Ubiquitin ligase STUB1 destabilizes IFNγ-receptor complex to suppress tumor IFNγ signaling

The cytokine IFNγ differentially impacts on tumors upon immune checkpoint blockade (ICB). Despite our understanding of downstream signaling events, less is known about regulation of its receptor (IFNγ-R1). With an unbiased genome-wide CRISPR/Cas9 screen for critical regulators of IFNγ-R1 cell surfac...

Full description

Bibliographic Details
Main Authors: Altelaar, M. (Author), Altinok, S. (Author), Apriamashvili, G. (Author), Bleijerveld, O.B (Author), Boshuizen, J. (Author), D’Empaire Altimari, D. (Author), de Bruijn, B. (Author), Krijgsman, O. (Author), Ligtenberg, M.A (Author), Lin, C.-P (Author), Londino, J.D (Author), Oswalt, L.E (Author), Peeper, D.S (Author), Sanchez-Hodge, R. (Author), Schisler, J.C (Author), Traets, J.J.H (Author), van Vliet, A. (Author), Visser, N.L (Author), Vredevoogd, D.W (Author)
Format: Article
Language:English
Published: Nature Research 2022
Subjects:
Online Access:View Fulltext in Publisher
LEADER 02741nam a2200505Ia 4500
001 10-1038-s41467-022-29442-x
008 220425s2022 CNT 000 0 und d
020 |a 20411723 (ISSN) 
245 1 0 |a Ubiquitin ligase STUB1 destabilizes IFNγ-receptor complex to suppress tumor IFNγ signaling 
260 0 |b Nature Research  |c 2022 
856 |z View Fulltext in Publisher  |u https://doi.org/10.1038/s41467-022-29442-x 
520 3 |a The cytokine IFNγ differentially impacts on tumors upon immune checkpoint blockade (ICB). Despite our understanding of downstream signaling events, less is known about regulation of its receptor (IFNγ-R1). With an unbiased genome-wide CRISPR/Cas9 screen for critical regulators of IFNγ-R1 cell surface abundance, we identify STUB1 as an E3 ubiquitin ligase for IFNγ-R1 in complex with its signal-relaying kinase JAK1. STUB1 mediates ubiquitination-dependent proteasomal degradation of IFNγ-R1/JAK1 complex through IFNγ-R1K285 and JAK1K249. Conversely, STUB1 inactivation amplifies IFNγ signaling, sensitizing tumor cells to cytotoxic T cells in vitro. This is corroborated by an anticorrelation between STUB1 expression and IFNγ response in ICB-treated patients. Consistent with the context-dependent effects of IFNγ in vivo, anti-PD-1 response is increased in heterogenous tumors comprising both wildtype and STUB1-deficient cells, but not full STUB1 knockout tumors. These results uncover STUB1 as a critical regulator of IFNγ-R1, and highlight the context-dependency of STUB1-regulated IFNγ signaling for ICB outcome. © 2022, The Author(s). 
650 0 4 |a genetics 
650 0 4 |a human 
650 0 4 |a Humans 
650 0 4 |a metabolism 
650 0 4 |a signal transduction 
650 0 4 |a Signal Transduction 
650 0 4 |a STUB1 protein, human 
650 0 4 |a ubiquitin 
650 0 4 |a Ubiquitin 
650 0 4 |a ubiquitin protein ligase 
650 0 4 |a ubiquitination 
650 0 4 |a Ubiquitination 
650 0 4 |a Ubiquitin-Protein Ligases 
700 1 |a Altelaar, M.  |e author 
700 1 |a Altinok, S.  |e author 
700 1 |a Apriamashvili, G.  |e author 
700 1 |a Bleijerveld, O.B.  |e author 
700 1 |a Boshuizen, J.  |e author 
700 1 |a D’Empaire Altimari, D.  |e author 
700 1 |a de Bruijn, B.  |e author 
700 1 |a Krijgsman, O.  |e author 
700 1 |a Ligtenberg, M.A.  |e author 
700 1 |a Lin, C.-P.  |e author 
700 1 |a Londino, J.D.  |e author 
700 1 |a Oswalt, L.E.  |e author 
700 1 |a Peeper, D.S.  |e author 
700 1 |a Sanchez-Hodge, R.  |e author 
700 1 |a Schisler, J.C.  |e author 
700 1 |a Traets, J.J.H.  |e author 
700 1 |a van Vliet, A.  |e author 
700 1 |a Visser, N.L.  |e author 
700 1 |a Vredevoogd, D.W.  |e author 
773 |t Nature Communications