2022
DOI: 10.1101/2022.08.11.503623
|View full text |Cite
Preprint
|
Sign up to set email alerts
|

A lncRNA identifiesIRF8enhancer element in negative feedback control of dendritic cell differentiation

Abstract: Transcription factors play a determining role in lineage commitment and cell differentiation. Interferon regulatory factor 8 (IRF8), a hematopoietic transcription factor, is prominently upregulated in dendritic cells (DC) by autoactivation and controls DC differentiation and function. However, it is unclear how IRF8 autoactivation is controlled and eventually limited. Here we identified a novel long non-coding RNA transcribed from the +32 kb enhancer downstream of IRF8 transcription start site and expressed sp… Show more

Help me understand this report
View published versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1

Citation Types

0
2
0

Year Published

2023
2023
2023
2023

Publication Types

Select...
2
1

Relationship

3
0

Authors

Journals

citations
Cited by 3 publications
(2 citation statements)
references
References 81 publications
0
2
0
Order By: Relevance
“…In our recent study, we discovered that the long noncoding RNA lncIRF8 plays a critical role in DC differentiation. This finding was made possible by utilizing DC generated from HoxB8 cells that were genetically modified employing CRISPR/Cas9 technology to knock out specific IRF8 enhancer elements or silence the lnIRF8 promoter by CRISPR/dCas9 [43,44]. In addition to differentiation, also DC migratory mechanisms were studied employing CRISPR editing in HoxB8 cells, where HoxB8-DC CCR7 −/− showed a failure in CCL19-dependent migration [26].…”
Section: Discussionmentioning
confidence: 99%
“…In our recent study, we discovered that the long noncoding RNA lncIRF8 plays a critical role in DC differentiation. This finding was made possible by utilizing DC generated from HoxB8 cells that were genetically modified employing CRISPR/Cas9 technology to knock out specific IRF8 enhancer elements or silence the lnIRF8 promoter by CRISPR/dCas9 [43,44]. In addition to differentiation, also DC migratory mechanisms were studied employing CRISPR editing in HoxB8 cells, where HoxB8-DC CCR7 −/− showed a failure in CCL19-dependent migration [26].…”
Section: Discussionmentioning
confidence: 99%
“…3b). We collected ChIP-seq data of the transcription factors PU.1 and IRF8, and five histone modifications (i.e., H3K4me1, H3K4me3, H3K9me3, H3K27me3, and H3K27ac) for each of the cell types [4, 16, 23, 24] (Supplementary Table 1). PU.1 is one of the master regulators of hematopoiesis and is expressed by all hematopoietic cells [25] and IRF8 is believed to co-bind with PU.1 to control the differentiation of DC progenitors (DCP) towards specific DC sub-types [4, 26].…”
Section: Methodsmentioning
confidence: 99%