NADPH oxidase controls neutrophilic response to sterile inflammation in mice by regulating the IL-1α/G-CSF axis.

TitleNADPH oxidase controls neutrophilic response to sterile inflammation in mice by regulating the IL-1α/G-CSF axis.
Publication TypeJournal Article
Year of Publication2015
AuthorsBagaitkar J, Pech NK, Ivanov S, Austin A, Zeng MYue, Pallat S, Huang G, Randolph GJ, Dinauer MC
JournalBlood
Volume126
Issue25
Pagination2724-33
Date Published2015 Dec 17
ISSN1528-0020
KeywordsAnimals, Disease Models, Animal, Flow Cytometry, Granulocyte Colony-Stimulating Factor, Granulomatous Disease, Chronic, Inflammation, Interleukin-1alpha, Mice, Mice, Inbred C57BL, Mice, Knockout, NADPH Oxidases, Neutrophils, Signal Transduction
Abstract

The leukocyte nicotinamide adenine dinucleotide phosphate (NADPH) oxidase generates reactive oxygen species essential in microbial killing and regulation of inflammation. Inactivating mutations in this enzyme lead to chronic granulomatous disease (CGD), associated with increased susceptibility to both pyogenic infections and to inflammatory disorders. The role of the NADPH oxidase in regulating inflammation driven by nonmicrobial stimuli is poorly understood. Here, we show that NADPH oxidase deficiency enhances the early local release of interleukin-1α (IL-1α) in response to damaged cells, promoting an excessive granulocyte colony-stimulating factor (G-CSF)-regulated neutrophilic response and prolonged inflammation. In peritoneal inflammation elicited by tissue injury, X-linked Cybb-null (X-CGD) mice exhibited increased release of IL-1α and IL-1 receptor -mediated G-CSF production. In turn, higher levels of systemic G-CSF increased peripheral neutrophilia, which amplified neutrophilic peritoneal inflammation in X-CGD mice. Dampening early neutrophil recruitment by neutralization of IL-1α, G-CSF, or neutrophil depletion itself promoted resolution of otherwise prolonged inflammation in X-CGD. IL-1β played little role. Thus, we identified an excessive IL-1α/G-CSF response as a major driver of enhanced sterile inflammation in CGD in the response to damaged cells. More broadly, these results provide new insights into the regulation of sterile inflammation, and identify the NADPH oxidase in regulating the amplitude of the early neutrophilic response.

DOI10.1182/blood-2015-05-644773
Custom 1

https://www.ncbi.nlm.nih.gov/pubmed/26443623?dopt=Abstract

Alternate JournalBlood
PubMed ID26443623
PubMed Central IDPMC4683333
Grant ListR37 AI049653 / AI / NIAID NIH HHS / United States
AI049653 / AI / NIAID NIH HHS / United States
P30 DK056341 / DK / NIDDK NIH HHS / United States
R01 AI049653 / AI / NIAID NIH HHS / United States
T32 DK094775 / DK / NIDDK NIH HHS / United States

Weill Cornell Medicine Gale and Ira Drukier Institute for Children's Health 413 E. 69th Street New York, NY 10021