Xin Zhao's research while affiliated with Chinese Academy of Sciences and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (9)


Development of metabolic models with multiple constraints: a review
  • Literature Review
  • Full-text available

February 2022

·

79 Reads

·

1 Citation

Chinese Journal of Biotechnology

·

Peiji Zhang

·

·

[...]

·

Constraint-based genome-scale metabolic network models (genome-scale metabolic models, GEMs) have been widely used to predict metabolic phenotypes. In addition to stoichiometric constraints, other constraints such as enzyme availability and thermodynamic feasibility may also limit the cellular phenotype solution space. Recently, extended GEM models considering either enzymatic or thermodynamic constraints have been developed to improve model prediction accuracy. This review summarizes the recent progresses on metabolic models with multiple constraints (MCGEMs). We presented the construction methods and various applications of MCGEMs including the simulation of gene knockout, prediction of biologically feasible pathways and identification of bottleneck steps. By integrating multiple constraints in a consistent modeling framework, MCGEMs can predict the metabolic bottlenecks and key controlling and modification targets for pathway optimization more precisely, and thus may provide more reliable design results to guide metabolic engineering of industrially important microorganisms.

Download
Share

ECMpy, a Simplified Workflow for Constructing Enzymatic Constrained Metabolic Network Model

January 2022

·

50 Reads

·

24 Citations

Biomolecules

Genome-scale metabolic models (GEMs) have been widely used for the phenotypic prediction of microorganisms. However, the lack of other constraints in the stoichiometric model often leads to a large metabolic solution space being inaccessible. Inspired by previous studies that take an allocation of macromolecule resources into account, we developed a simplified Python-based workflow for constructing enzymatic constrained metabolic network model (ECMpy) and constructed an enzyme-constrained model for Escherichia coli (eciML1515) by directly adding a total enzyme amount constraint in the latest version of GEM for E. coli (iML1515), considering the protein subunit composition in the reaction, and automated calibration of enzyme kinetic parameters. Using eciML1515, we predicted the overflow metabolism of E. coli and revealed that redox balance was the key reason for the difference between E. coli and Saccharomyces cerevisiae in overflow metabolism. The growth rate predictions on 24 single-carbon sources were improved significantly when compared with other enzyme-constrained models of E. coli. Finally, we revealed the tradeoff between enzyme usage efficiency and biomass yield by exploring the metabolic behaviours under different substrate consumption rates. Enzyme-constrained models can improve simulation accuracy and thus can predict cellular phenotypes under various genetic perturbations more precisely, providing reliable guidance for metabolic engineering.


Figure 3. Comparison of simulation results of the enzyme-constrained model eciML1515 and the stoichiometric model iML1515. Simulation of overflow metabolism at different growth rates using eciML1515 (a) and iML1515 (b). c. Simulated different overflow metabolism of E. coli and S. cerevisiae. d. The different overflow metabolic pathways of E. coli and S. cerevisiae.
Figure 5. (a) Simulated growth rates at different glucose uptake rates. (b) The trade-off between biomass yield and enzyme efficiency.
Comparison of the construction methods of enzyme-constrained model
ECMpy, a Simplified Workflow for Constructing Enzymatic Constrained Metabolic Network Model

December 2021

·

70 Reads

Genome-scale metabolic models (GEMs) have been widely used for phenotypic prediction of microorganisms. However, the lack of other constraints in the stoichiometric model often leads to a large metabolic solution space inaccessible. Inspired by previous studies that take allocation of macromolecule resources into account, we developed a simplified Python-based workflow for constructing enzymatic constrained metabolic network model (ECMpy) and constructed an enzyme-constrained model for Escherichia coli (eciML1515) by directly adding a total enzyme amount constraint in the latest version of GEM for E. coli (iML1515), considering the protein subunit composition in the reaction, and automated calibration of enzyme kinetic parameters. Using eciML1515, we predicted the overflow metabolism of E. coli and revealed that redox balance was the key reason for the difference between E. coli and Saccharomyces cerevisiae in overflow metabolism. The growth rate predictions on 24 single-carbon sources were improved significantly when compared with other enzyme-constrained models of E. coli. Finally, we revealed the tradeoff between enzyme usage efficiency and biomass yield by exploring the metabolic behaviors under different substrate consumption rates. Enzyme-constrained models can improve simulation accuracy and thus can predict cellular phenotypes under various genetic perturbations more precisely, providing reliable guidance for metabolic engineering.




Fig. 3. The simulation results of 22 product synthesis rates based on various models. (A) iML1515; (B) EcoTCM; (C) EcoECM; (D) EcoETM. The order of names in the legend is the same as the order of the final values of the production curves (from top to bottom). The molar amounts of products were normalized based on glucose (6 C-atoms). Original data for the figures can be seen in Supplementary file 2.
Fig. 4. The predicted synthesis rates for various products by various models. (A) (S)-Dihydroorotate (Dhor_S); (B) N-Carb-L-aspartate (Cbasp); (C) Carbamoyl phosphate (Cbp); (D) Orotidine-5-P (Orot5p); (E) Orotate (Orot); (F) l-Arginine (L-Arg). The molar amounts of products were normalized based on glucose (6 C-atoms).
Fig. 5. The MDF of product synthesis pathways under different constraints. (A) Carbamoyl phosphate; (B) L-Arginine; (C) Oxaloacetate; (D) Citrate; (E) Isocitrate; (F) Glyoxylate.
Fig. 6. Prediction of the L-arginine synthesis pathway by iML1515 (A) and EcoETM (B). Shown are: the structural change of the pathway (red arrow); the reaction with the highest enzyme cost (red thick arrow); the thermodynamic bottleneck reaction (blue arrow); the limiting metabolite (yellow background); and the simplified pathways of EMP and PP (navy-blue background). The unit of the flux value is mmol/gDW/h (blue, on the top); the unit of the enzyme cost is mg/gDW (purple, in the middle); and the unit of the maximum thermodynamic driving force is in kJ/mol (orange, at the bottom). (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)
Comparison of parameters for the two Cbp synthesis reactions.
Integrating thermodynamic and enzymatic constraints into genome-scale metabolic models

June 2021

·

144 Reads

·

31 Citations

Metabolic Engineering

Stoichiometric genome-scale metabolic network models (GEMs) have been widely used to predict metabolic phenotypes. In addition to stoichiometric ratios, other constraints such as enzyme availability and thermodynamic feasibility can also limit the phenotype solution space. Extended GEM models considering either enzymatic or thermodynamic constraints have been shown to improve prediction accuracy. In this paper, we propose a novel method that integrates both enzymatic and thermodynamic constraints in a single Pyomo modeling framework (ETGEMs). We applied this method to construct the EcoETM (E. coli metabolic model with enzymatic and thermodynamic constraints). Using this model, we calculated the optimal pathways for cellular growth and the production of 22 metabolites. When comparing the results with those of iML1515 and models with one of the two constraints, we observed that many thermodynamically unfavorable and/or high enzyme cost pathways were excluded from EcoETM. For example, the synthesis pathway of carbamoyl-phosphate (Cbp) from iML1515 is both thermodynamically unfavorable and enzymatically costly. After introducing the new constraints, the production pathways and yields of several Cbp-derived products (e.g. L-arginine, orotate) calculated using EcoETM were more realistic. The results of this study demonstrate the great application potential of metabolic models with multiple constraints for pathway analysis and phenotype prediction.


Fig. 4. The predicted synthesis rates for various products by various models. (A)
Integrating thermodynamic and enzymatic constraints into genome-scale metabolic models

December 2020

·

75 Reads

·

2 Citations

Stoichiometric genome-scale metabolic network models (GEMs) have been widely used to predict metabolic phenotypes. In addition to stoichiometric ratios, other constraints such as enzyme availability and thermodynamic feasibility can also limit the phenotype solution space. Extended GEM models considering either enzymatic or thermodynamic constraints have been shown to improve prediction accuracy. In this paper, we propose a novel method that integrates both enzymatic and thermodynamic constraints in a single Pyomo modeling framework (ETGEMs). We applied this method to construct the EcoETM, the E. coli metabolic model iML1515 with enzymatic and thermodynamic constraints. Using this model, we calculated the optimal pathways for cellular growth and the production of 22 metabolites. When comparing the results with those of iML1515 and models with one of the two constraints, we observed that many thermodynamically unfavorable and/or high enzyme cost pathways were excluded from EcoETM. For example, the synthesis pathway of carbamoyl-phosphate (Cbp) from iML1515 is both thermodynamically unfavorable and enzymatically costly. After introducing the new constraints, the production pathways and yields of several Cbp-derived products (e.g. L-arginine, orotate) calculated using EcoETM were more realistic. The results of this study demonstrate the great application potential of metabolic models with multiple constraints for pathway analysis and phenotype predication.


Correspondence between abbreviations and names
Progress and application of metabolic network model based on enzyme constraints

October 2019

·

90 Reads

·

1 Citation

Chinese Journal of Biotechnology

Genome-scale metabolic network models have been successfully applied to guide metabolic engineering. However, the conventional flux balance analysis only considers stoichiometry and reaction direction constraints, and the simulation results cannot accurately describe certain phenomena such as overflow metabolism and diauxie growth on two substrates. Recently, researchers proposed new constraint-based methods to simulate the cellular behavior under different conditions more precisely by introducing new constraints such as limited enzyme content and thermodynamics feasibility. Here we review several enzyme-constrained models, giving a comprehensive introduction on the biological basis and mathematical representation for the enzyme constraint, the optimization function, the impact on the calculated flux distribution and their application in identification of metabolic engineering targets. The main problems in these existing methods and the perspectives on this emerging research field are also discussed. By introducing new constraints, metabolic network models can simulate and predict cellular behavior under various environmental and genetic perturbations more accurately, and thus can provide more reliable guidance to strain engineering.


Systematic design and in vitro validation of novel one-carbon assimilation pathways

September 2019

·

1,206 Reads

·

64 Citations

Metabolic Engineering

The utilization of one-carbon (C1) assimilation pathways to produce chemicals and fuels from low-cost C1 compounds could greatly reduce the substrate-related production costs, and would also alleviate the pressure of the resource supply for bio-manufacturing. However, the natural C1 assimilation pathways normally involve ATP consumption or the loss of carbon resources as CO2, resulting in low product yields, making the design of novel pathways highly pertinent. Here we present several new ATP-independent and carbon-conserving C1 assimilation cycles with 100% theoretical carbon yield, which were discovered by computational analysis of metabolic reaction set with 6578 natural reactions from MetaCyc database and 73 computationally predicted aldolase reactions from ATLAS database. Then, kinetic evaluation of these cycles was conducted and the cycles without kinetic traps were chosen for further experimental verification. Finally, we used the two engineered enzymes Gals and TalBF178Y for the artificial reactions to construct a novel C1 assimilation pathway in vitro and optimized the pathway to achieve 88% carbon yield. These results demonstrate the usefulness of computational design in finding novel metabolic pathways for the efficient utilization of C1 compounds and shedding light on other promising pathways.

Citations (4)


... Notably, this integration is evident in frameworks such as GECKO (If no version is specified, the default is GECKO 3.0) [3] and AutoPACMEN [4]. These sophisticated models have significantly expanded the scope of classical flux balance analysis (FBA), providing insights into overflow metabolism and cellular growth across diverse environments for a range of organisms, including Escherichia coli [5,6], Saccharomyces cerevisiae [7], Yarrowia lipolytica [8], Aspergillus niger [9], Corynebacterium ...

Reference:

ECMpy 2.0: A Python Package for Automated Construction and Analysis of Enzyme-Constrained Models
ECMpy, a Simplified Workflow for Constructing Enzymatic Constrained Metabolic Network Model

Biomolecules

... However, given the inherent complexity of biological systems, relying solely on enzymatic constraints proves insufficient for a comprehensive description. Hence, there is a necessity to incorporate additional biological data into novel composite constraints, such as thermodynamics [29] and regulatory networks [30], or to construct a whole-cell GEM [31]. Furthermore, in terms of visualization of results, there is a lack of pathway visualization functionality similar to what CAVE [32] provides. ...

Integrating thermodynamic and enzymatic constraints into genome-scale metabolic models

Metabolic Engineering

... The iML1515 model [Mon+17] is a newer model of E. coli K-12 MG1655 released in 2017, which contains 189 new genes and 196 new reactions compared to iJO1366. It has been gradually accepted as a good replacement for iJO1366 in many researches, including [Yan+20] and [Kar+18]. In our project, we will use iML1515 as the GEM model because the metabolic simulation based on it is supposed to be more similar to the metabolism in the real world. ...

Integrating thermodynamic and enzymatic constraints into genome-scale metabolic models

... methanol assimilation research. [11][12][13] Meanwhile, researchers have endeavored to create novel CO 2 assimilation pathways, aiming to simulate and surpass the natural Calvin-Benson cycle. [14][15][16][17] In recent years, the efficiency of CO 2 electrocatalytic synthesis of one-carbon organic compounds has seen notable improvement. ...

Systematic design and in vitro validation of novel one-carbon assimilation pathways

Metabolic Engineering