenotypic states. The COBRA Toolbox is a comprehensive desktop software suite of interoperable COBRA methods. It has found widespread application in biology, biomedicine, and biotechnology because its functions can be flexibly combined to implement tailored COBRA protocols for any biochemical network. This protocol is an update to the COBRA Toolbox v.1.0 and v.2.0. Version 3.0 includes new methods for quality-controlled reconstruction, modeling, topological analysis, strain and experimental design, and network visualization, as well as network integration of chemoinformatic, metabolomic, transcriptomic, proteomic, and thermochemical data. New multi-lingual code integration also enables an expansion in COBRA application scope via high-precision, high-performance, and nonlinear numerical optimization solvers for multi-scale, multi-cellular, and reaction kinetic modeling, respectively. This protocol provides an overview of all these new features and can be adapted to generate and analyze constraint-based models in a wide variety of scenarios. The COBRA Toolbox v.3.0 provides an unparalleled depth of COBRA methods.
Collapse
Affiliation(s)
- Laurent Heirendt
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Sylvain Arreckx
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Thomas Pfau
- Life Sciences Research Unit, University of Luxembourg, Belvaux, Luxembourg
| | - Sebastián N Mendoza
- Center for Genome Regulation (Fondap 15090007), University of Chile, Santiago, Chile
- Mathomics, Center for Mathematical Modeling, University of Chile, Santiago, Chile
| | - Anne Richelle
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Almut Heinken
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Hulda S Haraldsdóttir
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Jacek Wachowiak
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Sarah M Keating
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, UK
| | - Vanja Vlasov
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Stefania Magnusdóttir
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Chiam Yu Ng
- Department of Chemical Engineering, The Pennsylvania State University, State College, PA, USA
| | - German Preciat
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Alise Žagare
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Siu H J Chan
- Department of Chemical Engineering, The Pennsylvania State University, State College, PA, USA
| | - Maike K Aurich
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Catherine M Clancy
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Jennifer Modamio
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - John T Sauls
- Department of Physics, and Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA
| | - Alberto Noronha
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | | | - Benjamin Cousins
- Algorithms and Randomness Center, School of Computer Science, Georgia Institute of Technology, Atlanta, GA, USA
| | - Diana C El Assal
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Luis V Valcarcel
- Biomedical Engineering and Sciences Department, TECNUN, University of Navarra, San Sebastián, Spain
| | - Iñigo Apaolaza
- Biomedical Engineering and Sciences Department, TECNUN, University of Navarra, San Sebastián, Spain
| | - Susan Ghaderi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Masoud Ahookhosh
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Marouen Ben Guebila
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Andrejs Kostromins
- Institute of Microbiology and Biotechnology, University of Latvia, Riga, Latvia
| | - Nicolas Sompairac
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - Hoai M Le
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Ding Ma
- Department of Management Science and Engineering, Stanford University, Stanford, CA, USA
| | - Yuekai Sun
- Department of Statistics, University of Michigan, Ann Arbor, MI, USA
| | - Lin Wang
- Department of Chemical Engineering, The Pennsylvania State University, State College, PA, USA
| | - James T Yurkovich
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Miguel A P Oliveira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Phan T Vuong
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Lemmer P El Assal
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Inna Kuperstein
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - H Scott Hinton
- Utah State University Research Foundation, North Logan, UT, USA
| | - William A Bryant
- Centre for Integrative Systems Biology and Bioinformatics, Department of Life Sciences, Imperial College London, London, UK
| | | | - Francisco J Planes
- Biomedical Engineering and Sciences Department, TECNUN, University of Navarra, San Sebastián, Spain
| | - Egils Stalidzans
- Institute of Microbiology and Biotechnology, University of Latvia, Riga, Latvia
| | - Alejandro Maass
- Center for Genome Regulation (Fondap 15090007), University of Chile, Santiago, Chile
- Mathomics, Center for Mathematical Modeling, University of Chile, Santiago, Chile
| | - Santosh Vempala
- Algorithms and Randomness Center, School of Computer Science, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael Hucka
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA
| | - Michael A Saunders
- Department of Management Science and Engineering, Stanford University, Stanford, CA, USA
| | - Costas D Maranas
- Department of Chemical Engineering, The Pennsylvania State University, State College, PA, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, USA
- Novo Nordisk Foundation Center for Biosustainability, University of California, San Diego, La Jolla, CA, USA
| | - Thomas Sauter
- Life Sciences Research Unit, University of Luxembourg, Belvaux, Luxembourg
| | - Bernhard Ø Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Lyngby, Denmark
| | - Ines Thiele
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Ronan M T Fleming
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg.
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
19
|
Ma Z, Ye C, Deng W, Xu M, Wang Q, Liu G, Wang F, Liu L, Xu Z, Shi G, Ding Z. Reconstruction and Analysis of a Genome-Scale Metabolic Model of Ganoderma lucidum for Improved Extracellular Polysaccharide Production. Front Microbiol 2018; 9:3076. [PMID: 30619160 PMCID: PMC6298397 DOI: 10.3389/fmicb.2018.03076] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/29/2018] [Indexed: 12/19/2022] Open
Abstract
In this study, we reconstructed for the first time a genome-scale metabolic model (GSMM) of Ganoderma lucidum strain CGMCC5.26, termed model iZBM1060, containing 1060 genes, 1202 metabolites, and 1404 reactions. Important findings based on model iZBM1060 and its predictions are as follows: (i) The extracellular polysaccharide (EPS) biosynthetic pathway was elucidated completely. (ii) A new fermentation strategy is proposed: addition of phenylalanine increased EPS production by 32.80% in simulations and by 38.00% in experiments. (iii) Eight genes for key enzymes were proposed for EPS overproduction. Model iZBM1060 provides a useful platform for regulating EPS production in terms of system metabolic engineering for G. lucidum, as well as a guide for future metabolic pathway construction of other high value-added edible/ medicinal mushroom species.
Collapse
Affiliation(s)
- Zhongbao Ma
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Chao Ye
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Weiwei Deng
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Mengmeng Xu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Qiong Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Gaoqiang Liu
- Key Laboratory of Cultivation and Protection for Non-Wood Forest Trees, Ministry of Education, College of Life Science and Technology, Central South University of Forestry and Technology, Changsha, China
| | - Feng Wang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Liming Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Zhenghong Xu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Guiyang Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| | - Zhongyang Ding
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
20
|
Koduru L, Lakshmanan M, Lee DY. In silico model-guided identification of transcriptional regulator targets for efficient strain design. Microb Cell Fact 2018; 17:167. [PMID: 30359263 PMCID: PMC6201637 DOI: 10.1186/s12934-018-1015-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/20/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Cellular metabolism is tightly regulated by hard-wired multiple layers of biological processes to achieve robust and homeostatic states given the limited resources. As a result, even the most intuitive enzyme-centric metabolic engineering endeavours through the up-/down-regulation of multiple genes in biochemical pathways often deliver insignificant improvements in the product yield. In this regard, targeted engineering of transcriptional regulators (TRs) that control several metabolic functions in modular patterns is an interesting strategy. However, only a handful of in silico model-added techniques are available for identifying the TR manipulation candidates, thus limiting its strain design application. RESULTS We developed hierarchical-Beneficial Regulatory Targeting (h-BeReTa) which employs a genome-scale metabolic model and transcriptional regulatory network (TRN) to identify the relevant TR targets suitable for strain improvement. We then applied this method to industrially relevant metabolites and cell factory hosts, Escherichia coli and Corynebacterium glutamicum. h-BeReTa suggested several promising TR targets, many of which have been validated through literature evidences. h-BeReTa considers the hierarchy of TRs in the TRN and also accounts for alternative metabolic pathways which may divert flux away from the product while identifying suitable metabolic fluxes, thereby performing superior in terms of global TR target identification. CONCLUSIONS In silico model-guided strain design framework, h-BeReTa, was presented for identifying transcriptional regulator targets. Its efficacy and applicability to microbial cell factories were successfully demonstrated via case studies involving two cell factory hosts, as such suggesting several intuitive targets for overproducing various value-added compounds.
Collapse
Affiliation(s)
- Lokanand Koduru
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117576, Singapore
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, Singapore, 138668, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, Singapore, 138668, Singapore.
- School of Chemical Engineering, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
21
|
Ang KS, Lakshmanan M, Lee NR, Lee DY. Metabolic Modeling of Microbial Community Interactions for Health, Environmental and Biotechnological Applications. Curr Genomics 2018; 19:712-722. [PMID: 30532650 PMCID: PMC6225453 DOI: 10.2174/1389202919666180911144055] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 11/08/2017] [Accepted: 11/11/2017] [Indexed: 02/08/2023] Open
Abstract
In nature, microbes do not exist in isolation but co-exist in a variety of ecological and biological environments and on various host organisms. Due to their close proximity, these microbes interact among themselves, and also with the hosts in both positive and negative manners. Moreover, these interactions may modulate dynamically upon external stimulus as well as internal community changes. This demands systematic techniques such as mathematical modeling to understand the intrinsic community behavior. Here, we reviewed various approaches for metabolic modeling of microbial communities. If detailed species-specific information is available, segregated models of individual organisms can be constructed and connected via metabolite exchanges; otherwise, the community may be represented as a lumped network of metabolic reactions. The constructed models can then be simulated to help fill knowledge gaps, and generate testable hypotheses for designing new experiments. More importantly, such community models have been developed to study microbial interactions in various niches such as host microbiome, biogeochemical and bioremediation, waste water treatment and synthetic consortia. As such, the metabolic modeling efforts have allowed us to gain new insights into the natural and synthetic microbial communities, and design interventions to achieve specific goals. Finally, potential directions for future development in metabolic modeling of microbial communities were also discussed.
Collapse
Affiliation(s)
- Kok Siong Ang
- 1Bioprocessing Technology Institute (BTI), ASTAR, Singapore 138668, Singapore; 2Department of Chemical and Biomolecular Engineering, and NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117585, Singapore; 3School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do16419, Republic of Korea
| | - Meiyappan Lakshmanan
- 1Bioprocessing Technology Institute (BTI), ASTAR, Singapore 138668, Singapore; 2Department of Chemical and Biomolecular Engineering, and NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117585, Singapore; 3School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do16419, Republic of Korea
| | - Na-Rae Lee
- 1Bioprocessing Technology Institute (BTI), ASTAR, Singapore 138668, Singapore; 2Department of Chemical and Biomolecular Engineering, and NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117585, Singapore; 3School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do16419, Republic of Korea
| | - Dong-Yup Lee
- 1Bioprocessing Technology Institute (BTI), ASTAR, Singapore 138668, Singapore; 2Department of Chemical and Biomolecular Engineering, and NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117585, Singapore; 3School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do16419, Republic of Korea
| |
Collapse
|
22
|
Botero D, Valdés I, Rodríguez MJ, Henao D, Danies G, González AF, Restrepo S. A Genome-Scale Metabolic Reconstruction of Phytophthora infestans With the Integration of Transcriptional Data Reveals the Key Metabolic Patterns Involved in the Interaction of Its Host. Front Genet 2018; 9:244. [PMID: 30042788 PMCID: PMC6048221 DOI: 10.3389/fgene.2018.00244] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 06/21/2018] [Indexed: 11/30/2022] Open
Abstract
Phytophthora infestans, the causal agent of late blight disease, affects potatoes and tomatoes worldwide. This plant pathogen has a hemibiotrophic lifestyle, having an initial biotrophic infection phase during which the pathogen spreads within the host tissue, followed by a necrotrophic phase in which host cell death is induced. Although increasing information is available on the molecular mechanisms, underlying the distinct phases of the hemibiotrophic lifestyle, studies that consider the entire metabolic processes in the pathogen while undergoing the biotrophic, transition to necrotrophic, and necrotrophic phases have not been conducted. In this study, the genome-scale metabolic reconstruction of P. infestans was achieved. Subsequently, transcriptional data (microarrays, RNA-seq) was integrated into the metabolic reconstruction to obtain context-specific (metabolic) models (CSMs) of the infection process, using constraint-based reconstruction and analysis. The goal was to identify specific metabolic markers for distinct stages of the pathogen's life cycle. Results indicate that the overall metabolism show significant changes during infection. The most significant changes in metabolism were observed at the latest time points of infection. Metabolic activity associated with purine, pyrimidine, fatty acid, fructose and mannose, arginine, glycine, serine, and threonine amino acids appeared to be the most important metabolisms of the pathogen during the course of the infection, showing high number of reactions associated with them and expression switches at important stages of the life cycle. This study provides a framework for future throughput studies of the metabolic changes during the hemibiotrophic life cycle of this important plant pathogen.
Collapse
Affiliation(s)
- David Botero
- Laboratory of Mycology and Phytopathology (LAMFU), Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Iván Valdés
- Laboratory of Mycology and Phytopathology (LAMFU), Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - María-Juliana Rodríguez
- Laboratory of Mycology and Phytopathology (LAMFU), Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Diana Henao
- Laboratory of Mycology and Phytopathology (LAMFU), Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| | - Giovanna Danies
- Department of Design, Universidad de los Andes, Bogotá, Colombia
| | - Andrés F González
- Group of Product and Process Design, Department of Chemical Engineering, Universidad de Los Andes, Bogotá, Colombia
| | - Silvia Restrepo
- Laboratory of Mycology and Phytopathology (LAMFU), Department of Biological Sciences, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
23
|
Kelley JJ, Maor S, Kim MK, Lane A, Lun DS. MOST-visualization: software for producing automated textbook-style maps of genome-scale metabolic networks. Bioinformatics 2018; 33:2596-2597. [PMID: 28430868 DOI: 10.1093/bioinformatics/btx240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 04/14/2017] [Indexed: 11/13/2022] Open
Abstract
Summary Visualization of metabolites, reactions and pathways in genome-scale metabolic networks (GEMs) can assist in understanding cellular metabolism. Three attributes are desirable in software used for visualizing GEMs: (i) automation, since GEMs can be quite large; (ii) production of understandable maps that provide ease in identification of pathways, reactions and metabolites; and (iii) visualization of the entire network to show how pathways are interconnected. No software currently exists for visualizing GEMs that satisfies all three characteristics, but MOST-Visualization, an extension of the software package MOST (Metabolic Optimization and Simulation Tool), satisfies (i), and by using a pre-drawn overview map of metabolism based on the Roche map satisfies (ii) and comes close to satisfying (iii). Availability and Implementation MOST is distributed for free on the GNU General Public License. The software and full documentation are available at http://most.ccib.rutgers.edu/. Contact dslun@rutgers.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- James J Kelley
- Department of Computer Science, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA
| | - Shay Maor
- Department of Computer Science, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA
| | - Min Kyung Kim
- Department of Computer Science, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA
| | - Anatoliy Lane
- Department of Computer Science, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA
| | - Desmond S Lun
- Department of Computer Science, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ 08102, USA.,Department of Plant Biology and Pathology, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
24
|
Özcan E, Çakır T. Genome-Scale Brain Metabolic Networks as Scaffolds for the Systems Biology of Neurodegenerative Diseases: Mapping Metabolic Alterations. ADVANCES IN NEUROBIOLOGY 2018; 21:195-217. [PMID: 30334223 DOI: 10.1007/978-3-319-94593-4_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Systems-based investigation of diseases requires integrated analysis of cellular networks and high-throughput data of gene products. The use of genome-scale metabolic networks for such integration has led to the elucidation of cellular mechanisms for several cell types from microorganisms to plants. It has become easier and cheaper to generate high-throughput data over years in the form of transcriptome, proteome and metabolome. This has tremendously improved the quality and quantity of information extracted from such data enabling the documentation of active pathways and reactions in cell metabolism. A number of omics-based datasets for several neurodegenerative diseases are now available in public repositories. This increases the potential of using genome-scale brain metabolic networks as a scaffold for this type of data to map metabolic alterations for the purpose of elucidating disease mechanisms and for the diagnosis and treatment of such disorders. This chapter first reviews omics data collected for neurodegenerative diseases to map their effect on metabolism. Later, the potential for genome-scale metabolic modeling of such data is reviewed and discussed in light of recently reconstructed brain metabolic networks at genome-scale.
Collapse
Affiliation(s)
- Emrah Özcan
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey.
| |
Collapse
|
25
|
Abstract
The science and art of Genome scale metabolic network reconstructions have been explicitly documented in the literature for organisms across all the three kingdoms of life. Constraints-based models derived from such reconstructions have been used to assess metabolic phenotypes of their complex connections to genotype accurately. The problem of infectious disease is complex due to the multifactorial response of the host to the pathogen. Systems biology approaches and modeling allow one to study, understand, and predict emergent properties of such complex responses. The integration of the host and pathogen metabolic networks and the subsequent merger of their stoichiometric matrices is nontrivial and requires understanding of both pathogen and host metabolism and physiologies. The protocol here describes the detailed process of network and stoichiometric matrix merger using a salmonella-mouse macrophage model. The protocol also discusses the interfacial and objective functions required to actually embark on the analysis of host-pathogen interaction models.
Collapse
|
26
|
Koduru L, Kim Y, Bang J, Lakshmanan M, Han NS, Lee DY. Genome-scale modeling and transcriptome analysis of Leuconostoc mesenteroides unravel the redox governed metabolic states in obligate heterofermentative lactic acid bacteria. Sci Rep 2017; 7:15721. [PMID: 29147021 PMCID: PMC5691038 DOI: 10.1038/s41598-017-16026-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/06/2017] [Indexed: 11/09/2022] Open
Abstract
Obligate heterofermentative lactic acid bacteria (LAB) are well-known for their beneficial health effects in humans. To delineate the incompletely characterized metabolism that currently limits their exploitation, at systems-level, we developed a genome-scale metabolic model of the representative obligate heterofermenting LAB, Leuconostoc mesenteroides (iLME620). Constraint-based flux analysis was then used to simulate several qualitative and quantitative phenotypes of L. mesenteroides, thereby evaluating the model validity. With established predictive capabilities, we subsequently employed iLME620 to elucidate unique metabolic characteristics of L. mesenteroides, such as the limited ability to utilize amino acids as energy source, and to substantiate the role of malolactic fermentation (MLF) in the reduction of pH-homeostatic burden on F0F1-ATPase. We also reported new hypothesis on the MLF mechanism that could be explained via a substrate channelling-like phenomenon mainly influenced by intracellular redox state rather than the intermediary reactions. Model simulations further revealed possible proton-symporter dependent activity of the energy efficient glucose-phosphotransferase system in obligate heterofermentative LAB. Moreover, integrated transcriptomic analysis allowed us to hypothesize transcriptional regulatory bias affecting the intracellular redox state. The insights gained here about the low ATP-yielding metabolism of L. mesenteroides, dominantly controlled by the cellular redox state, could potentially aid strain design for probiotic and cell factory applications.
Collapse
Affiliation(s)
- Lokanand Koduru
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117576, Singapore
| | - Yujin Kim
- Brain Korea 21 Center for Bio-Resource Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Jeongsu Bang
- Brain Korea 21 Center for Bio-Resource Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, Singapore, 138668, Singapore
| | - Nam Soo Han
- Brain Korea 21 Center for Bio-Resource Development, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| | - Dong-Yup Lee
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117576, Singapore.
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, Singapore, 138668, Singapore.
| |
Collapse
|
27
|
Kyriakopoulos S, Ang KS, Lakshmanan M, Huang Z, Yoon S, Gunawan R, Lee DY. Kinetic Modeling of Mammalian Cell Culture Bioprocessing: The Quest to Advance Biomanufacturing. Biotechnol J 2017; 13:e1700229. [DOI: 10.1002/biot.201700229] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/27/2017] [Accepted: 10/11/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Sarantos Kyriakopoulos
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
| | - Kok Siong Ang
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
| | - Zhuangrong Huang
- Department of Chemical Engineering; University of Massachusetts Lowell; Lowell MA USA
| | - Seongkyu Yoon
- Department of Chemical Engineering; University of Massachusetts Lowell; Lowell MA USA
| | - Rudiyanto Gunawan
- Institute for Chemical and Bioengineering; ETH Zurich; Zurich Switzerland
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science; Technology and Research (A*STAR); Singapore
- Department of Chemical and Biomolecular Engineering; National University of Singapore; Singapore
| |
Collapse
|
28
|
Rejc Ž, Magdevska L, Tršelič T, Osolin T, Vodopivec R, Mraz J, Pavliha E, Zimic N, Cvitanović T, Rozman D, Moškon M, Mraz M. Computational modelling of genome-scale metabolic networks and its application to CHO cell cultures. Comput Biol Med 2017; 88:150-160. [PMID: 28732234 DOI: 10.1016/j.compbiomed.2017.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 01/30/2023]
Abstract
Genome-scale metabolic models (GEMs) have become increasingly important in recent years. Currently, GEMs are the most accurate in silico representation of the genotype-phenotype link. They allow us to study complex networks from the systems perspective. Their application may drastically reduce the amount of experimental and clinical work, improve diagnostic tools and increase our understanding of complex biological phenomena. GEMs have also demonstrated high potential for the optimisation of bio-based production of recombinant proteins. Herein, we review the basic concepts, methods, resources and software tools used for the reconstruction and application of GEMs. We overview the evolution of the modelling efforts devoted to the metabolism of Chinese Hamster Ovary (CHO) cells. We present a case study on CHO cell metabolism under different amino acid depletions. This leads us to the identification of the most influential as well as essential amino acids in selected CHO cell lines.
Collapse
Affiliation(s)
- Živa Rejc
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Lidija Magdevska
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Tilen Tršelič
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Timotej Osolin
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Rok Vodopivec
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Jakob Mraz
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Eva Pavliha
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nikolaj Zimic
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Cvitanović
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Damjana Rozman
- Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Moškon
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia.
| | - Miha Mraz
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
29
|
He L, Wu SG, Zhang M, Chen Y, Tang YJ. WUFlux: an open-source platform for 13C metabolic flux analysis of bacterial metabolism. BMC Bioinformatics 2016; 17:444. [PMID: 27814681 PMCID: PMC5096001 DOI: 10.1186/s12859-016-1314-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 10/26/2016] [Indexed: 12/21/2022] Open
Abstract
Background Flux analyses, including flux balance analysis (FBA) and 13C-metabolic flux analysis (13C-MFA), offer direct insights into cell metabolism, and have been widely used to characterize model and non-model microbial species. Nonetheless, constructing the 13C-MFA model and performing flux calculation are demanding for new learners, because they require knowledge of metabolic networks, carbon transitions, and computer programming. To facilitate and standardize the 13C-MFA modeling work, we set out to publish a user-friendly and programming-free platform (WUFlux) for flux calculations in MATLAB®. Results We constructed an open-source platform for steady-state 13C-MFA. Using GUIDE (graphical user interface design environment) in MATLAB, we built a user interface that allows users to modify models based on their own experimental conditions. WUFlux is capable of directly correcting mass spectrum data of TBDMS (N-tert-butyldimethylsilyl-N-methyltrifluoroacetamide)-derivatized proteinogenic amino acids by removing background noise. To simplify 13C-MFA of different prokaryotic species, the software provides several metabolic network templates, including those for chemoheterotrophic bacteria and mixotrophic cyanobacteria. Users can modify the network and constraints, and then analyze the microbial carbon and energy metabolisms of various carbon substrates (e.g., glucose, pyruvate/lactate, acetate, xylose, and glycerol). WUFlux also offers several ways of visualizing the flux results with respect to the constructed network. To validate our model’s applicability, we have compared and discussed the flux results obtained from WUFlux and other MFA software. We have also illustrated how model constraints of cofactor and ATP balances influence fluxome results. Conclusion Open-source software for 13C-MFA, WUFlux, with a user-friendly interface and easy-to-modify templates, is now available at http://www.13cmfa.org/or (http://tang.eece.wustl.edu/ToolDevelopment.htm). We will continue documenting curated models of non-model microbial species and improving WUFlux performance. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1314-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lian He
- Department of Energy, Environmental and Chemical Engineering, Washington University, St. Louis, MO, 63130, USA.
| | - Stephen G Wu
- Department of Energy, Environmental and Chemical Engineering, Washington University, St. Louis, MO, 63130, USA
| | - Muhan Zhang
- Department of Computer Science and Engineering, Washington University, St. Louis, MO, 63130, USA
| | - Yixin Chen
- Department of Computer Science and Engineering, Washington University, St. Louis, MO, 63130, USA
| | - Yinjie J Tang
- Department of Energy, Environmental and Chemical Engineering, Washington University, St. Louis, MO, 63130, USA.
| |
Collapse
|
30
|
Pfau T, Pacheco MP, Sauter T. Towards improved genome-scale metabolic network reconstructions: unification, transcript specificity and beyond. Brief Bioinform 2016; 17:1060-1069. [PMID: 26615025 PMCID: PMC5142010 DOI: 10.1093/bib/bbv100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/20/2015] [Indexed: 12/24/2022] Open
Abstract
Genome-scale metabolic network reconstructions provide a basis for the investigation of the metabolic properties of an organism. There are reconstructions available for multiple organisms, from prokaryotes to higher organisms and methods for the analysis of a reconstruction. One example is the use of flux balance analysis to improve the yields of a target chemical, which has been applied successfully. However, comparison of results between existing reconstructions and models presents a challenge because of the heterogeneity of the available reconstructions, for example, of standards for presenting gene-protein-reaction associations, nomenclature of metabolites and reactions or selection of protonation states. The lack of comparability for gene identifiers or model-specific reactions without annotated evidence often leads to the creation of a new model from scratch, as data cannot be properly matched otherwise. In this contribution, we propose to improve the predictive power of metabolic models by switching from gene-protein-reaction associations to transcript-isoform-reaction associations, thus taking advantage of the improvement of precision in gene expression measurements. To achieve this precision, we discuss available databases that can be used to retrieve this type of information and point at issues that can arise from their neglect. Further, we stress issues that arise from non-standardized building pipelines, like inconsistencies in protonation states. In addition, problems arising from the use of non-specific cofactors, e.g. artificial futile cycles, are discussed, and finally efforts of the metabolic modelling community to unify model reconstructions are highlighted.
Collapse
|
31
|
Carbonell P, Currin A, Jervis AJ, Rattray NJW, Swainston N, Yan C, Takano E, Breitling R. Bioinformatics for the synthetic biology of natural products: integrating across the Design-Build-Test cycle. Nat Prod Rep 2016; 33:925-32. [PMID: 27185383 PMCID: PMC5063057 DOI: 10.1039/c6np00018e] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Indexed: 12/11/2022]
Abstract
Covering: 2000 to 2016Progress in synthetic biology is enabled by powerful bioinformatics tools allowing the integration of the design, build and test stages of the biological engineering cycle. In this review we illustrate how this integration can be achieved, with a particular focus on natural products discovery and production. Bioinformatics tools for the DESIGN and BUILD stages include tools for the selection, synthesis, assembly and optimization of parts (enzymes and regulatory elements), devices (pathways) and systems (chassis). TEST tools include those for screening, identification and quantification of metabolites for rapid prototyping. The main advantages and limitations of these tools as well as their interoperability capabilities are highlighted.
Collapse
Affiliation(s)
- Pablo Carbonell
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| | - Andrew Currin
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| | - Adrian J. Jervis
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| | - Nicholas J. W. Rattray
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| | - Neil Swainston
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| | - Cunyu Yan
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| | - Eriko Takano
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| | - Rainer Breitling
- Manchester Centre for Fine and Specialty Chemicals (SYNBIOCHEM) , Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , UK . ;
| |
Collapse
|
32
|
Cuevas DA, Edirisinghe J, Henry CS, Overbeek R, O’Connell TG, Edwards RA. From DNA to FBA: How to Build Your Own Genome-Scale Metabolic Model. Front Microbiol 2016; 7:907. [PMID: 27379044 PMCID: PMC4911401 DOI: 10.3389/fmicb.2016.00907] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/27/2016] [Indexed: 11/19/2022] Open
Abstract
Microbiological studies are increasingly relying on in silico methods to perform exploration and rapid analysis of genomic data, and functional genomics studies are supplemented by the new perspectives that genome-scale metabolic models offer. A mathematical model consisting of a microbe's entire metabolic map can be rapidly determined from whole-genome sequencing and annotating the genomic material encoded in its DNA. Flux-balance analysis (FBA), a linear programming technique that uses metabolic models to predict the phenotypic responses imposed by environmental elements and factors, is the leading method to simulate and manipulate cellular growth in silico. However, the process of creating an accurate model to use in FBA consists of a series of steps involving a multitude of connections between bioinformatics databases, enzyme resources, and metabolic pathways. We present the methodology and procedure to obtain a metabolic model using PyFBA, an extensible Python-based open-source software package aimed to provide a platform where functional annotations are used to build metabolic models (http://linsalrob.github.io/PyFBA). Backed by the Model SEED biochemistry database, PyFBA contains methods to reconstruct a microbe's metabolic map, run FBA upon different media conditions, and gap-fill its metabolism. The extensibility of PyFBA facilitates novel techniques in creating accurate genome-scale metabolic models.
Collapse
Affiliation(s)
- Daniel A. Cuevas
- Computational Science Research Center, San Diego State University, San DiegoCA, USA
| | - Janaka Edirisinghe
- Mathematics and Computer Science Division, Argonne National Laboratory, ArgonneIL, USA
| | - Chris S. Henry
- Mathematics and Computer Science Division, Argonne National Laboratory, ArgonneIL, USA
| | - Ross Overbeek
- Fellowship for Interpretation of Genomes, Burr RidgeIL, USA
| | - Taylor G. O’Connell
- Biological and Medical Informatics Research Center, San Diego State University, San DiegoCA, USA
| | - Robert A. Edwards
- Computational Science Research Center, San Diego State University, San DiegoCA, USA
- Biological and Medical Informatics Research Center, San Diego State University, San DiegoCA, USA
- Department of Computer Science, San Diego State University, San DiegoCA, USA
- Department of Biology, San Diego State University, San DiegoCA, USA
| |
Collapse
|
33
|
Guo W, Feng X. OM-FBA: Integrate Transcriptomics Data with Flux Balance Analysis to Decipher the Cell Metabolism. PLoS One 2016; 11:e0154188. [PMID: 27100883 PMCID: PMC4839607 DOI: 10.1371/journal.pone.0154188] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/11/2016] [Indexed: 11/19/2022] Open
Abstract
Constraint-based metabolic modeling such as flux balance analysis (FBA) has been widely used to simulate cell metabolism. Thanks to its simplicity and flexibility, numerous algorithms have been developed based on FBA and successfully predicted the phenotypes of various biological systems. However, their phenotype predictions may not always be accurate in FBA because of using the objective function that is assumed for cell metabolism. To overcome this challenge, we have developed a novel computational framework, namely omFBA, to integrate multi-omics data (e.g. transcriptomics) into FBA to obtain omics-guided objective functions with high accuracy. In general, we first collected transcriptomics data and phenotype data from published database (e.g. GEO database) for different microorganisms such as Saccharomyces cerevisiae. We then developed a “Phenotype Match” algorithm to derive an objective function for FBA that could lead to the most accurate estimation of the known phenotype (e.g. ethanol yield). The derived objective function was next correlated with the transcriptomics data via regression analysis to generate the omics-guided objective function, which was next used to accurately simulate cell metabolism at unknown conditions. We have applied omFBA in studying sugar metabolism of S. cerevisiae and found that the ethanol yield could be accurately predicted in most of the cases tested (>80%) by using transcriptomics data alone, and revealed valuable metabolic insights such as the dynamics of flux ratios. Overall, omFBA presents a novel platform to potentially integrate multi-omics data simultaneously and could be incorporated with other FBA-derived tools by replacing the arbitrary objective function with the omics-guided objective functions.
Collapse
Affiliation(s)
- Weihua Guo
- Department of Biological Systems Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Xueyang Feng
- Department of Biological Systems Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
34
|
Mishra P, Park GY, Lakshmanan M, Lee HS, Lee H, Chang MW, Ching CB, Ahn J, Lee DY. Genome-scale metabolic modeling and in silico analysis of lipid accumulating yeast Candida tropicalis for dicarboxylic acid production. Biotechnol Bioeng 2016; 113:1993-2004. [PMID: 26915092 DOI: 10.1002/bit.25955] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/25/2015] [Accepted: 02/14/2016] [Indexed: 02/02/2023]
Abstract
Recently, the bio-production of α,ω-dicarboxylic acids (DCAs) has gained significant attention, which potentially leads to the replacement of the conventional petroleum-based products. In this regard, the lipid accumulating yeast Candida tropicalis, has been recognized as a promising microbial host for DCA biosynthesis: it possess the unique ω-oxidation pathway where the terminal carbon of α-fatty acids is oxidized to form DCAs with varying chain lengths. However, despite such industrial importance, its cellular physiology and lipid accumulation capability remain largely uncharacterized. Thus, it is imperative to better understand the metabolic behavior of this lipogenic yeast, which could be achieved by a systems biological approach. To this end, herein, we reconstructed the genome-scale metabolic model of C. tropicalis, iCT646, accounting for 646 unique genes, 945 metabolic reactions, and 712 metabolites. Initially, the comparative network analysis of iCT646 with other yeasts revealed several distinctive metabolic reactions, mainly within the amino acid and lipid metabolism including the ω-oxidation pathway. Constraints-based flux analysis was, then, employed to predict the in silico growth rates of C. tropicalis which are highly consistent with the cellular phenotype observed in glucose and xylose minimal media chemostat cultures. Subsequently, the lipid accumulation capability of C. tropicalis was explored in comparison with Saccharomyces cerevisiae, indicating that the formation of "citrate pyruvate cycle" is essential to the lipid accumulation in oleaginous yeasts. The in silico flux analysis also highlighted the enhanced ability of pentose phosphate pathway as NADPH source rather than malic enzyme during lipogenesis. Finally, iCT646 was successfully utilized to highlight the key directions of C. tropicalis strain design for the whole cell biotransformation application to produce long-chain DCAs from alkanes. Biotechnol. Bioeng. 2016;113: 1993-2004. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pranjul Mishra
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore
| | - Gyu-Yeon Park
- Biotechnology Process Engineering Center, KRIBB, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju, 363-883, Korea.,Bioprocess Department, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon, 305-350, Korea
| | - Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Hee-Seok Lee
- Biotechnology Process Engineering Center, KRIBB, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju, 363-883, Korea.,Bioprocess Department, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon, 305-350, Korea
| | - Hongweon Lee
- Biotechnology Process Engineering Center, KRIBB, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju, 363-883, Korea.,Bioprocess Department, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon, 305-350, Korea
| | - Matthew Wook Chang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, 117456, Singapore
| | - Chi Bun Ching
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore
| | - Jungoh Ahn
- Biotechnology Process Engineering Center, KRIBB, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju, 363-883, Korea. .,Bioprocess Department, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon, 305-350, Korea.
| | - Dong-Yup Lee
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117585, Singapore. .,Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, 138668, Singapore. .,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, 117456, Singapore.
| |
Collapse
|
35
|
Lakshmanan M, Cheung CYM, Mohanty B, Lee DY. Modeling Rice Metabolism: From Elucidating Environmental Effects on Cellular Phenotype to Guiding Crop Improvement. FRONTIERS IN PLANT SCIENCE 2016; 7:1795. [PMID: 27965696 PMCID: PMC5126141 DOI: 10.3389/fpls.2016.01795] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 11/15/2016] [Indexed: 05/20/2023]
Abstract
Crop productivity is severely limited by various biotic and abiotic stresses. Thus, it is highly needed to understand the underlying mechanisms of environmental stress response and tolerance in plants, which could be addressed by systems biology approach. To this end, high-throughput omics profiling and in silico modeling can be considered to explore the environmental effects on phenotypic states and metabolic behaviors of rice crops at the systems level. Especially, the advent of constraint-based metabolic reconstruction and analysis paves a way to characterize the plant cellular physiology under various stresses by combining the mathematical network models with multi-omics data. Rice metabolic networks have been reconstructed since 2013 and currently six such networks are available, where five are at genome-scale. Since their publication, these models have been utilized to systematically elucidate the rice abiotic stress responses and identify agronomic traits for crop improvement. In this review, we summarize the current status of the existing rice metabolic networks and models with their applications. Furthermore, we also highlight future directions of rice modeling studies, particularly stressing how these models can be used to contextualize the affluent multi-omics data that are readily available in the public domain. Overall, we envisage a number of studies in the future, exploiting the available metabolic models to enhance the yield and quality of rice and other food crops.
Collapse
Affiliation(s)
- Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and ResearchSingapore, Singapore
| | - C. Y. Maurice Cheung
- Department of Chemical and Biomolecular Engineering, National University of SingaporeSingapore, Singapore
| | - Bijayalaxmi Mohanty
- Department of Chemical and Biomolecular Engineering, National University of SingaporeSingapore, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and ResearchSingapore, Singapore
- Department of Chemical and Biomolecular Engineering, National University of SingaporeSingapore, Singapore
- Synthetic Biology for Clinical and Technological Innovation, Life Sciences Institute, National University of SingaporeSingapore, Singapore
- *Correspondence: Dong-Yup Lee,
| |
Collapse
|
36
|
Olivier BG, Swat MJ, Moné MJ. Modeling and Simulation Tools: From Systems Biology to Systems Medicine. Methods Mol Biol 2016; 1386:441-63. [PMID: 26677194 DOI: 10.1007/978-1-4939-3283-2_19] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Modeling is an integral component of modern biology. In this chapter we look into the role of the model, as it pertains to Systems Medicine, and the software that is required to instantiate and run it. We do this by comparing the development, implementation, and characteristics of tools that have been developed to work with two divergent methodologies: Systems Biology and Pharmacometrics. From the Systems Biology perspective we consider the concept of "Software as a Medical Device" and what this may imply for the migration of research-oriented, simulation software into the domain of human health.In our second perspective, we see how in practice hundreds of computational tools already accompany drug discovery and development at every stage of the process. Standardized exchange formats are required to streamline the model exchange between tools, which would minimize translation errors and reduce the required time. With the emergence, almost 15 years ago, of the SBML standard, a large part of the domain of interest is already covered and models can be shared and passed from software to software without recoding them. Until recently the last stage of the process, the pharmacometric analysis used in clinical studies carried out on subject populations, lacked such an exchange medium. We describe a new emerging exchange format in Pharmacometrics which covers the non-linear mixed effects models, the standard statistical model type used in this area. By interfacing these two formats the entire domain can be covered by complementary standards and subsequently the according tools.
Collapse
Affiliation(s)
- Brett G Olivier
- Systems Bioinformatics, VU University Amsterdam, Amsterdam, The Netherlands.
| | - Maciej J Swat
- EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, UK
| | - Martijn J Moné
- Molecular Cell Physiology, VU University Amsterdam, Amsterdam, The Netherlands.,Systems and Synthetic Biology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
37
|
Marmiesse L, Peyraud R, Cottret L. FlexFlux: combining metabolic flux and regulatory network analyses. BMC SYSTEMS BIOLOGY 2015; 9:93. [PMID: 26666757 PMCID: PMC4678642 DOI: 10.1186/s12918-015-0238-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 11/27/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND Expression of cell phenotypes highly depends on metabolism that supplies matter and energy. To achieve proper utilisation of the different metabolic pathways, metabolism is tightly regulated by a complex regulatory network composed of diverse biological entities (genes, transcripts, proteins, signalling molecules…). The integrated analysis of both regulatory and metabolic networks appears very insightful but is not straightforward because of the distinct characteristics of both networks. The classical method used for metabolic flux analysis is Flux Balance Analysis (FBA), which is constraint-based and relies on the assumption of steady-state metabolite concentrations throughout the network. Regarding regulatory networks, a broad spectrum of methods are dedicated to their analysis although logical modelling remains the major method to take charge of large-scale networks. RESULTS We present FlexFlux, an application implementing a new way to combine the analysis of both metabolic and regulatory networks, based on simulations that do not require kinetic parameters and can be applied to genome-scale networks. FlexFlux is based on seeking regulatory network steady-states by performing synchronous updates of multi-state qualitative initial values. FlexFlux is then able to use the calculated steady-state values as constraints for metabolic flux analyses using FBA. As input, FlexFlux uses the standards Systems Biology Markup Language (SBML) and SBML Qualitative Models Package ("qual") extension (SBML-qual) file formats and provides a set of FBA based functions. CONCLUSIONS FlexFlux is an open-source java software with executables and full documentation available online at http://lipm-bioinfo.toulouse.inra.fr/flexflux/. It can be defined as a research tool that enables a better understanding of both regulatory and metabolic networks based on steady-state simulations. FlexFlux integrates well in the flux analysis ecosystem thanks to the support of standard file formats and can thus be used as a complementary tool to existing software featuring other types of analyses.
Collapse
Affiliation(s)
- Lucas Marmiesse
- INRA, Laboratoire des Interactions Plantes-Microrganismes (LIPM), UMR441, 24 chemin de Borde Rouge - Auzeville, CS52627, Castanet-Tolosan Cedex, F31326, France. .,CNRS, Laboratoire des Interactions Plantes-Microrganismes (LIPM), UMR2594, 24 chemin de Borde Rouge - Auzeville, CS52627, Castanet-Tolosan Cedex, F31326, France.
| | - Rémi Peyraud
- INRA, Laboratoire des Interactions Plantes-Microrganismes (LIPM), UMR441, 24 chemin de Borde Rouge - Auzeville, CS52627, Castanet-Tolosan Cedex, F31326, France. .,CNRS, Laboratoire des Interactions Plantes-Microrganismes (LIPM), UMR2594, 24 chemin de Borde Rouge - Auzeville, CS52627, Castanet-Tolosan Cedex, F31326, France.
| | - Ludovic Cottret
- INRA, Laboratoire des Interactions Plantes-Microrganismes (LIPM), UMR441, 24 chemin de Borde Rouge - Auzeville, CS52627, Castanet-Tolosan Cedex, F31326, France. .,CNRS, Laboratoire des Interactions Plantes-Microrganismes (LIPM), UMR2594, 24 chemin de Borde Rouge - Auzeville, CS52627, Castanet-Tolosan Cedex, F31326, France.
| |
Collapse
|
38
|
Guan Z, Xue D, Abdallah II, Dijkshoorn L, Setroikromo R, Lv G, Quax WJ. Metabolic engineering of Bacillus subtilis for terpenoid production. Appl Microbiol Biotechnol 2015; 99:9395-406. [PMID: 26373726 PMCID: PMC4628092 DOI: 10.1007/s00253-015-6950-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/17/2015] [Accepted: 08/20/2015] [Indexed: 11/04/2022]
Abstract
Terpenoids are the largest group of small-molecule natural products, with more than 60,000 compounds made from isopentenyl diphosphate (IPP) and its isomer dimethylallyl diphosphate (DMAPP). As the most diverse group of small-molecule natural products, terpenoids play an important role in the pharmaceutical, food, and cosmetic industries. For decades, Escherichia coli (E. coli) and Saccharomyces cerevisiae (S. cerevisiae) were extensively studied to biosynthesize terpenoids, because they are both fully amenable to genetic modifications and have vast molecular resources. On the other hand, our literature survey (20 years) revealed that terpenoids are naturally more widespread in Bacillales. In the mid-1990s, an inherent methylerythritol phosphate (MEP) pathway was discovered in Bacillus subtilis (B. subtilis). Since B. subtilis is a generally recognized as safe (GRAS) organism and has long been used for the industrial production of proteins, attempts to biosynthesize terpenoids in this bacterium have aroused much interest in the scientific community. This review discusses metabolic engineering of B. subtilis for terpenoid production, and encountered challenges will be discussed. We will summarize some major advances and outline future directions for exploiting the potential of B. subtilis as a desired "cell factory" to produce terpenoids.
Collapse
Affiliation(s)
- Zheng Guan
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Building 3215, room 917, 9713 AV, Groningen, The Netherlands
- Institute of Materia Medica, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dan Xue
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Building 3215, room 917, 9713 AV, Groningen, The Netherlands
| | - Ingy I Abdallah
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Building 3215, room 917, 9713 AV, Groningen, The Netherlands
| | - Linda Dijkshoorn
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Building 3215, room 917, 9713 AV, Groningen, The Netherlands
| | - Rita Setroikromo
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Building 3215, room 917, 9713 AV, Groningen, The Netherlands
| | - Guiyuan Lv
- Institute of Materia Medica, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wim J Quax
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Building 3215, room 917, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
39
|
Lakshmanan M, Kim TY, Chung BKS, Lee SY, Lee DY. Flux-sum analysis identifies metabolite targets for strain improvement. BMC SYSTEMS BIOLOGY 2015; 9:73. [PMID: 26510838 PMCID: PMC4625974 DOI: 10.1186/s12918-015-0198-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/18/2015] [Indexed: 11/21/2022]
Abstract
Background Rational design of microbial strains for enhanced cellular physiology through in silico analysis has been reported in many metabolic engineering studies. Such in silico techniques typically involve the analysis of a metabolic model describing the metabolic and physiological states under various perturbed conditions, thereby identifying genetic targets to be manipulated for strain improvement. More often than not, the activation/inhibition of multiple reactions is necessary to produce a predicted change for improvement of cellular properties or states. However, as it is more computationally cumbersome to simulate all possible combinations of reaction perturbations, it is desirable to consider alternative techniques for identifying such metabolic engineering targets. Results In this study, we present the modified version of previously developed metabolite-centric approach, also known as flux-sum analysis (FSA), for identifying metabolic engineering targets. Utility of FSA was demonstrated by applying it to Escherichia coli, as case studies, for enhancing ethanol and succinate production, and reducing acetate formation. Interestingly, most of the identified metabolites correspond to gene targets that have been experimentally validated in previous works on E. coli strain improvement. A notable example is that pyruvate, the metabolite target for enhancing succinate production, was found to be associated with multiple reaction targets that were only identifiable through more computationally expensive means. In addition, detailed analysis of the flux-sum perturbed conditions also provided valuable insights into how previous metabolic engineering strategies have been successful in enhancing cellular physiology. Conclusions The application of FSA under the flux balance framework can identify novel metabolic engineering targets from the metabolite-centric perspective. Therefore, the current approach opens up a new research avenue for rational design and engineering of industrial microbes in the field of systems metabolic engineering. Electronic supplementary material The online version of this article (doi:10.1186/s12918-015-0198-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
| | - Tae Yong Kim
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 program), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea. .,Center for Systems and Synthetic Biotechnology, Bioinformatics Research Center, Institute for the BioCentury, and Department of Bio and Brain Engineering, KAIST, Daejeon, 305-701, Republic of Korea.
| | - Bevan K S Chung
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
| | - Sang Yup Lee
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 program), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea. .,Center for Systems and Synthetic Biotechnology, Bioinformatics Research Center, Institute for the BioCentury, and Department of Bio and Brain Engineering, KAIST, Daejeon, 305-701, Republic of Korea.
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore. .,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117576, Singapore. .,Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore.
| |
Collapse
|
40
|
Lakshmanan M, Yu K, Koduru L, Lee DY. In silico model-driven cofactor engineering strategies for improving the overall NADP(H) turnover in microbial cell factories. J Ind Microbiol Biotechnol 2015; 42:1401-14. [PMID: 26254041 DOI: 10.1007/s10295-015-1663-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/23/2015] [Indexed: 02/03/2023]
Abstract
Optimizing the overall NADPH turnover is one of the key challenges in various value-added biochemical syntheses. In this work, we first analyzed the NADPH regeneration potentials of common cell factories, including Escherichia coli, Saccharomyces cerevisiae, Bacillus subtilis, and Pichia pastoris across multiple environmental conditions and determined E. coli and glycerol as the best microbial chassis and most suitable carbon source, respectively. In addition, we identified optimal cofactor specificity engineering (CSE) enzyme targets, whose cofactors when switched from NAD(H) to NADP(H) improve the overall NADP(H) turnover. Among several enzyme targets, glyceraldehyde-3-phosphate dehydrogenase was recognized as a global candidate since its CSE improved the NADP(H) regeneration under most of the conditions examined. Finally, by analyzing the protein structures of all CSE enzyme targets via homology modeling, we established that the replacement of conserved glutamate or aspartate with serine in the loop region could change the cofactor dependence from NAD(H) to NADP(H).
Collapse
Affiliation(s)
- Meiyappan Lakshmanan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, Singapore, 138668, Singapore
| | - Kai Yu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore
| | - Lokanand Koduru
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Centros, Singapore, 138668, Singapore. .,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore. .,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore.
| |
Collapse
|
41
|
Flux Balance Analysis with Objective Function Defined by Proteomics Data-Metabolism of Mycobacterium tuberculosis Exposed to Mefloquine. PLoS One 2015. [PMID: 26218987 PMCID: PMC4517854 DOI: 10.1371/journal.pone.0134014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We present a study of the metabolism of the Mycobacterium tuberculosis after exposure to antibiotics using proteomics data and flux balance analysis (FBA). The use of FBA to study prokaryotic organisms is well-established and allows insights into the metabolic pathways chosen by the organisms under different environmental conditions. To apply FBA a specific objective function must be selected that represents the metabolic goal of the organism. FBA estimates the metabolism of the cell by linear programming constrained by the stoichiometry of the reactions in an in silico metabolic model of the organism. It is assumed that the metabolism of the organism works towards the specified objective function. A common objective is the maximization of biomass. However, this goal is not suitable for situations when the bacterium is exposed to antibiotics, as the goal of organisms in these cases is survival and not necessarily optimal growth. In this paper we propose a new approach for defining the FBA objective function in studies when the bacterium is under stress. The function is defined based on protein expression data. The proposed methodology is applied to the case when the bacterium is exposed to the drug mefloquine, but can be easily extended to other organisms, conditions or drugs. We compare our method with an alternative method that uses experimental data for adjusting flux constraints. We perform comparisons in terms of essential enzymes and agreement using enzyme abundances. Results indicate that using proteomics data to define FBA objective functions yields less essential reactions with zero flux and lower error rates in prediction accuracy. With flux variability analysis we observe that overall variability due to alternate optima is reduced with the incorporation of proteomics data. We believe that incorporating proteomics data in the objective function used in FBA may help obtain metabolic flux representations that better support experimentally observed features.
Collapse
|
42
|
Bayer T, Milker S, Wiesinger T, Rudroff F, Mihovilovic MD. Designer Microorganisms for Optimized Redox Cascade Reactions - Challenges and Future Perspectives. Adv Synth Catal 2015. [DOI: 10.1002/adsc.201500202] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
43
|
Ferrer Valenzuela J, Pinuer LA, García Cancino A, Bórquez Yáñez R. Metabolic Fluxes in Lactic Acid Bacteria—A Review. FOOD BIOTECHNOL 2015. [DOI: 10.1080/08905436.2015.1027913] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Membrane transporter engineering in industrial biotechnology and whole cell biocatalysis. Trends Biotechnol 2015; 33:237-46. [DOI: 10.1016/j.tibtech.2015.02.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/15/2015] [Accepted: 02/02/2015] [Indexed: 02/06/2023]
|
45
|
Using bioconductor package BiGGR for metabolic flux estimation based on gene expression changes in brain. PLoS One 2015; 10:e0119016. [PMID: 25806817 PMCID: PMC4373785 DOI: 10.1371/journal.pone.0119016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 01/08/2015] [Indexed: 01/13/2023] Open
Abstract
Predicting the distribution of metabolic fluxes in biochemical networks is of major interest in systems biology. Several databases provide metabolic reconstructions for different organisms. Software to analyze flux distributions exists, among others for the proprietary MATLAB environment. Given the large user community for the R computing environment, a simple implementation of flux analysis in R appears desirable and will facilitate easy interaction with computational tools to handle gene expression data. We extended the R software package BiGGR, an implementation of metabolic flux analysis in R. BiGGR makes use of public metabolic reconstruction databases, and contains the BiGG database and the reconstruction of human metabolism Recon2 as Systems Biology Markup Language (SBML) objects. Models can be assembled by querying the databases for pathways, genes or reactions of interest. Fluxes can then be estimated by maximization or minimization of an objective function using linear inverse modeling algorithms. Furthermore, BiGGR provides functionality to quantify the uncertainty in flux estimates by sampling the constrained multidimensional flux space. As a result, ensembles of possible flux configurations are constructed that agree with measured data within precision limits. BiGGR also features automatic visualization of selected parts of metabolic networks using hypergraphs, with hyperedge widths proportional to estimated flux values. BiGGR supports import and export of models encoded in SBML and is therefore interoperable with different modeling and analysis tools. As an application example, we calculated the flux distribution in healthy human brain using a model of central carbon metabolism. We introduce a new algorithm termed Least-squares with equalities and inequalities Flux Balance Analysis (Lsei-FBA) to predict flux changes from gene expression changes, for instance during disease. Our estimates of brain metabolic flux pattern with Lsei-FBA for Alzheimer’s disease agree with independent measurements of cerebral metabolism in patients. This second version of BiGGR is available from Bioconductor.
Collapse
|
46
|
Asgari Y, Zabihinpour Z, Salehzadeh-Yazdi A, Schreiber F, Masoudi-Nejad A. Alterations in cancer cell metabolism: the Warburg effect and metabolic adaptation. Genomics 2015; 105:275-81. [PMID: 25773945 DOI: 10.1016/j.ygeno.2015.03.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/09/2015] [Accepted: 03/04/2015] [Indexed: 12/13/2022]
Abstract
The Warburg effect means higher glucose uptake of cancer cells compared to normal tissues, whereas a smaller fraction of this glucose is employed for oxidative phosphorylation. With the advent of high throughput technologies and computational systems biology, cancer cell metabolism has been reinvestigated over the last decades toward identifying various events underlying "how" and "why" a cancer cell employs aerobic glycolysis. Significant progress has been shaped to revise the Warburg effect. In this study, we have integrated the gene expression of 13 different cancer cells with the genome-scale metabolic network of human (Recon1) based on the E-Flux method, and analyzed them based on constraint-based modeling. Results show that regardless of significant up- and down-regulated metabolic genes, the distribution of metabolic changes is similar in different cancer types. These findings support the theory that the Warburg effect is a consequence of metabolic adaptation in cancer cells.
Collapse
Affiliation(s)
- Yazdan Asgari
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Zahra Zabihinpour
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Salehzadeh-Yazdi
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Falk Schreiber
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle, Germany; Clayton School of Information Technology, Monash University, Clayton, VIC, Australia.
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
47
|
Ravikrishnan A, Raman K. Critical assessment of genome-scale metabolic networks: the need for a unified standard. Brief Bioinform 2015; 16:1057-68. [PMID: 25725218 DOI: 10.1093/bib/bbv003] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Indexed: 12/17/2022] Open
Abstract
Genome-scale metabolic networks have been reconstructed for several organisms. These metabolic networks provide detailed information about the metabolism inside the cells, coupled with the genomic, proteomic and thermodynamic information. These networks are widely simulated using 'constraint-based' modelling techniques and find applications ranging from strain improvement for metabolic engineering to prediction of drug targets in pathogenic organisms. Components of these metabolic networks are represented in multiple file formats and also using different markup languages, with varying levels of annotations; this leads to inconsistencies and increases the complexities in comparing and analysing reconstructions on multiple platforms. In this work, we critically examine nearly 100 published genome-scale metabolic networks and their corresponding constraint-based models and discuss various issues with respect to model quality. One of the major concerns is the lack of annotations using standard identifiers that can uniquely describe several components such as metabolites, genes, proteins and reactions. We also find that many models do not have complete information regarding constraints on reactions fluxes and objective functions for carrying out simulations. Overall, our analysis highlights the need for a widely acceptable standard for representing constraint-based models. A rigorous standard can help in streamlining the process of reconstruction and improve the quality of reconstructed metabolic models.
Collapse
|
48
|
Systems Approaches to Study Infectious Diseases. SYSTEMS AND SYNTHETIC BIOLOGY 2015. [DOI: 10.1007/978-94-017-9514-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
49
|
Narang P, Khan S, Hemrom AJ, Lynn AM. MetaNET--a web-accessible interactive platform for biological metabolic network analysis. BMC SYSTEMS BIOLOGY 2014; 8:130. [PMID: 25779921 PMCID: PMC4267457 DOI: 10.1186/s12918-014-0130-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 11/12/2014] [Indexed: 11/10/2022]
Abstract
Background Metabolic reactions have been extensively studied and compiled over the last century. These have provided a theoretical base to implement models, simulations of which are used to identify drug targets and optimize metabolic throughput at a systemic level. While tools for the perturbation of metabolic networks are available, their applications are limited and restricted as they require varied dependencies and often a commercial platform for full functionality. We have developed MetaNET, an open source user-friendly platform-independent and web-accessible resource consisting of several pre-defined workflows for metabolic network analysis. Result MetaNET is a web-accessible platform that incorporates a range of functions which can be combined to produce different simulations related to metabolic networks. These include (i) optimization of an objective function for wild type strain, gene/catalyst/reaction knock-out/knock-down analysis using flux balance analysis. (ii) flux variability analysis (iii) chemical species participation (iv) cycles and extreme paths identification and (v) choke point reaction analysis to facilitate identification of potential drug targets. The platform is built using custom scripts along with the open-source Galaxy workflow and Systems Biology Research Tool as components. Pre-defined workflows are available for common processes, and an exhaustive list of over 50 functions are provided for user defined workflows. Conclusion MetaNET, available at http://metanet.osdd.net, provides a user-friendly rich interface allowing the analysis of genome-scale metabolic networks under various genetic and environmental conditions. The framework permits the storage of previous results, the ability to repeat analysis and share results with other users over the internet as well as run different tools simultaneously using pre-defined workflows, and user-created custom workflows.
Collapse
Affiliation(s)
- Pankaj Narang
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, 110067, India,
| | | | | | | | | |
Collapse
|
50
|
Kelley JJ, Lane A, Li X, Mutthoju B, Maor S, Egen D, Lun DS. MOST: a software environment for constraint-based metabolic modeling and strain design. Bioinformatics 2014; 31:610-1. [PMID: 25677126 DOI: 10.1093/bioinformatics/btu685] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
SUMMARY MOST (metabolic optimization and simulation tool) is a software package that implements GDBB (genetic design through branch and bound) in an intuitive user-friendly interface with excel-like editing functionality, as well as implementing FBA (flux balance analysis), and supporting systems biology markup language and comma-separated values files. GDBB is currently the fastest algorithm for finding gene knockouts predicted by FBA to increase production of desired products, but GDBB has only been available on a command line interface, which is difficult to use for those without programming knowledge, until the release of MOST. AVAILABILITY AND IMPLEMENTATION MOST is distributed for free on the GNU General Public License. The software and full documentation are available at http://most.ccib.rutgers.edu/. CONTACT dslun@rutgers.edu.
Collapse
Affiliation(s)
- James J Kelley
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Anatoliy Lane
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Xiaowei Li
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Brahmaji Mutthoju
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Shay Maor
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Dennis Egen
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Desmond S Lun
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ 08102, USA and Phenomics and Bioinformatics Research Centre and School of Mathematics and Statistics, University of South Australia, Mawson Lakes, SA 5095, Australia
| |
Collapse
|