Faculty Opinions recommendation of IL-4 receptor engagement in human neutrophils impairs their migration and extracellular trap formation.

Author(s):  
Warren J Leonard
2021 ◽  
pp. 2003312
Author(s):  
Sofia Winslow ◽  
Lina Odqvist ◽  
Sarah Diver ◽  
Rebecca Riise ◽  
Suado Abdillahi ◽  
...  

BackgroundIL-6 trans-signalling (IL-6TS) is emerging as a pathogenic mechanism in chronic respiratory diseases, however the drivers of IL-6TS in the airways and the phenotypic characteristic of patients with increased IL-6TS pathway activation remain poorly understood.ObjectiveOur aim was to identify and characterize COPD patients with increased airway IL-6TS and to elucidate the biological drivers of IL-6TS pathway activation.MethodsWe used an IL-6TS-specific sputum biomarker profile (sIL-6R, IL-6, IL-1β, IL-8, MIP-1β) to stratify sputum data from patients with COPD (n=74; BEAT-COPD) by hierarchical clustering. The IL-6TS signature was related to clinical characteristics and sputum microbiome profiles. The induction of neutrophil extracellular trap formation (NETosis) and IL-6TS by Haemophilus influenzae were studied in human neutrophils.ResultsHierarchical clustering revealed an IL-6TS-high subset (n=24) of COPD patients, which shared phenotypic traits with an IL-6TS-high subset previously identified in asthma. The subset was characterized by increased sputum cell counts (p=0.0001), persistent sputum neutrophilia (p=0.0004), reduced quality of life (CRQ total score; p=0.008), and increased levels of pro-inflammatory mediators and MMPs in sputum. IL-6TS-high COPD patients showed an increase in Proteobacteria, with Haemophilus as the dominating genus. NETosis induced by H. influenzae was identified as a potential mechanism for increased soluble IL-6 receptor (sIL-6R) levels. This was supported by a significant positive correlation between sIL-6R and NETosis markers in bronchoalveolar lavage fluid from COPD patients.ConclusionIL-6TS pathway activation due to chronic colonization with Haemophilus may be an important disease driver in a subset of COPD patients.


2016 ◽  
Vol 93 ◽  
pp. 190-203 ◽  
Author(s):  
Deepika Awasthi ◽  
Sheela Nagarkoti ◽  
Amit Kumar ◽  
Megha Dubey ◽  
Abhishek Kumar Singh ◽  
...  

PLoS ONE ◽  
2015 ◽  
Vol 10 (4) ◽  
pp. e0124082 ◽  
Author(s):  
Giselle A. Funchal ◽  
Natália Jaeger ◽  
Rafael S. Czepielewski ◽  
Mileni S. Machado ◽  
Stéfanie P. Muraro ◽  
...  

Discoveries ◽  
2014 ◽  
Vol 2 (2) ◽  
pp. e19 ◽  
Author(s):  
Mona Saffarzadeh ◽  
Hector A. Cabrera-Fuentes ◽  
Florian Veit ◽  
Dongsheng Jiang ◽  
Karin Scharffetter-Kochanek ◽  
...  

2019 ◽  
Vol 144 (1) ◽  
pp. 267-279.e4 ◽  
Author(s):  
Daniela Impellizzieri ◽  
Frederike Ridder ◽  
Miro E. Raeber ◽  
Cecilie Egholm ◽  
Janine Woytschak ◽  
...  

2020 ◽  
Vol 4 (2) ◽  
pp. 312-321 ◽  
Author(s):  
Brianna M. Craver ◽  
Gajalakshmi Ramanathan ◽  
Summer Hoang ◽  
Xinyue Chang ◽  
Laura F. Mendez Luque ◽  
...  

Abstract Thrombosis is a major cause of mortality in patients with myeloproliferative neoplasms (MPNs), though there is currently little to offer patients with MPN beyond aspirin and cytoreductive therapies such as hydroxyurea for primary prevention. Thrombogenesis in MPN involves multiple cellular mechanisms, including platelet activation and neutrophil-extracellular trap formation; therefore, an antithrombotic agent that targets one or more of these processes would be of therapeutic benefit in MPN. Here, we treated the JAK2V617F knockin mouse model of polycythemia vera with N-acetylcysteine (NAC), a sulfhydryl-containing compound with broad effects on glutathione replenishment, free radical scavenging, and reducing disulfide bonds, to investigate its antithrombotic effects in the context of MPN. Strikingly, NAC treatment extended the lifespan of JAK2V617F mice without impacting blood counts or splenomegaly. Using an acute pulmonary thrombosis model in vivo, we found that NAC reduced thrombus formation to a similar extent as the irreversible platelet inhibitor aspirin. In vitro analysis of platelet activation revealed that NAC reduced thrombin-induced platelet-leukocyte aggregate formation in JAK2V617F mice. Furthermore, NAC reduced neutrophil extracellular trap formation in primary human neutrophils from patients with MPN as well as healthy controls. These results provide evidence that N-acetylcysteine inhibits thrombosis in JAK2V617F mice and provide a pre-clinical rationale for investigating NAC as a therapeutic to reduce thrombotic risk in MPN.


Sign in / Sign up

Export Citation Format

Share Document