2022
DOI: 10.1038/s41929-022-00798-z
|View full text |Cite
|
Sign up to set email alerts
|

Deep learning-based kcat prediction enables improved enzyme-constrained model reconstruction

Abstract: Enzyme turnover numbers (kcat) are key to understanding cellular metabolism, proteome allocation and physiological diversity, but experimentally measured kcat data are sparse and noisy. Here we provide a deep learning approach (DLKcat) for high-throughput kcat prediction for metabolic enzymes from any organism merely from substrate structures and protein sequences. DLKcat can capture kcat changes for mutated enzymes and identify amino acid residues with a strong impact on kcat values. We applied this approach … Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
3
1
1

Citation Types

0
206
0

Year Published

2022
2022
2024
2024

Publication Types

Select...
6
3

Relationship

0
9

Authors

Journals

citations
Cited by 171 publications
(244 citation statements)
references
References 70 publications
0
206
0
Order By: Relevance
“…Unfortunately, there is little data on both for C. glutamicum. There are three ways to improve the coverage of enzyme kinetic parameters in the model: 1) directly populate unknown reactions with mean or median values of enzyme kinetic parameters from other reactions [16][17][18]; 2) expand the EC number annotation information of model reactions using EC number prediction tools [54,55]; and 3) directly predict reactions with unknown parameters based on existing enzyme kinetic parameters via machine learning or deep learning approaches [25,56]. In addition, the kinetic data are mainly sourced from the BRENDA and SABIO-RK databases, which mostly collect in vitro measurements that differ somewhat from the in vivo data.…”
Section: Discussionmentioning
confidence: 99%
See 1 more Smart Citation
“…Unfortunately, there is little data on both for C. glutamicum. There are three ways to improve the coverage of enzyme kinetic parameters in the model: 1) directly populate unknown reactions with mean or median values of enzyme kinetic parameters from other reactions [16][17][18]; 2) expand the EC number annotation information of model reactions using EC number prediction tools [54,55]; and 3) directly predict reactions with unknown parameters based on existing enzyme kinetic parameters via machine learning or deep learning approaches [25,56]. In addition, the kinetic data are mainly sourced from the BRENDA and SABIO-RK databases, which mostly collect in vitro measurements that differ somewhat from the in vivo data.…”
Section: Discussionmentioning
confidence: 99%
“…In the enzyme-constrained models, kcat and molecular weight (MW) of an enzyme set constraints on the fluxes of the reactions catalyzed by that enzyme. Previous studies have made efforts to automatically acquire kcat values from databases and fill missing values using methods like machine learning [25]. In contrast, few studies paid attention to molecular weight.…”
Section: Introductionmentioning
confidence: 99%
“…Moreover, the gradient descent algorithm changes the turnover numbers of all the enzymes simultaneously, thus the imputed enzyme concentrations effectively get changed relative to the initial estimates, attenuating dramatic changes in turnover number estimates of single reactions. Thus, given the increasing number of machine learning generated turnover number estimates [14,48], this algorithm is well suited as a refinement step to increase their accuracy.…”
Section: Estimating Enzyme Turnover Numbersmentioning
confidence: 99%
“…In the enzyme-constrained models, k cat and molecular weight (MW) of an enzyme set constraints on the fluxes of the reactions catalyzed by that enzyme. Previous studies have made efforts to automatically acquire k cat values from databases and fill missing values using methods like machine learning [ 25 ]. In contrast, few studies have paid attention to molecular weight.…”
Section: Introductionmentioning
confidence: 99%