Butorphanol Promotes Macrophage Phenotype Transition to Inhibit Inflammatory Lung Injury Through κ Receptor

2021 ◽  
Author(s):  
Guangxin Luan ◽  
Fan Pan ◽  
Lina Bu ◽  
Kaixuan Wu ◽  
Aizhong Wang ◽  
...  
2019 ◽  
Vol 116 (33) ◽  
pp. 16513-16518 ◽  
Author(s):  
Saroj Nepal ◽  
Chinnaswamy Tiruppathi ◽  
Yoshikazu Tsukasaki ◽  
Joseph Farahany ◽  
Manish Mittal ◽  
...  

Efferocytosis of apoptotic neutrophils (PMNs) by alveolar macrophages (AMФs) is vital for resolution of inflammation and tissue injury. Here, we investigated the role of AMФ polarization and expression of the efferocytic ligand Gas6 in restoring homeostasis. In the murine model of lipopolysaccharide (LPS)-induced acute lung injury (ALI), we observed augmented temporal generation of cytokines IL-4 and TSG6 in bronchoalveolar fluid (BALF). Interestingly, we also observed increased expression of antiinflammatory markers consistent with a phenotype shift in AMФs. In particular, AMФs expressed the efferocytic ligand Gas6. In vitro priming of bone marrow–derived macrophages (BMMФs) with IL-4 or TSG6 also induced MФ transition and expression of Gas6. TSG6- or IL-4–primed BMMФs induced efferocytosis of apoptotic PMNs compared with control BMMФs. Adoptive transfer of TSG6- or IL-4–primed BMMФs i.t. into LPS-challenged mice more rapidly and effectively cleared PMNs in lungs compared with control BMMФs. We demonstrated that expression of Gas6 during AMФ transition was due to activation of the transcription factor signal transducer and activator of transcription-6 (STAT6) downstream of IL-4 or TSG6 signaling. Adoptive transfer of Gas6-depleted BMMФs failed to clear PMNs in lungs following LPS challenge and mice showed severely defective resolution of lung injury. Thus, activation of STAT6-mediated Gas6 expression during macrophage phenotype transition resulting in efferocytosis of PMNs plays a crucial role in the resolution of inflammatory lung injury.


2016 ◽  
Vol 113 (50) ◽  
pp. E8151-E8158 ◽  
Author(s):  
Manish Mittal ◽  
Chinnaswamy Tiruppathi ◽  
Saroj Nepal ◽  
You-Yang Zhao ◽  
Dagmara Grzych ◽  
...  

TNFα-stimulated gene-6 (TSG6), a 30-kDa protein generated by activated macrophages, modulates inflammation; however, its mechanism of action and role in the activation of macrophages are not fully understood. Here we observed markedly augmented LPS-induced inflammatory lung injury and mortality in TSG6−/− mice compared with WT (TSG6+/+) mice. Treatment of mice with intratracheal instillation of TSG6 prevented LPS-induced lung injury and neutrophil sequestration, and increased survival in mice. We found that TSG6 inhibited the association of TLR4 with MyD88, thereby suppressing NF-κB activation. TSG6 also prevented the expression of proinflammatory proteins (iNOS, IL-6, TNFα, IL-1β, and CXCL1) while increasing the expression of anti-inflammatory proteins (CD206, Chi3l3, IL-4, and IL-10) in macrophages. This shift was associated with suppressed activation of proinflammatory transcription factors STAT1 and STAT3. In addition, we observed that LPS itself up-regulated the expression of TSG6 in TSG6+/+ mice, suggesting an autocrine role for TSG6 in transitioning macrophages. Thus, TSG6 functions by converting macrophages from a proinflammatory to an anti-inflammatory phenotype secondary to suppression of TLR4/NF-κB signaling and STAT1 and STAT3 activation.


2015 ◽  
Vol 128 (16) ◽  
pp. 3094-3105 ◽  
Author(s):  
J. Thangavel ◽  
S. Samanta ◽  
S. Rajasingh ◽  
B. Barani ◽  
Y.-T. Xuan ◽  
...  

2013 ◽  
Vol 2013 ◽  
pp. 1-12 ◽  
Author(s):  
Mansoor A. Syed ◽  
Vineet Bhandari

Rationale. Hyperoxia exposure to developing lungs—critical in the pathogenesis of bronchopulmonary dysplasia—may augment lung inflammation by inhibiting anti-inflammatory mediators in alveolar macrophages.Objective. We sought to determine the O2-induced effects on the polarization of macrophages and the role of anti-inflammatory BRP-39 in macrophage phenotype and neonatal lung injury.Methods. We used RAW264.7, peritoneal, and bone marrow derived macrophages for polarization (M1/M2) studies. Forin vivostudies, wild-type (WT) and BRP-39−/−mice received continuous exposure to 21% O2(control mice) or 100% O2from postnatal (PN) 1 to PN7 days, along with intranasal lipopolysaccharide (LPS) administered on alternate days (PN2, -4, and -6). Lung histology, bronchoalveolar lavage (BAL) cell counts, BAL protein, and cytokines measurements were performed.Measurements and Main Results. Hyperoxia differentially contributed to macrophage polarization by enhancing LPS induced M1 and inhibiting interleukin-4 induced M2 phenotype. BRP-39 absence led to further enhancement of the hyperoxia and LPS induced M1 phenotype. In addition, BRP-39−/−mice were significantly more sensitive to LPS plus hyperoxia induced lung injury and mortality compared to WT mice.Conclusions. These findings collectively indicate that BRP-39 is involved in repressing the M1 proinflammatory phenotype in hyperoxia, thereby deactivating inflammatory responses in macrophages and preventing neonatal lung injury.


2018 ◽  
Vol 234 (4) ◽  
pp. 4217-4231 ◽  
Author(s):  
Tianyi Yu ◽  
Min Gao ◽  
Peilang Yang ◽  
Dan Liu ◽  
Di Wang ◽  
...  

2014 ◽  
Vol 184 (4) ◽  
pp. 1167-1184 ◽  
Author(s):  
Hanzhou Wang ◽  
David W. Melton ◽  
Laurel Porter ◽  
Zaheer U. Sarwar ◽  
Linda M. McManus ◽  
...  

Biomaterials ◽  
2017 ◽  
Vol 149 ◽  
pp. 77-87 ◽  
Author(s):  
Qiankun Zhang ◽  
Jin Wook Hwang ◽  
Joung-Hwan Oh ◽  
Chan Ho Park ◽  
Shin Hye Chung ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document