1
|
Fitzpatrick TB. B Vitamins: An Update on Their Importance for Plant Homeostasis. ANNUAL REVIEW OF PLANT BIOLOGY 2024; 75:67-93. [PMID: 38424064 DOI: 10.1146/annurev-arplant-060223-025336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
B vitamins are a source of coenzymes for a vast array of enzyme reactions, particularly those of metabolism. As metabolism is the basis of decisions that drive maintenance, growth, and development, B vitamin-derived coenzymes are key components that facilitate these processes. For over a century, we have known about these essential compounds and have elucidated their pathways of biosynthesis, repair, salvage, and degradation in numerous organisms. Only now are we beginning to understand their importance for regulatory processes, which are becoming an important topic in plants. Here, I highlight and discuss emerging evidence on how B vitamins are integrated into vital processes, from energy generation and nutrition to gene expression, and thereby contribute to the coordination of growth and developmental programs, particularly those that concern maintenance of a stable state, which is the foundational tenet of plant homeostasis.
Collapse
|
2
|
Kochel B. Negative feedback systems for modelling NF-κB transcription factor oscillatory activity. Transcription 2024; 15:65-96. [PMID: 38739365 PMCID: PMC11810101 DOI: 10.1080/21541264.2024.2331887] [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] [Received: 10/27/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 05/14/2024] Open
Abstract
Low-dimensional negative feedback systems (NFSs) were developed within a signal flow model to describe the oscillatory activities of NF-κB caused by interactions with its inhibitor IκBα. The NFSs were established as 3rd- and 4th-order linear systems containing unperturbed and perturbed negative feedback (NF) loops with constant or time-varying NF strengths and a feed-forward loop. NF-related analytical solutions to the NFSs representing the time courses of NF-κB and IκBα were determined and their exact mathematical relationship was found. The NFS's parameters were determined to fit the experimental time courses of NF-κB in TNF-α-stimulated embryonic fibroblasts, rela-/- embryonic fibroblasts reconstituted with RelA, C9L cells, GFP-p65 knock-in embryonic fibroblasts and embryogenic fibroblasts lacking Iκβ and IκBε, LPS-stimulated IC-21 macrophages treated or not with DCPA, and anti-IgM-stimulated DT40 B-lymphocytes. The unperturbed and perturbed NFSs describing the above biosystems generated isochronous and non-isochronous solutions, depending on a constant or time-varying NF strength, respectively. The oscillation period of the NF-coupled solutions, the phase difference between them and the time delays in the appearance of cytoplasmic IκBα after stimulation of NF-κB were determined. A significant divergence between the IκBα solutions to the NFSs and the IκBα experimental courses led to a rejection of the NF coupling between NF-κB and IκBα in the above biosystems. It was shown that neither the linearity nor the low dimensionality of the NFSs altered the NF relationship and the divergence between the IκBα solutions to the NFS and IκBα experimental time courses. Although the NF relationship between IκBα and NF-κB was not confirmed in all the experimental data analyzed, delayed negative feedback was found in some cases.
Collapse
Affiliation(s)
- Bonawentura Kochel
- Immunotherapy Central Europe, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
3
|
Andrews SS, Wiley HS, Sauro HM. Design patterns of biological cells. Bioessays 2024; 46:e2300188. [PMID: 38247191 PMCID: PMC10922931 DOI: 10.1002/bies.202300188] [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: 09/30/2023] [Revised: 12/03/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024]
Abstract
Design patterns are generalized solutions to frequently recurring problems. They were initially developed by architects and computer scientists to create a higher level of abstraction for their designs. Here, we extend these concepts to cell biology to lend a new perspective on the evolved designs of cells' underlying reaction networks. We present a catalog of 21 design patterns divided into three categories: creational patterns describe processes that build the cell, structural patterns describe the layouts of reaction networks, and behavioral patterns describe reaction network function. Applying this pattern language to the E. coli central metabolic reaction network, the yeast pheromone response signaling network, and other examples lends new insights into these systems.
Collapse
Affiliation(s)
- Steven S. Andrews
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - H. Steven Wiley
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Herbert M. Sauro
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
4
|
Cornish-Bowden A, Cárdenas ML. Evolution of Henrik Kacser's thought: Early publications on the organization of the whole system. Biosystems 2023; 226:104883. [PMID: 36931555 DOI: 10.1016/j.biosystems.2023.104883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Although the papers of Kacser and Burns (1973) and Heinrich and Rapoport (1974a,b) are commonly taken as the birth of metabolic control analysis, many of the ideas in them are foreshadowed in earlier papers, from 1956 onwards, when Kacser first argued for taking a systemic view of genetics and biochemistry.
Collapse
|
5
|
Gabrielli N, Maga-Nteve C, Kafkia E, Rettel M, Loeffler J, Kamrad S, Typas A, Patil KR. Unravelling metabolic cross-feeding in a yeast-bacteria community using 13 C-based proteomics. Mol Syst Biol 2023; 19:e11501. [PMID: 36779294 PMCID: PMC10090948 DOI: 10.15252/msb.202211501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/14/2023] Open
Abstract
Cross-feeding is fundamental to the diversity and function of microbial communities. However, identification of cross-fed metabolites is often challenging due to the universality of metabolic and biosynthetic intermediates. Here, we use 13 C isotope tracing in peptides to elucidate cross-fed metabolites in co-cultures of Saccharomyces cerevisiae and Lactococcus lactis. The community was grown on lactose as the main carbon source with either glucose or galactose fraction of the molecule labelled with 13 C. Data analysis allowing for the possible mass-shifts yielded hundreds of peptides for which we could assign both species identity and labelling degree. The labelling pattern showed that the yeast utilized galactose and, to a lesser extent, lactic acid shared by L. lactis as carbon sources. While the yeast provided essential amino acids to the bacterium as expected, the data also uncovered a complex pattern of amino acid exchange. The identity of the cross-fed metabolites was further supported by metabolite labelling in the co-culture supernatant, and by diminished fitness of a galactose-negative yeast mutant in the community. Together, our results demonstrate the utility of 13 C-based proteomics for uncovering microbial interactions.
Collapse
Affiliation(s)
| | | | - Eleni Kafkia
- European Molecular Biology Laboratory, Heidelberg, Germany.,Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Mandy Rettel
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jakob Loeffler
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Stephan Kamrad
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | | | - Kiran Raosaheb Patil
- European Molecular Biology Laboratory, Heidelberg, Germany.,Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
6
|
Dynamics and Sensitivity of Signaling Pathways. CURRENT PATHOBIOLOGY REPORTS 2022; 10:11-22. [PMID: 36969954 PMCID: PMC10035447 DOI: 10.1007/s40139-022-00230-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Purpose of Review Signaling pathways serve to communicate information about extracellular conditions into the cell, to both the nucleus and cytoplasmic processes to control cell responses. Genetic mutations in signaling network components are frequently associated with cancer and can result in cells acquiring an ability to divide and grow uncontrollably. Because signaling pathways play such a significant role in cancer initiation and advancement, their constituent proteins are attractive therapeutic targets. In this review, we discuss how signaling pathway modeling can assist with identifying effective drugs for treating diseases, such as cancer. An achievement that would facilitate the use of such models is their ability to identify controlling biochemical parameters in signaling pathways, such as molecular abundances and chemical reaction rates, because this would help determine effective points of attack by therapeutics. Recent Findings We summarize the current state of understanding the sensitivity of phosphorylation cycles with and without sequestration. We also describe some basic properties of regulatory motifs including feedback and feedforward regulation. Summary Although much recent work has focused on understanding the dynamics and particularly the sensitivity of signaling networks in eukaryotic systems, there is still an urgent need to build more scalable models of signaling networks that can appropriately represent their complexity across different cell types and tumors.
Collapse
|
7
|
Hoermann R, Pekker MJ, Midgley JEM, Larisch R, Dietrich JW. Principles of Endocrine Regulation: Reconciling Tensions Between Robustness in Performance and Adaptation to Change. Front Endocrinol (Lausanne) 2022; 13:825107. [PMID: 35757421 PMCID: PMC9219553 DOI: 10.3389/fendo.2022.825107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/02/2022] [Indexed: 02/06/2023] Open
Abstract
Endocrine regulation in the hypothalamic-pituitary-thyroid (HPT) axis is orchestrated by physiological circuits which integrate multiple internal and external influences. Essentially, it provides either of the two responses to overt biological challenges: to defend the homeostatic range of a target hormone or adapt it to changing environmental conditions. Under certain conditions, such flexibility may exceed the capability of a simple feedback control loop, rather requiring more intricate networks of communication between the system's components. A new minimal mathematical model, in the form of a parametrized nonlinear dynamical system, is here formulated as a proof-of-concept to elucidate the principles of the HPT axis regulation. In particular, it allows uncovering mechanisms for the homeostasis of the key biologically active hormone free triiodothyronine (FT3). One mechanism supports the preservation of FT3 homeostasis, whilst the other is responsible for the adaptation of the homeostatic state to a new level. Together these allow optimum resilience in stressful situations. Preservation of FT3 homeostasis, despite changes in FT4 and TSH levels, is found to be an achievable system goal by joining elements of top-down and bottom-up regulation in a cascade of targeted feedforward and feedback loops. Simultaneously, the model accounts for the combination of properties regarded as essential to endocrine regulation, namely sensitivity, the anticipation of an adverse event, robustness, and adaptation. The model therefore offers fundamental theoretical insights into the effective system control of the HPT axis.
Collapse
Affiliation(s)
- Rudolf Hoermann
- Department for Nuclear Medicine, Klinikum Lüdenscheid, Lüdenscheid, Germany
| | - Mark J. Pekker
- Mathematical Sciences Department, University of Alabama, Huntsville, AL, United States
| | | | - Rolf Larisch
- Department for Nuclear Medicine, Klinikum Lüdenscheid, Lüdenscheid, Germany
| | - Johannes W. Dietrich
- Diabetes, Endocrinology and Metabolism Section, Department of Medicine I, St. Josef Hospital, Ruhr-University of Bochum, Bochum, Germany
- Diabetes Centre Bochum/Hattingen, Ruhr University of Bochum, Bochum, Germany
- Ruhr Center for Rare Diseases (CeSER), Ruhr University of Bochum and Witten/Herdecke University, Bochum, Germany
| |
Collapse
|
8
|
Combined Transcriptomic and Protein Array Cytokine Profiling of Human Stem Cells from Dental Apical Papilla Modulated by Oral Bacteria. Int J Mol Sci 2022; 23:ijms23095098. [PMID: 35563488 PMCID: PMC9103834 DOI: 10.3390/ijms23095098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/22/2022] [Accepted: 04/30/2022] [Indexed: 11/18/2022] Open
Abstract
Stem cells from the apical papilla (SCAP) are a promising resource for use in regenerative endodontic treatment (RET) that may be adversely affected by oral bacteria, which in turn can exert an effect on the success of RET. Our work aims to study the cytokine profile of SCAP upon exposure to oral bacteria and their supernatants—Fusobacterium nucleatum and Enterococcus faecalis—as well as to establish their effect on the osteogenic and immunogenic potentials of SCAP. Further, we target the presence of key proteins of the Wnt/β-Catenin, TGF-β, and NF-κB signaling pathways, which play a crucial role in adult osteogenic differentiation of mesenchymal stem cells, using the Western blot (WB) technique. The membrane-based sandwich immunoassay and transcriptomic analysis showed that, under the influence of F. nucleatum (both bacteria and supernatant), the production of pro-inflammatory cytokines IL-6, IL-8, and MCP-1 occurred, which was also confirmed at the mRNA level. Conversely, E. faecalis reduced the secretion of the aforementioned cytokines at both mRNA and protein levels. WB analysis showed that SCAP co-cultivation with E. faecalis led to a decrease in the level of the key proteins of the Wnt/β-Catenin and NF-κB signaling pathways: β-Catenin (p = 0.0068 *), LRP-5 (p = 0.0059 **), and LRP-6 (p = 0.0329 *), as well as NF-kB (p = 0.0034 **) and TRAF6 (p = 0.0285 *). These results suggest that oral bacteria can up- and downregulate the immune and inflammatory responses of SCAP, as well as influence the osteogenic potential of SCAP, which may negatively regulate the success of RET.
Collapse
|
9
|
Goulooze SC, Snelder N, Seelmann A, Horvat-Broecker A, Brinker M, Joseph A, Garmann D, Lippert J, Eissing T. Finerenone Dose-Exposure-Serum Potassium Response Analysis of FIDELIO-DKD Phase III: The Role of Dosing, Titration, and Inclusion Criteria. Clin Pharmacokinet 2022; 61:451-462. [PMID: 34786651 PMCID: PMC8891103 DOI: 10.1007/s40262-021-01083-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Finerenone is a nonsteroidal selective mineralocorticoid receptor antagonist (MRA) that demonstrated efficacy in delaying the progression of chronic kidney disease (CKD) and reducing cardiovascular events in patients with CKD and type 2 diabetes mellitus in FIDELIO-DKD, where 5734 patients were randomized 1:1 to receive either finerenone or placebo, with a median follow-up of 2.6 years. Doses of finerenone 10 or 20 mg once daily were titrated based on (serum) potassium and estimated glomerular filtration rate. The MRA mode of action increases potassium. METHODS Nonlinear mixed-effects population pharmacokinetic/pharmacodynamic models were used to analyze the finerenone dose-exposure-response relationship for potassium in FIDELIO-DKD. Individual time-varying exposures from pharmacokinetic analyses were related to the potassium response via a maximal effect, indirect-response model informed by 148,384 serum potassium measurements. RESULTS Although observed potassium levels decreased with increasing dose (i.e., inverse relation), model-based simulations for a fixed-dose setting (i.e., no dose titration) revealed the intrinsic finerenone dose-exposure-potassium response, with potassium levels increasing in a dose- and exposure-dependent manner, thus explaining the apparent conflict. The potassium limit for inclusion and uptitration from finerenone 10 to 20 mg in FIDELIO-DKD was ≤ 4.8 mmol/L. Modified limits of ≤ 5.0 mmol/L were simulated, resulting in higher hyperkalemia frequencies for both the finerenone and the placebo arms, whereas the relative hyperkalemia risk of a finerenone treatment compared with placebo did not increase. CONCLUSIONS The analyses demonstrated the effectiveness of finerenone dose titration in managing serum potassium and provide a quantitative basis to guide safe clinical use.
Collapse
Affiliation(s)
| | - Nelleke Snelder
- Leiden Experts on Advanced Pharmacokinetics and Pharmacodynamics (LAP&P), Leiden, The Netherlands
| | - Andreas Seelmann
- Bayer AG, Pharmaceuticals R&D, Pharmacometrics, Leverkusen/Wuppertal/Berlin, Germany
| | | | - Meike Brinker
- Bayer AG, Pharmaceuticals R&D, Clinical Development, Wuppertal/Berlin, Germany
| | - Amer Joseph
- Bayer AG, Pharmaceuticals R&D, Clinical Development, Wuppertal/Berlin, Germany
| | - Dirk Garmann
- Bayer AG, Pharmaceuticals R&D, Pharmacometrics, Leverkusen/Wuppertal/Berlin, Germany
| | - Joerg Lippert
- Bayer AG, Pharmaceuticals R&D, Pharmacometrics, Leverkusen/Wuppertal/Berlin, Germany
| | - Thomas Eissing
- Bayer AG, Pharmaceuticals R&D, Pharmacometrics, Leverkusen/Wuppertal/Berlin, Germany.
| |
Collapse
|
10
|
Hancock EJ, Krycer JR, Ang J. Metabolic buffer analysis reveals the simultaneous, independent control of ATP and adenylate energy ratios. J R Soc Interface 2021; 18:20200976. [PMID: 33906384 PMCID: PMC8086841 DOI: 10.1098/rsif.2020.0976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/15/2021] [Indexed: 11/12/2022] Open
Abstract
Determining the underlying principles behind biological regulation is important for understanding the principles of life, treating complex diseases and creating de novo synthetic biology. Buffering-the use of reservoirs of molecules to maintain molecular concentrations-is a widespread and important mechanism for biological regulation. However, a lack of theory has limited our understanding of its roles and quantified effects. Here, we study buffering in energy metabolism using control theory and novel buffer analysis. We find that buffering can enable the simultaneous, independent control of multiple coupled outputs. In metabolism, adenylate kinase and AMP deaminase enable simultaneous control of ATP and adenylate energy ratios, while feedback on metabolic pathways is fundamentally limited to controlling one of these outputs. We also quantify the regulatory effects of the phosphagen system-the above buffers and creatine kinase-revealing which mechanisms regulate which outputs. The results are supported by human muscle and mouse adipocyte data. Together, these results illustrate the synergy of feedback and buffering in molecular biology to simultaneously control multiple outputs.
Collapse
Affiliation(s)
- Edward J. Hancock
- School of Mathematics and Statistics, Sydney, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, 2006, Australia
| | - James R. Krycer
- School of Life and Environmental Sciences, Sydney, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, 2006, Australia
| | - Jordan Ang
- Synthace Ltd, London, W12 7FQ, UK
- Department of Chemical and Physical Sciences, University of Toronto, Mississauga, ON L5L1C6, Canada
- Department of Immunology, University of Toronto, Toronto, ON, M5S1A8, Canada
| |
Collapse
|
11
|
Gao H, Ma H, Sun J, Xu W, Gao W, Lai X, Yan B. Expression and function analysis of crustacyanin gene family involved in resistance to heavy metal stress and body color formation in Exopalaemon carinicauda. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2021; 336:352-363. [PMID: 33465290 DOI: 10.1002/jez.b.23025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 11/09/2022]
Abstract
Crustacyanin has the function of binding astaxanthin which is the best antioxidant, and plays an important role in the body color variation of crustaceans. To investigate the causes of body color variation of the ridgetail white prawn, Exopalaemon carinicauda, the present study obtained four subtypes of crustacyanin gene: C1, C2, A1, and A2. Based on fluorescence quantitative polymerase chain reaction, lipocalin-C1 is mainly expressed in the eyestalk, lipocalin-C2 is in the ventral nerve cord, and lipocalin-A1 and lipocalin-A2 are in subcutaneous adipose tissues. Under the inhibiting effect of Cd2+ stress, the expression of four subtypes first increases and then decreases within 24 h, and reaches the maximum at 6 or 12 h. RNA interference experiments showed a decrease in the expression of lipocalin genes in subcutaneous adipose tissue for each subtype, with the body color changing from transparent to red, and the dark red spots on the epidermis changing to bright red. Moreover, the blue protein in the subcutaneous adipose tissue largely disappeared, based on the light micrographs. In view of these findings, the crustacyanin gene appears to fulfill some function in the resistance to heavy metal stress and body color formation of E. carinicauda.
Collapse
Affiliation(s)
- Huan Gao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, China.,Marine Resource Development institute of Jiangsu (Lianyungang), Lianyungang, Jiangsu, China.,The Jiangsu Provincial Infrastructure for Conservation and Utilization of Agricultural Germplasm, Nanjing, China
| | - Hangke Ma
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, China
| | - Jinqiu Sun
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, China
| | - Wanyuan Xu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, China
| | - Wei Gao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, China
| | - Xiaofang Lai
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, China.,Marine Resource Development institute of Jiangsu (Lianyungang), Lianyungang, Jiangsu, China.,The Jiangsu Provincial Infrastructure for Conservation and Utilization of Agricultural Germplasm, Nanjing, China
| | - Binlun Yan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, China.,Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, China.,Marine Resource Development institute of Jiangsu (Lianyungang), Lianyungang, Jiangsu, China.,The Jiangsu Provincial Infrastructure for Conservation and Utilization of Agricultural Germplasm, Nanjing, China
| |
Collapse
|
12
|
Desmet S, Brouckaert M, Boerjan W, Morreel K. Seeing the forest for the trees: Retrieving plant secondary biochemical pathways from metabolome networks. Comput Struct Biotechnol J 2020; 19:72-85. [PMID: 33384856 PMCID: PMC7753198 DOI: 10.1016/j.csbj.2020.11.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 02/06/2023] Open
Abstract
Over the last decade, a giant leap forward has been made in resolving the main bottleneck in metabolomics, i.e., the structural characterization of the many unknowns. This has led to the next challenge in this research field: retrieving biochemical pathway information from the various types of networks that can be constructed from metabolome data. Searching putative biochemical pathways, referred to as biotransformation paths, is complicated because several flaws occur during the construction of metabolome networks. Multiple network analysis tools have been developed to deal with these flaws, while in silico retrosynthesis is appearing as an alternative approach. In this review, the different types of metabolome networks, their flaws, and the various tools to trace these biotransformation paths are discussed.
Collapse
Affiliation(s)
- Sandrien Desmet
- Ghent University, Department of Plant Biotechnology and Bioinformatics, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Marlies Brouckaert
- Ghent University, Department of Plant Biotechnology and Bioinformatics, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Wout Boerjan
- Ghent University, Department of Plant Biotechnology and Bioinformatics, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Kris Morreel
- Ghent University, Department of Plant Biotechnology and Bioinformatics, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| |
Collapse
|
13
|
A brief note on the properties of linear pathways. Biochem Soc Trans 2020; 48:1379-1395. [PMID: 32830848 DOI: 10.1042/bst20190842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 11/17/2022]
Abstract
Linear metabolic pathways are the simplest network architecture we find in metabolism and are a good starting point to gain insight into the operating principles of metabolic control. Linear pathways possess some well-known properties, such as a bias of flux control towards the first few steps of the pathway as well as the lack of flux control at reactions close to equilibrium. In both cases, a rationale for these behaviors is given in terms of how elasticities transmit changes through a pathway. A discussion is given on the fundamental role that two reaction step sections play in a linear pathway when transmitting changes. For a pathway with irreversible steps, the deconstruction is straight forward and includes a product of local response coefficients that cascade along the pathway. When reversibility is included, the picture became more complex but a relationship in terms of the local response coefficients if derived that includes the reverse response coefficients and highlights the interplay between the forward and backward transmission of changes during a perturbation.
Collapse
|
14
|
Bromig L, Kremling A, Marin-Sanguino A. Understanding biochemical design principles with ensembles of canonical non-linear models. PLoS One 2020; 15:e0230599. [PMID: 32353072 PMCID: PMC7192416 DOI: 10.1371/journal.pone.0230599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/03/2020] [Indexed: 12/22/2022] Open
Abstract
Systems biology applies concepts from engineering in order to understand biological networks. If such an understanding was complete, biologists would be able to design ad hoc biochemical components tailored for different purposes, which is the goal of synthetic biology. Needless to say that we are far away from creating biological subsystems as intricate and precise as those found in nature, but mathematical models and high throughput techniques have brought us a long way in this direction. One of the difficulties that still needs to be overcome is finding the right values for model parameters and dealing with uncertainty, which is proving to be an extremely difficult task. In this work, we take advantage of ensemble modeling techniques, where a large number of models with different parameter values are formulated and then tested according to some performance criteria. By finding features shared by successful models, the role of different components and the synergies between them can be better understood. We will address some of the difficulties often faced by ensemble modeling approaches, such as the need to sample a space whose size grows exponentially with the number of parameters, and establishing useful selection criteria. Some methods will be shown to reduce the predictions from many models into a set of understandable “design principles” that can guide us to improve or manufacture a biochemical network. Our proposed framework formulates models within standard formalisms in order to integrate information from different sources and minimize the dimension of the parameter space. Additionally, the mathematical properties of the formalism enable a partition of the parameter space into independent subspaces. Each of these subspaces can be paired with a set of criteria that depend exclusively on it, thus allowing a separate sampling/screening in spaces of lower dimension. By applying tests in a strict order where computationally cheaper tests are applied first to each subspace and applying computationally expensive tests to the remaining subset thereafter, the use of resources is optimized and a larger number of models can be examined. This can be compared to a complex database query where the order of the requests can make a huge difference in the processing time. The method will be illustrated by analyzing a classical model of a metabolic pathway with end-product inhibition. Even for such a simple model, the method provides novel insight.
Collapse
Affiliation(s)
- Lukas Bromig
- Specialty Division for Systems Biotechnology, Technische Universität München, Garching, Germany
| | - Andreas Kremling
- Specialty Division for Systems Biotechnology, Technische Universität München, Garching, Germany
| | - Alberto Marin-Sanguino
- Specialty Division for Systems Biotechnology, Technische Universität München, Garching, Germany
- * E-mail:
| |
Collapse
|
15
|
Franco da Cunha F, Andrade-Oliveira V, Candido de Almeida D, Borges da Silva T, Naffah de Souza Breda C, Costa Cruz M, Faquim-Mauro EL, Antonio Cenedeze M, Ioshie Hiyane M, Pacheco-Silva A, Aparecida Cavinato R, Torrecilhas AC, Olsen Saraiva Câmara N. Extracellular Vesicles isolated from Mesenchymal Stromal Cells Modulate CD4 + T Lymphocytes Toward a Regulatory Profile. Cells 2020; 9:cells9041059. [PMID: 32340348 PMCID: PMC7226573 DOI: 10.3390/cells9041059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) can generate immunological tolerance due to their regulatory activity in many immune cells. Extracellular vesicles (EVs) release is a pivotal mechanism by which MSCs exert their actions. In this study, we evaluate whether mesenchymal stromal cell extracellular vesicles (MSC-EVs) can modulate T cell response. MSCs were expanded and EVs were obtained by differential ultracentrifugation of the supernatant. The incorporation of MSC-EVs by T cells was detected by confocal microscopy. Expression of surface markers was detected by flow cytometry or CytoFLEX and cytokines were detected by RT-PCR, FACS and confocal microscopy and a miRNA PCR array was performed. We demonstrated that MSC-EVs were incorporated by lymphocytes in vitro and decreased T cell proliferation and Th1 differentiation. Interestingly, in Th1 polarization, MSC-EVs increased Foxp3 expression and generated a subpopulation of IFN-γ+/Foxp3+T cells with suppressive capacity. A differential expression profile of miRNAs in MSC-EVs-treated Th1 cells was seen, and also a modulation of one of their target genes, TGFbR2. MSC-EVs altered the metabolism of Th1-differentiated T cells, suggesting the involvement of the TGF-β pathway in this metabolic modulation. The addition of MSC-EVs in vivo, in an OVA immunization model, generated cells Foxp3+. Thus, our findings suggest that MSC-EVs are able to specifically modulate activated T cells at an alternative regulatory profile by miRNAs and metabolism shifting.
Collapse
Affiliation(s)
- Flavia Franco da Cunha
- Departamento de Nefrologia, UNIFESP, Rua Pedro de Toledo 669, São Paulo 04039-032, Brazil; (D.C.d.A.); (T.B.d.S.); (M.A.C.); (A.P.-S.); (R.A.C.)
- Correspondence: (F.F.d.C.); (N.O.S.C.)
| | - Vinicius Andrade-Oliveira
- Departamento de Imunologia, USP, Avenida Prof. Lineu Prestes 1730, ICB IV, São Paulo 05508-000, Brazil; (V.A.-O.); (C.N.d.S.B.); (M.C.C.); (M.I.H.)
| | - Danilo Candido de Almeida
- Departamento de Nefrologia, UNIFESP, Rua Pedro de Toledo 669, São Paulo 04039-032, Brazil; (D.C.d.A.); (T.B.d.S.); (M.A.C.); (A.P.-S.); (R.A.C.)
| | - Tamiris Borges da Silva
- Departamento de Nefrologia, UNIFESP, Rua Pedro de Toledo 669, São Paulo 04039-032, Brazil; (D.C.d.A.); (T.B.d.S.); (M.A.C.); (A.P.-S.); (R.A.C.)
| | - Cristiane Naffah de Souza Breda
- Departamento de Imunologia, USP, Avenida Prof. Lineu Prestes 1730, ICB IV, São Paulo 05508-000, Brazil; (V.A.-O.); (C.N.d.S.B.); (M.C.C.); (M.I.H.)
| | - Mario Costa Cruz
- Departamento de Imunologia, USP, Avenida Prof. Lineu Prestes 1730, ICB IV, São Paulo 05508-000, Brazil; (V.A.-O.); (C.N.d.S.B.); (M.C.C.); (M.I.H.)
| | - Eliana L. Faquim-Mauro
- Laboratório de Imunopatologia, Instituto Butantan, Av. Vital Brasil 1500, São Paulo 05503-900, Brazil;
| | - Marcos Antonio Cenedeze
- Departamento de Nefrologia, UNIFESP, Rua Pedro de Toledo 669, São Paulo 04039-032, Brazil; (D.C.d.A.); (T.B.d.S.); (M.A.C.); (A.P.-S.); (R.A.C.)
| | - Meire Ioshie Hiyane
- Departamento de Imunologia, USP, Avenida Prof. Lineu Prestes 1730, ICB IV, São Paulo 05508-000, Brazil; (V.A.-O.); (C.N.d.S.B.); (M.C.C.); (M.I.H.)
| | - Alvaro Pacheco-Silva
- Departamento de Nefrologia, UNIFESP, Rua Pedro de Toledo 669, São Paulo 04039-032, Brazil; (D.C.d.A.); (T.B.d.S.); (M.A.C.); (A.P.-S.); (R.A.C.)
- Hospital Israelita Albert Einstein, Av. Albert Einstein, São Paulo 627–05652-900, Brazil
| | - Regiane Aparecida Cavinato
- Departamento de Nefrologia, UNIFESP, Rua Pedro de Toledo 669, São Paulo 04039-032, Brazil; (D.C.d.A.); (T.B.d.S.); (M.A.C.); (A.P.-S.); (R.A.C.)
| | - Ana Claudia Torrecilhas
- Departamento de Ciências Farmacêuticas, UNIFESP, Rua São Nicolau 210, Diadema 09913-030, São Paulo, Brazil;
| | - Niels Olsen Saraiva Câmara
- Departamento de Nefrologia, UNIFESP, Rua Pedro de Toledo 669, São Paulo 04039-032, Brazil; (D.C.d.A.); (T.B.d.S.); (M.A.C.); (A.P.-S.); (R.A.C.)
- Departamento de Imunologia, USP, Avenida Prof. Lineu Prestes 1730, ICB IV, São Paulo 05508-000, Brazil; (V.A.-O.); (C.N.d.S.B.); (M.C.C.); (M.I.H.)
- Correspondence: (F.F.d.C.); (N.O.S.C.)
| |
Collapse
|
16
|
Song Y, Peng H, Bu D, Ding X, Yang F, Zhu Z, Tian X, Zhang L, Wang X, Tang C, Huang Y, Du J, Jin H. Negative auto-regulation of sulfur dioxide generation in vascular endothelial cells: AAT1 S-sulfenylation. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30306-5. [PMID: 32087961 DOI: 10.1016/j.bbrc.2020.02.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 02/06/2020] [Indexed: 01/10/2023]
Abstract
Recently, endogenous sulfur dioxide (SO2) has been found to exert an important function in the cardiovascular system. However, the regulatory mechanism for SO2 generation has not been entirely clarified. Hence, we aimed to explore the possible auto-regulation of endogenous SO2 generation and its mechanisms in vascular endothelial cells. We showed that SO2 did not affect the protein expression of aspartate aminotransferase 1 (AAT1), a major SO2 synthesis enzyme, but significantly inhibited AAT activity in primary human umbilical vein endothelial cells (HUVECs) and porcine purified AAT1 protein. An AAT1 enzymatic kinetic study showed that SO2 reduced the Vmax (1.89 ± 0.10 vs 2.55 ± 0.12, μmol/mg/min, P < 0.05) and increased the Km (35.97 ± 9.54 vs 19.33 ± 1.76 μmol/L, P < 0.05) values. Furthermore, SO2 induced S-sulfenylation of AAT1 in primary HUVECs and purified AAT1 protein. LC-MS/MS analysis indicated that SO2 sulfenylated AAT1 at Cys192. Mechanistically, thiol reductant DTT treatment or C192S mutation prevented SO2-induced AAT1 sulfenylation and the subsequent inhibition of AAT activity in purified AAT1 protein and primary HUVECs. Our findings reveal, for the first time, a mechanism of auto-regulation of SO2 generation through sulfenylation of AAT1 at Cys192 to suppress AAT activity in vascular endothelial cells. These findings will greatly deepen the understanding of regulatory mechanisms in the cardiovascular homeostasis.
Collapse
Affiliation(s)
- Yunjia Song
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China; Research Unit of Clinical Diagnosis and Treatment of Pediatric Syncope and Cardiovascular Diseases, Chinese Academy of Medical Sciences, China
| | - Hanlin Peng
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Dingfang Bu
- Research Center, Peking University First Hospital, Beijing, 100034, China
| | - Xiang Ding
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fuquan Yang
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhigang Zhu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Xiaoyu Tian
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Lulu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Xiuli Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre, Beijing, 100191, China; Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, 100191, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China; Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, 100191, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
17
|
Saavedra E, González-Chávez Z, Moreno-Sánchez R, Michels PA. Drug Target Selection for Trypanosoma cruzi Metabolism by Metabolic Control Analysis and Kinetic Modeling. Curr Med Chem 2019; 26:6652-6671. [DOI: 10.2174/0929867325666180917104242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 08/17/2018] [Accepted: 08/17/2018] [Indexed: 11/22/2022]
Abstract
In the search for therapeutic targets in the intermediary metabolism of trypanosomatids
the gene essentiality criterion as determined by using knock-out and knock-down genetic
strategies is commonly applied. As most of the evaluated enzymes/transporters have
turned out to be essential for parasite survival, additional criteria and approaches are clearly
required for suitable drug target prioritization. The fundamentals of Metabolic Control
Analysis (MCA; an approach in the study of control and regulation of metabolism) and kinetic
modeling of metabolic pathways (a bottom-up systems biology approach) allow quantification
of the degree of control that each enzyme exerts on the pathway flux (flux control coefficient)
and metabolic intermediate concentrations (concentration control coefficient). MCA
studies have demonstrated that metabolic pathways usually have two or three enzymes with
the highest control of flux; their inhibition has more negative effects on the pathway function
than inhibition of enzymes exerting low flux control. Therefore, the enzymes with the highest
pathway control are the most convenient targets for therapeutic intervention. In this review,
the fundamentals of MCA as well as experimental strategies to determine the flux control coefficients
and metabolic modeling are analyzed. MCA and kinetic modeling have been applied
to trypanothione metabolism in Trypanosoma cruzi and the model predictions subsequently
validated in vivo. The results showed that three out of ten enzyme reactions analyzed
in the T. cruzi anti-oxidant metabolism were the most controlling enzymes. Hence, MCA and
metabolic modeling allow a further step in target prioritization for drug development against
trypanosomatids and other parasites.
Collapse
Affiliation(s)
- Emma Saavedra
- Departamento de Bioquimica, Instituto Nacional de Cardiologia Ignacio Chavez. Mexico City, Mexico
| | - Zabdi González-Chávez
- Departamento de Bioquimica, Instituto Nacional de Cardiologia Ignacio Chavez. Mexico City, Mexico
| | - Rafael Moreno-Sánchez
- Departamento de Bioquimica, Instituto Nacional de Cardiologia Ignacio Chavez. Mexico City, Mexico
| | - Paul A.M. Michels
- Centre for Immunity, Infection and Evolution (CIIE) and Centre for Translational and Chemical Biology (CTCB), School of Biological Sciences, The University of Edinburgh, Edinburgh, Scotland
| |
Collapse
|
18
|
Jones SJ, Taylor AF, Beales PA. Towards feedback-controlled nanomedicines for smart, adaptive delivery. Exp Biol Med (Maywood) 2019; 244:283-293. [PMID: 30205721 PMCID: PMC6435888 DOI: 10.1177/1535370218800456] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
IMPACT STATEMENT The timing and rate of release of pharmaceuticals from advanced drug delivery systems is an important property that has received considerable attention in the scientific literature. Broadly, these mostly fall into two classes: controlled release with a prolonged release rate or triggered release where the drug is rapidly released in response to an environmental stimulus. This review aims to highlight the potential for developing adaptive release systems that more subtlety modulate the drug release profile through continuous communication with its environment facilitated through feedback control. By reviewing the key elements of this approach in one place (fundamental principles of nanomedicine, enzymatic nanoreactors for medical therapies and feedback-controlled chemical systems) and providing additional motivating case studies in the context of chronobiology, we hope to inspire innovative development of novel "chrononanomedicines."
Collapse
Affiliation(s)
- Stephen J. Jones
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Annette F. Taylor
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - Paul A Beales
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
19
|
Ellis GFR, Kopel J. The Dynamical Emergence of Biology From Physics: Branching Causation via Biomolecules. Front Physiol 2019; 9:1966. [PMID: 30740063 PMCID: PMC6355675 DOI: 10.3389/fphys.2018.01966] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/31/2018] [Indexed: 01/30/2023] Open
Abstract
Biology differs fundamentally from the physics that underlies it. This paper proposes that the essential difference is that while physics at its fundamental level is Hamiltonian, in biology, once life has come into existence, causation of a contextual branching nature occurs at every level of the hierarchy of emergence at each time. The key feature allowing this to happen is the way biomolecules such as voltage-gated ion channels can act to enable branching logic to arise from the underlying physics, despite that physics per se being of a deterministic nature. Much randomness occurs at the molecular level, which enables higher level functions to select lower level outcomes according to higher level needs. Intelligent causation occurs when organisms engage in deduction, enabling prediction and planning. This is possible because ion channels enable action potentials to propagate in axons. The further key feature is that such branching biological behavior acts down to cause the underlying physical interactions to also exhibit a contextual branching behavior.
Collapse
Affiliation(s)
- George F. R. Ellis
- Mathematics Department, University of Cape Town, Cape Town, South Africa
| | - Jonathan Kopel
- Texas Tech University Health Sciences Center (TTUHSC), Lubbock, TX, United States
| |
Collapse
|
20
|
Christensen CD, Hofmeyr JHS, Rohwer JM. Delving deeper: Relating the behaviour of a metabolic system to the properties of its components using symbolic metabolic control analysis. PLoS One 2018; 13:e0207983. [PMID: 30485345 PMCID: PMC6261606 DOI: 10.1371/journal.pone.0207983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/11/2018] [Indexed: 11/22/2022] Open
Abstract
High-level behaviour of metabolic systems results from the properties of, and interactions between, numerous molecular components. Reaching a complete understanding of metabolic behaviour based on the system’s components is therefore a difficult task. This problem can be tackled by constructing and subsequently analysing kinetic models of metabolic pathways since such models aim to capture all the relevant properties of the system components and their interactions. Symbolic control analysis is a framework for analysing pathway models in order to reach a mechanistic understanding of their behaviour. By providing algebraic expressions for the sensitivities of system properties, such as metabolic flux or steady-state concentrations, in terms of the properties of individual reactions it allows one to trace the high level behaviour back to these low level components. Here we apply this method to a model of pyruvate branch metabolism in Lactococcus lactis in order to explain a previously observed negative flux response towards an increase in substrate concentration. With this method we are able to show, first, that the sensitivity of flux towards changes in reaction rates (represented by flux control coefficients) is determined by the individual metabolic branches of the pathway, and second, how the sensitivities of individual reaction rates towards their substrates (represented by elasticity coefficients) contribute to this flux control. We also quantify the contributions of enzyme binding and mass-action to enzyme elasticity separately, which allows for an even finer-grained understanding of flux control. These analytical tools allow us to analyse the control properties of a metabolic model and to arrive at a mechanistic understanding of the quantitative contributions of each of the enzymes to this control. Our analysis provides an example of the descriptive power of the general principles of symbolic control analysis.
Collapse
Affiliation(s)
- Carl D. Christensen
- Laboratory for Molecular Systems Biology, Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Jan-Hendrik S. Hofmeyr
- Laboratory for Molecular Systems Biology, Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
- Centre for Complex Systems in Transition, Stellenbosch University, Stellenbosch, South Africa
| | - Johann M. Rohwer
- Laboratory for Molecular Systems Biology, Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
- * E-mail:
| |
Collapse
|
21
|
Optimal control of bacterial growth for the maximization of metabolite production. J Math Biol 2018; 78:985-1032. [PMID: 30334073 DOI: 10.1007/s00285-018-1299-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 09/18/2018] [Indexed: 12/24/2022]
Abstract
Microorganisms have evolved complex strategies for controlling the distribution of available resources over cellular functions. Biotechnology aims at interfering with these strategies, so as to optimize the production of metabolites and other compounds of interest, by (re)engineering the underlying regulatory networks of the cell. The resulting reallocation of resources can be described by simple so-called self-replicator models and the maximization of the synthesis of a product of interest formulated as a dynamic optimal control problem. Motivated by recent experimental work, we are specifically interested in the maximization of metabolite production in cases where growth can be switched off through an external control signal. We study various optimal control problems for the corresponding self-replicator models by means of a combination of analytical and computational techniques. We show that the optimal solutions for biomass maximization and product maximization are very similar in the case of unlimited nutrient supply, but diverge when nutrients are limited. Moreover, external growth control overrides natural feedback growth control and leads to an optimal scheme consisting of a first phase of growth maximization followed by a second phase of product maximization. This two-phase scheme agrees with strategies that have been proposed in metabolic engineering. More generally, our work shows the potential of optimal control theory for better understanding and improving biotechnological production processes.
Collapse
|
22
|
Choi K, Medley JK, König M, Stocking K, Smith L, Gu S, Sauro HM. Tellurium: An extensible python-based modeling environment for systems and synthetic biology. Biosystems 2018; 171:74-79. [PMID: 30053414 PMCID: PMC6108935 DOI: 10.1016/j.biosystems.2018.07.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 10/28/2022]
Abstract
Here we present Tellurium, a Python-based environment for model building, simulation, and analysis that facilitates reproducibility of models in systems and synthetic biology. Tellurium is a modular, cross-platform, and open-source simulation environment composed of multiple libraries, plugins, and specialized modules and methods. Tellurium is a self-contained modeling platform which comes with a fully configured Python distribution. Two interfaces are provided, one based on the Spyder IDE which has an accessible user interface akin to MATLAB and a second based on the Jupyter Notebook, which is a format that contains live code, equations, visualizations, and narrative text. Tellurium uses libRoadRunner as the default SBML simulation engine which supports deterministic simulations, stochastic simulations, and steady-state analyses. Tellurium also includes Antimony, a human-readable model definition language which can be converted to and from SBML. Other standard Python scientific libraries such as NumPy, SciPy, and matplotlib are included by default. Additionally, we include several user-friendly plugins and advanced modules for a wide-variety of applications, ranging from complex algorithms for bifurcation analysis to multidimensional parameter scanning. By combining multiple libraries, plugins, and modules into a single package, Tellurium provides a unified but extensible solution for biological modeling and analysis for both novices and experts. AVAILABILITY tellurium.analogmachine.org.
Collapse
Affiliation(s)
- Kiri Choi
- Department of Bioengineering, University of Washington, William H. Foege Building, Box 355061, Seattle, WA 98195, USA.
| | - J Kyle Medley
- Department of Bioengineering, University of Washington, William H. Foege Building, Box 355061, Seattle, WA 98195, USA.
| | - Matthias König
- Institute for Biology, Institute for Theoretical Biology, Humboldt University, Berlin, Germany.
| | - Kaylene Stocking
- Department of Bioengineering, University of Washington, William H. Foege Building, Box 355061, Seattle, WA 98195, USA.
| | - Lucian Smith
- Department of Bioengineering, University of Washington, William H. Foege Building, Box 355061, Seattle, WA 98195, USA.
| | - Stanley Gu
- Department of Bioengineering, University of Washington, William H. Foege Building, Box 355061, Seattle, WA 98195, USA.
| | - Herbert M Sauro
- Department of Bioengineering, University of Washington, William H. Foege Building, Box 355061, Seattle, WA 98195, USA.
| |
Collapse
|
23
|
Asrani KH, Cheng L, Cheng CJ, Subramanian RR. Arginase I mRNA therapy - a novel approach to rescue arginase 1 enzyme deficiency. RNA Biol 2018; 15:914-922. [PMID: 29923457 DOI: 10.1080/15476286.2018.1475178] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Arginase I (ARG1) deficiency is an autosomal recessive urea cycle disorder, caused by deficiency of the enzyme Arginase I, resulting in accumulation of arginine in blood. Current Standard of Care (SOC) for ARG1 deficiency in patients or those having detrimental mutations of ARG1 gene is diet control. Despite diet and drug therapy with nitrogen scavengers, ~25% of patients suffer from severe mental deficits and loss of ambulation. 75% of patients whose symptoms can be managed through diet therapy continue to suffer neuro-cognitive deficits. In our research, we demonstrate in vitro and in vivo that administration of ARG1 mRNA increased ARG1 protein expression and specific activity in relevant cell types, including ARG1-deficient patient cell lines, as well as in wild type mice for up to 4 days. These studies demonstrate that ARG1 mRNA treatment led to increased functional protein expression of ARG1 and subsequently an increase in urea. Hence, ARG1 mRNA therapy could be a potential treatment option to develop for patients.
Collapse
Affiliation(s)
- Kirtika H Asrani
- a Discovery Research Cambridge , Alexion Pharmaceuticals, Inc ., Cambridge , MA , USA
| | - Lei Cheng
- a Discovery Research Cambridge , Alexion Pharmaceuticals, Inc ., Cambridge , MA , USA
| | - Christopher J Cheng
- b Nucleic Acid Technology , Alexion Pharmaceuticals, Inc ., New Haven , CT , USA
| | - Romesh R Subramanian
- a Discovery Research Cambridge , Alexion Pharmaceuticals, Inc ., Cambridge , MA , USA
| |
Collapse
|
24
|
Cuperlovic-Culf M. Machine Learning Methods for Analysis of Metabolic Data and Metabolic Pathway Modeling. Metabolites 2018; 8:E4. [PMID: 29324649 PMCID: PMC5875994 DOI: 10.3390/metabo8010004] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/15/2023] Open
Abstract
Machine learning uses experimental data to optimize clustering or classification of samples or features, or to develop, augment or verify models that can be used to predict behavior or properties of systems. It is expected that machine learning will help provide actionable knowledge from a variety of big data including metabolomics data, as well as results of metabolism models. A variety of machine learning methods has been applied in bioinformatics and metabolism analyses including self-organizing maps, support vector machines, the kernel machine, Bayesian networks or fuzzy logic. To a lesser extent, machine learning has also been utilized to take advantage of the increasing availability of genomics and metabolomics data for the optimization of metabolic network models and their analysis. In this context, machine learning has aided the development of metabolic networks, the calculation of parameters for stoichiometric and kinetic models, as well as the analysis of major features in the model for the optimal application of bioreactors. Examples of this very interesting, albeit highly complex, application of machine learning for metabolism modeling will be the primary focus of this review presenting several different types of applications for model optimization, parameter determination or system analysis using models, as well as the utilization of several different types of machine learning technologies.
Collapse
Affiliation(s)
- Miroslava Cuperlovic-Culf
- Digital Technologies Research Center, National Research Council of Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada.
| |
Collapse
|