Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Standard

Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells. / Liu, Yawei; Marin, Andrea; Ejlerskov, Patrick; Rasmussen, Louise Munk; Prinz, Marco; Issazadeh-Navikas, Shohreh.

I: Nature Communications, Bind 8, 14709, 24.04.2017.

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Harvard

Liu, Y, Marin, A, Ejlerskov, P, Rasmussen, LM, Prinz, M & Issazadeh-Navikas, S 2017, 'Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells', Nature Communications, bind 8, 14709. https://doi.org/10.1038/ncomms14709

APA

Liu, Y., Marin, A., Ejlerskov, P., Rasmussen, L. M., Prinz, M., & Issazadeh-Navikas, S. (2017). Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells. Nature Communications, 8, [14709]. https://doi.org/10.1038/ncomms14709

Vancouver

Liu Y, Marin A, Ejlerskov P, Rasmussen LM, Prinz M, Issazadeh-Navikas S. Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells. Nature Communications. 2017 apr. 24;8. 14709. https://doi.org/10.1038/ncomms14709

Author

Liu, Yawei ; Marin, Andrea ; Ejlerskov, Patrick ; Rasmussen, Louise Munk ; Prinz, Marco ; Issazadeh-Navikas, Shohreh. / Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells. I: Nature Communications. 2017 ; Bind 8.

Bibtex

@article{e8429e33d4a44274849f8b98a13c777c,
title = "Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells",
abstract = "Neurons reprogramme encephalitogenic T cells (Tenc) to regulatory T cells (Tregs), either FoxP3(+)Tregs or FoxA1(+)Tregs. We reported previously that neuronal ability to generate FoxA1(+)Tregs was central to preventing neuroinflammation in experimental autoimmune encephalomyelitis (EAE). Mice lacking interferon (IFN)-β were defective in generating FoxA1(+)Tregs in the brain. Here we show that lack of neuronal IFNβ signalling is associated with the absence of programme death ligand-1 (PDL1), which prevents their ability to reprogramme Tenc cells to FoxA1(+)Tregs. Passive transfer-EAE via IFNβ-competent Tenc cells to mice lacking IFNβ and active induced-EAE in mice lacking its receptor, IFNAR, in the brain (Nes(Cre):Ifnar(fl/fl)) result in defective FoxA1(+)Tregs generation and aggravated neuroinflammation. IFNβ activates neuronal PI3K/Akt signalling and Akt binds to transcription factor FoxA1 that translocates to the nucleus and induces PDL1. Conversely, inhibition of PI3K/Akt, FoxA1 and PDL1 blocked neuronal ability to generate FoxA1(+)Tregs. We characterize molecular factors central for neuronal ability to reprogramme pathogenic T cells to FoxA1(+)Tregs preventing neuroinflammation.",
author = "Yawei Liu and Andrea Marin and Patrick Ejlerskov and Rasmussen, {Louise Munk} and Marco Prinz and Shohreh Issazadeh-Navikas",
year = "2017",
month = apr,
day = "24",
doi = "10.1038/ncomms14709",
language = "English",
volume = "8",
journal = "Nature Communications",
issn = "2041-1723",
publisher = "nature publishing group",

}

RIS

TY - JOUR

T1 - Neuronal IFN-beta-induced PI3K/Akt-FoxA1 signalling is essential for generation of FoxA1(+)Treg cells

AU - Liu, Yawei

AU - Marin, Andrea

AU - Ejlerskov, Patrick

AU - Rasmussen, Louise Munk

AU - Prinz, Marco

AU - Issazadeh-Navikas, Shohreh

PY - 2017/4/24

Y1 - 2017/4/24

N2 - Neurons reprogramme encephalitogenic T cells (Tenc) to regulatory T cells (Tregs), either FoxP3(+)Tregs or FoxA1(+)Tregs. We reported previously that neuronal ability to generate FoxA1(+)Tregs was central to preventing neuroinflammation in experimental autoimmune encephalomyelitis (EAE). Mice lacking interferon (IFN)-β were defective in generating FoxA1(+)Tregs in the brain. Here we show that lack of neuronal IFNβ signalling is associated with the absence of programme death ligand-1 (PDL1), which prevents their ability to reprogramme Tenc cells to FoxA1(+)Tregs. Passive transfer-EAE via IFNβ-competent Tenc cells to mice lacking IFNβ and active induced-EAE in mice lacking its receptor, IFNAR, in the brain (Nes(Cre):Ifnar(fl/fl)) result in defective FoxA1(+)Tregs generation and aggravated neuroinflammation. IFNβ activates neuronal PI3K/Akt signalling and Akt binds to transcription factor FoxA1 that translocates to the nucleus and induces PDL1. Conversely, inhibition of PI3K/Akt, FoxA1 and PDL1 blocked neuronal ability to generate FoxA1(+)Tregs. We characterize molecular factors central for neuronal ability to reprogramme pathogenic T cells to FoxA1(+)Tregs preventing neuroinflammation.

AB - Neurons reprogramme encephalitogenic T cells (Tenc) to regulatory T cells (Tregs), either FoxP3(+)Tregs or FoxA1(+)Tregs. We reported previously that neuronal ability to generate FoxA1(+)Tregs was central to preventing neuroinflammation in experimental autoimmune encephalomyelitis (EAE). Mice lacking interferon (IFN)-β were defective in generating FoxA1(+)Tregs in the brain. Here we show that lack of neuronal IFNβ signalling is associated with the absence of programme death ligand-1 (PDL1), which prevents their ability to reprogramme Tenc cells to FoxA1(+)Tregs. Passive transfer-EAE via IFNβ-competent Tenc cells to mice lacking IFNβ and active induced-EAE in mice lacking its receptor, IFNAR, in the brain (Nes(Cre):Ifnar(fl/fl)) result in defective FoxA1(+)Tregs generation and aggravated neuroinflammation. IFNβ activates neuronal PI3K/Akt signalling and Akt binds to transcription factor FoxA1 that translocates to the nucleus and induces PDL1. Conversely, inhibition of PI3K/Akt, FoxA1 and PDL1 blocked neuronal ability to generate FoxA1(+)Tregs. We characterize molecular factors central for neuronal ability to reprogramme pathogenic T cells to FoxA1(+)Tregs preventing neuroinflammation.

U2 - 10.1038/ncomms14709

DO - 10.1038/ncomms14709

M3 - Journal article

C2 - 28436428

VL - 8

JO - Nature Communications

JF - Nature Communications

SN - 2041-1723

M1 - 14709

ER -

ID: 178255917