2023
DOI: 10.1038/s41587-023-01728-5
|View full text |Cite
|
Sign up to set email alerts
|

A relay velocity model infers cell-dependent RNA velocity

Abstract: RNA velocity provides an approach for inferring cellular state transitions from single-cell RNA sequencing (scRNA-seq) data. Conventional RNA velocity models infer universal kinetics from all cells in an scRNA-seq experiment, resulting in unpredictable performance in experiments with multi-stage and/or multi-lineage transition of cell states where the assumption of the same kinetic rates for all cells no longer holds. Here we present cellDancer, a scalable deep neural network that locally infers velocity for e… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

0
14
0

Year Published

2023
2023
2024
2024

Publication Types

Select...
6
3

Relationship

1
8

Authors

Journals

citations
Cited by 35 publications
(21 citation statements)
references
References 53 publications
(93 reference statements)
0
14
0
Order By: Relevance
“…To further understand the phenotypic transition of SMCs, we performed RNA velocity analysis using cellDancer 27 (Figure 2C and 2D). The RNA velocity estimated the 2 directions of cell transition: from contractile toward migration/inflammatory phenotype and from contractile/ inflammatory toward other cell types (mostly macrophages; Figure 2C).…”
Section: Progressive Reduction Of Contractile Genes and Induction Of ...mentioning
confidence: 99%
“…To further understand the phenotypic transition of SMCs, we performed RNA velocity analysis using cellDancer 27 (Figure 2C and 2D). The RNA velocity estimated the 2 directions of cell transition: from contractile toward migration/inflammatory phenotype and from contractile/ inflammatory toward other cell types (mostly macrophages; Figure 2C).…”
Section: Progressive Reduction Of Contractile Genes and Induction Of ...mentioning
confidence: 99%
“…For further research in the future, we need to combine proteomics to evaluate protein velocity and even combine epigenomics with chromatin velocity to understand the regulation mechanism of the cell cycle more comprehensively. More broadly, when coupled with remarkable advances in singlecell approaches, including multi-velocity, [89] construction of vector fields, [36] as well as spatial multi-omics, [90] we will enable a deeper, quantitative understanding of the spatiotemporally un-derlying mechanism of the cell cycle and other biological systems via nonequilibrium dynamics and thermodynamics to help prevent various diseases, including cancer and neurodegeneration, and open up new possibilities for precision medicine.…”
Section: Discussionmentioning
confidence: 99%
“…For further research in the future, we need to combine proteomics to evaluate protein velocity and even combine epigenomics with chromatin velocity to understand the regulation mechanism of the cell cycle more comprehensively. More broadly, when coupled with remarkable advances in single-cell approaches, including multi-velocity (83), construction of vector fields (35), as well as spatial multi-omics (84), we will enable a deeper, quantitative understanding of the spatiotemporally underlying mechanism of the cell cycle and other biological systems via non-equilibrium dynamics and thermodynamics to help prevent various diseases, including cancer and neurodegeneration, and open up new possibilities for precision medicine.…”
Section: Discussionmentioning
confidence: 99%