4.7 Article

Human skin dendritic cell fate is differentially regulated by the monocyte identity factor Kruppel-like factor 4 during steady state and inflammation

Journal

JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY
Volume 139, Issue 6, Pages 1873-+

Publisher

MOSBY-ELSEVIER
DOI: 10.1016/j.jaci.2016.09.018

Keywords

Kruppel-like factor 4; Runt-related transcription factor 3; TGF-beta 1 signaling; Notch; lineage decision; monocyte differentiation

Funding

  1. Austrian Science Fund (FWF) [P19474-B13, P22441-B13, P23228-B19, P22058, P19245, P25720, SFB-2304]
  2. [W1212]
  3. [W1248-B13]
  4. [W1241]
  5. Austrian Science Fund (FWF) [P 25720] Funding Source: researchfish
  6. Austrian Science Fund (FWF) [W1212, W1241, P25720] Funding Source: Austrian Science Fund (FWF)

Ask authors/readers for more resources

Background: Langerhans cell (LC) networks play key roles in immunity and tolerance at body surfaces. LCs are established prenatally and can be replenished from blood monocytes. Unlike skin-resident dermal DCs (dDCs)/interstitial-type DCs and inflammatory dendritic epidermal cells appearing in dermatitis/eczema lesions, LCs lack key monocyte-affiliated markers. Inversely, LCs express various epithelial genes critical for their long-term peripheral tissue residency. Objective: Dendritic cells (DCs) are functionally involved in inflammatory diseases; however, the mechanisms remained poorly understood. Methods: In vitro differentiation models of human DCs, gene profiling, gene transduction, and immunohistology were used to identify molecules involved in DC subset specification. Results: Here we identified the monocyte/macrophage lineage identity transcription factor Kruppel-like factor 4 (KLF4) to be inhibited during LC differentiation from human blood monocytes. Conversely, KLF4 is maintained or induced during dermal DC and monocyte-derived dendritic cell/inflammatory dendritic epidermal cell differentiation. We showed that in monocytic cells KLF4 has to be repressed to allow their differentiation into LCs. Moreover, respective KLF4 levels in DC subsets positively correlate with proinflammatory characteristics. We identified epithelial Notch signaling to repress KLF4 in monocytes undergoing LC commitment. Loss of KLF4 in monocytes transcriptionally derepresses Runt-related transcription factor 3 in response to TGF-beta 1, thereby allowing LC differentiation marked by a low cytokine expression profile. Conclusion: Monocyte differentiation into LCs depends on activation of Notch signaling and the concomitant loss of KLF4.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available