2021
DOI: 10.1016/j.gpb.2020.11.008
|View full text |Cite
|
Sign up to set email alerts
|

scGET: Predicting Cell Fate Transition During Early Embryonic Development by Single-Cell Graph Entropy

Abstract: During early embryonic development, cell fate commitment represents a critical transition or “tipping point” of embryonic differentiation, at which there is a drastic and qualitative shift of the cell populations. In this study, we presented a computational approach, scGET, to explore the gene–gene associations based on single-cell RNA sequencing (scRNA-seq) data for critical transition prediction. Specifically, by transforming the gene expression data to the local network entropy, the single-cell graph entrop… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1

Citation Types

0
6
0

Year Published

2021
2021
2023
2023

Publication Types

Select...
6

Relationship

1
5

Authors

Journals

citations
Cited by 21 publications
(10 citation statements)
references
References 55 publications
0
6
0
Order By: Relevance
“… 42 Importantly, DICE displayed a clear transition-like behavior, unlike the individual TF variances which increased but in a mostly asynchronous manner ( Figure 6 E). We also compared DICE to BioTIP, 19 a tool designed to detect cell-fate transitions using the concept of a dynamical network biomarker (DNB), 17 , 18 which also relies on covariation patterns, albeit not just of TFs but of specific subsets of all genes. In line with this, BioTIP’s criticality index also displayed a clear transition, although at an earlier timepoint compared to DICE, whilst also displaying larger fluctuations during the timecourse ( Figures S12 A and S12B).…”
Section: Resultsmentioning
confidence: 99%
See 2 more Smart Citations
“… 42 Importantly, DICE displayed a clear transition-like behavior, unlike the individual TF variances which increased but in a mostly asynchronous manner ( Figure 6 E). We also compared DICE to BioTIP, 19 a tool designed to detect cell-fate transitions using the concept of a dynamical network biomarker (DNB), 17 , 18 which also relies on covariation patterns, albeit not just of TFs but of specific subsets of all genes. In line with this, BioTIP’s criticality index also displayed a clear transition, although at an earlier timepoint compared to DICE, whilst also displaying larger fluctuations during the timecourse ( Figures S12 A and S12B).…”
Section: Resultsmentioning
confidence: 99%
“…It will be interesting if future work were to perform a more comprehensive comparison to include other tipping point algorithms such as scGET. 18 …”
Section: Discussionmentioning
confidence: 99%
See 1 more Smart Citation
“…The recently proposed dynamical network biomarker (DNB) theory shows a possible way of detecting the criticality of complex disease by regarding the progression of a disease as a high-dimensional nonlinear dynamic system and the critical transition as the state shift at the bifurcation point [ 1 , 7 , 8 ]. The DNB method and its modified versions have been applied in a variety of biomedical fields to successfully detect the pre-disease state of metabolic syndromes [ 9 , 10 ], identify immune checkpoint blockades [ 11 ] and assess cell fate commitment [ 12 ]. However, the DNB theory is not suitable for the analysis of datasets with small sample sizes since it requires multiple samples at each time point to evaluate its three statistical conditions, which restricts its application for biological and clinical data.…”
Section: Introductionmentioning
confidence: 99%
“…Meanwhile, a webserver, GranatumX was also presented in this special issue, which enables biologists to access the latest single-cell bioinformatics methods in a web-based graphical environment [22] . On the other hand, as a method-oriented work, the computational model proposed by Zhong et al [23] can explore the gene–gene associations based on scRNA-seq data for critical transition prediction. Li and Li proposed the scLink method, which used statistical network modeling to understand the co-expression relationships among genes and construct sparse gene co-expression networks from single-cell gene expression data [24] .…”
mentioning
confidence: 99%