RIPK3-MLKL-Mediated Neutrophil Death Requires Concurrent Activation of Fibroblast Activation Protein-α.

Wang, Xiaoliang; Gessier, Francois; Perozzo, Remo; Stojkov, Darko; Hosseini, Aref; Amirshahrokhi, Keyvan; Kuchen, Stefan; Yousefi, Shida; Lötscher, Pius; Simon, Hans-Uwe (2020). RIPK3-MLKL-Mediated Neutrophil Death Requires Concurrent Activation of Fibroblast Activation Protein-α. Journal of immunology, 205(6), pp. 1653-1663. American Association of Immunologists 10.4049/jimmunol.2000113

[img] Text
Simon_RIPK3-MLKL-Mediated Neutrophil Death Requires.pdf - Published Version
Restricted to registered users only
Available under License Publisher holds Copyright.

Download (1MB) | Request a copy

Cytokine-primed neutrophils can undergo a nonapoptotic type of cell death using components of the necroptotic pathway, including receptor-interacting protein kinase-3 (RIPK3), mixed lineage kinase-like (MLKL) and NADPH oxidase. In this report, we provide evidence for a potential role of serine proteases in CD44-mediated necroptotic death of GM-CSF-primed human neutrophils. Specifically, we observed that several inhibitors known to block the enzymatic function of fibroblast activation protein-α (FAP-α) were able to block CD44-mediated reactive oxygen species production and cell death, but not FAS receptor-mediated apoptosis. To understand how FAP-α is involved in this nonapoptotic death pathway, we performed immunoblotting experiments in the presence and absence of inhibitors of RIPK3, MLKL, p38 MAPK, PI3K, and FAP-α. The results of these experiments suggested that FAP-α is active in parallel with RIPK3, MLKL, and p38 MAPK activation but proximal to PI3K and NADPH oxidase activation. Interestingly, neutrophils isolated from the joints of patients suffering from rheumatoid arthritis underwent a GM-CSF-independent necroptosis following CD44 ligation; this effect was also blocked by both FAP-α and MLKL inhibitors. Taken together, our evidence shows that the RIPK3-MLKL pathway activates NADPH oxidase but requires, in addition to p38 MAPK and PI3K, a serine protease activity, whereby FAP-α is the most likely candidate. Thus, FAP-α could be a potential drug target in neutrophilic inflammatory responses to avoid exaggerated nonapoptotic neutrophil death, leading to tissue damage.

Item Type:

Journal Article (Original Article)

Division/Institute:

04 Faculty of Medicine > Department of Dermatology, Urology, Rheumatology, Nephrology, Osteoporosis (DURN) > Clinic of Rheumatology and Immunology
04 Faculty of Medicine > Pre-clinic Human Medicine > Institute of Pharmacology
09 Interdisciplinary Units > Microscopy Imaging Center (MIC)

UniBE Contributor:

Wang, Xiaoliang, Stojkov, Darko, Hosseini, Aref, Kuchen, Stefan, Yousefi, Shida, Simon, Hans-Uwe

Subjects:

600 Technology > 610 Medicine & health

ISSN:

0022-1767

Publisher:

American Association of Immunologists

Language:

English

Submitter:

Celine Joray

Date Deposited:

24 Sep 2020 11:16

Last Modified:

05 Dec 2022 15:40

Publisher DOI:

10.4049/jimmunol.2000113

PubMed ID:

32796025

BORIS DOI:

10.7892/boris.146660

URI:

https://boris.unibe.ch/id/eprint/146660

Actions (login required)

Edit item Edit item
Provide Feedback