Thymic stromal lymphopoietin (TSLP) and Toluene-diisocyanate-induced airway inflammation: Alleviation by TSLP neutralizing antibody

2019 ◽  
Vol 317 ◽  
pp. 59-67 ◽  
Author(s):  
Gongchang Yu ◽  
Yu Zhang ◽  
Xiaoqun Wang ◽  
Linlin Sai ◽  
Cunxiang Bo ◽  
...  
1991 ◽  
Vol 143 (3_pt_2) ◽  
pp. S37-S38 ◽  
Author(s):  
Leonardo M. Fabbri ◽  
Piero Maestrelli ◽  
Marina Saetta ◽  
Cristina E. Mapp

2020 ◽  
Author(s):  
Xianru Peng ◽  
Minyu Huang ◽  
Wenqu Zhao ◽  
Zihan Lan ◽  
Xiaohua Wang ◽  
...  

Abstract BackgroundExposure to toluene diisocyanate (TDI) is a significant pathogenic factor for asthma. We previously reported that receptor for advanced glycation end products (RAGE) plays a key role in TDI-induced asthma; however, the mechanism is not clear. Epigenetic alterations of histone deacetylase (HDAC) are associated with allergic asthma. However, its effect in TDI-induced asthma is not known. The purpose of this study was to determine the role of RAGE and HDAC1 in the regulation of airway inflammation using a TDI-induced asthma model.MethodsBALB/c mice were sensitized, and challenged by TDI to establish murine asthma models. FPS-ZM1 (RAGE inhibitor), JNJ-26482585 and romidepsin (HDAC inhibitor) were given intraperitoneally before each challenge. The human bronchial epithelial cell line 16HBE was stimulated by TDI-human serum albumin (TDI-HSA) in vitro. RAGE knockdown cells were constructed and evaluated, and MK2006 (AKT inhibitor) was used in in vitro experiments.ResultsIn the TDI-induced asthmatic mice, airway reactivity, the level of Th2 cytokines in lymph supernatant, IgE, airway inflammation, and goblet cell metaplasia were all significantly increased. The increases were suppressed by FPS-ZM1, JNJ-26482585, and romidepsin. The expression of HDAC1, RAGE, and p-AKT/t-AKT was also upregulated in TDI-induced asthmatic mice, and the expressions were attenuated by FPS-ZM1. Knockdown of RAGE attenuated the upregulation of HDAC1 and phospho-AKT (p-AKT) in 16HBE cells stimulated by TDI-HSA. Treatment with the AKT inhibitor MK2006 suppressed TDI-induced HDAC1 expression. ConclusionRAGE mediates airway inflammation in a TDI-induced murine asthma model, partly via the HDAC1 pathway. Key words: Toluene diisocyanate, asthma, histone deacetylase 1, advanced glycosylation end product receptor


Author(s):  
Paul Harris ◽  
Jonathan Phillips ◽  
Ruoqi Peng ◽  
Lisa Burns ◽  
Lorena Renteria ◽  
...  

Respiration ◽  
1991 ◽  
Vol 58 (1) ◽  
pp. 18-21 ◽  
Author(s):  
Leonardo M. Fabbri ◽  
Marina Saetta ◽  
Guido Picotti ◽  
Christine E. Mapp

2005 ◽  
Vol 6 (10) ◽  
pp. 1047-1053 ◽  
Author(s):  
Baohua Zhou ◽  
Michael R Comeau ◽  
Thibaut De Smedt ◽  
H Denny Liggitt ◽  
Martin E Dahl ◽  
...  

2009 ◽  
Vol 29 (6) ◽  
pp. 786-794 ◽  
Author(s):  
Gyu-Young Hur ◽  
Sung-Ho Kim ◽  
Sang Myun Park ◽  
Young-Min Ye ◽  
Cheol-Woo Kim ◽  
...  

2014 ◽  
Vol 2014 ◽  
pp. 1-11 ◽  
Author(s):  
Fang Zhang ◽  
Gang Huang ◽  
Bo Hu ◽  
Li-Ping Fang ◽  
E-Hong Cao ◽  
...  

We demonstrate that high mobility group box 1 protein (HMGB1) directs Th17 skewing by regulating dendritic cell (DC) function. First, ourin vitrostudies reveal that recombinant HMGB1 (rHMGB1) activates myeloid DCs to produce IL-23in vitro, and rHMGB1-activated DCs prime naïve lymphocytes to produce the Th17 cytokine IL-17A. Second, we demonstrate that anti-HMGB1 neutralizing antibody attenuates HMGB1 expression, neutrophilic inflammation, airway hyperresponsiveness, and Th17-related cytokine secretionin vivoby using a murine model of neutrophilic asthma induced by ovalbumin (OVA) plus lipopolysaccharide (LPS). Furthermore, anti-HMGB1 neutralizing antibody decreases the number of Th17 cells in lung cells and suppresses the production of IL-23 by lung CD11C+APCs. Finally, we show that intranasal adoptive transfer of rHMGB1-activated DCs was sufficient to restore lung neutrophilic inflammation and the Th17 response in a DC-driven model of asthma, whereas the transfer of rHMGB1 plus anti-HMGB1-treated mDCs significantly reduced these inflammation phenotypes. These data suggest, for the first time, that HMGB1 drives the DC-polarized Th17-type response in allergic lung inflammation and that blocking HMGB1 may benefit the attenuation of neutrophilic airway inflammation in asthma.


Sign in / Sign up

Export Citation Format

Share Document