Chronic and low-level particulate matter exposure can sustainably mediate lung damage and alter CD4 T cells during acute lung injury

2019 ◽  
Vol 112 ◽  
pp. 51-58 ◽  
Author(s):  
Yusheng Li ◽  
Tiancao Dong ◽  
Xiaoping Jiang ◽  
Chunmei Wang ◽  
Ying Zhang ◽  
...  
2020 ◽  
Vol 125 ◽  
pp. 109946 ◽  
Author(s):  
Yu-sen Chai ◽  
Yan-qing Chen ◽  
Shi-hui Lin ◽  
Ke Xie ◽  
Chuan-jiang Wang ◽  
...  

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 766-766
Author(s):  
Rick Kapur ◽  
Michael Kim ◽  
Johan Rebetz ◽  
Alisa Takabe-French ◽  
Noel Kim ◽  
...  

Abstract Transfusion related acute lung injury (TRALI) is a syndrome of respiratory distress which occurs within 6 hours of blood transfusion. It is the leading cause of transfusion-related fatalities and the pathogenesis is incompletely understood. In the majority of the cases, anti-leukocyte antibodies present in the transfused blood product, in combination with recipient predisposing risk-factors such as inflammation, are implicated to be responsible for the onset of TRALI. Unfortunately, no therapies are available for TRALI. Recently, using novel murine models of TRALI, CD4+T cells were found to be important protector cells against antibody-mediated TRALI and administration of interleukin (IL)-10 was demonstrated to be a successful treatment strategy for TRALI, rescuing mice therapeutically from pulmonary edema, the hallmark of acute lung injury (Kapur et al, Blood 2017, 129(18):2557-2569). Whether the gut microbiome plays any role in the development of TRALI is currently unknown. For that purpose, we compared the biological TRALI response in mice housed in a barrier-free (BF) setting versus mice housed in a specific pathogen-free (SPF) environment. We utilized our TRALI model in which C57BL/6 mice were first depleted of CD4+ T cells in vivo followed by injection of anti-major histocompatibility complex class I antibodies (clones 34-1-2s and AF6-88.5.5.3). The TRALI response was analyzed after 90 minutes for several parameters including pulmonary edema (lung wet-to-dry weight ratios, W/Ds), rectal temperatures (indicative of systemic shock), plasma levels of macrophage inflammatory protein (MIP)-2 (murine homologue of IL-8, a neutrophil chemoattractant), levels of pulmonary neutrophils (major effector cells in TRALI) and lung histology. We observed that after one week of housing, baseline rectal temperatures of BF mice significantly increased from 38.7o C to 39.5° C (p<0.001) while SPF mice remained stable around 38.3 °C. When comparing BF versus SPF mice, the baseline temperature of BF mice was significantly higher than SPF mice after one week of housing (P<0.0001). The gut flora was subsequently depleted in BF mice via administration of broad-spectrum antibiotics through the drinking water. When both aerobic and anaerobic gut microbes were efficiently depleted after a week of antibiotic treatment, rectal temperatures of the treated BF mice significantly dropped again to 38.7 °C (P<0.05). When inducing TRALI by depleting CD4+ T cells and injecting TRALI-antibodies, the rectal temperatures of the untreated BF mice remained low while the rectal temperatures of SPF mice and gut flora depleted-BF mice normalized again from 60 minutes onwards (34.9 versus 36.8 versus 37.8 °C, respectively). Moreover, the untreated BF mice suffered from TRALI while the SPF mice and the gut flora depleted-BF mice were protected from TRALI development (lung W/Ds 5.77 versus 4.63 versus 4.49, respectively) which was also evident from lung histology-analyses. The prevention of TRALI in SPF mice and gut flora depleted BF mice was paralleled by decreased plasma MIP-2 levels compared to the untreated BF mice which suffered from TRALI (MIP-2: 0.09 versus 0.08 versus 82.94 pg/ml, respectively) which also corresponded to pulmonary neutrophil numbers. As previously shown, low IL-10 levels were associated with TRALI development and IL-10 KO mice were found to be susceptible to antibody-mediated murine TRALI (without prior in vivo cell-depletion). However, when IL-10 KO mice were housed under SPF conditions and injected with TRALI-inducing antibodies, they did not display increased lung W/Ds levels compared to naïve SPF mice indicating that the gut flora precedes and dictates the biological TRALI response. These data collectively link the gut flora to the development of antibody-mediated TRALI in mice and antibiotic treatment prevents TRALI and may perhaps prevent the onset of human TRALI which may help in TRALI risk-assessment prior to transfusion. Disclosures Semple: Rigel: Consultancy; Novartis: Consultancy; Amgen: Consultancy; UCB: Consultancy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 92-92
Author(s):  
Rick Kapur ◽  
Michael Kim ◽  
Shanjee Shanmugabhavananthan ◽  
Jonathan Liu ◽  
Noel Kim ◽  
...  

Abstract Transfusion related acute lung injury (TRALI) is the leading cause of transfusion-induced fatalities and is characterized by acute respiratory distress following blood transfusion. Donor antibodies present in the transfused blood product such as anti-human leukocyte antigen (HLA) or anti-human neutrophil antigen (HNA) antibodies are frequently involved. Currently, there is no treatment available for TRALI apart from supportive measures such as oxygen. The pathogenesis the disorder is incompletely understood, however, several animal models have contributed to our understanding of TRALI disease pathology. Most TRALI reactions are considered to be due to a two-hit paradigm where the first hit is a predisposing patient factor such as inflammation while the second hit is the transfusion. It is widely believed that the second hit delivers antibodies that trigger TRALI in the recipient. The anti-MHC class I antibody, 34-1-2s, has been widely used as an agent that delivers the second TRALI hit in mice. We have previously shown that CD4+ T cells, more specifically, CD4+CD25+FoxP3+ T-regulatory cells (Tregs) convey protection against TRALI (Blood. 126 (23):2342, 2015; abstract #82075, manuscript submitted). In the current study, we utilized a C57BL/6 mouse model of severe TRALI by first depleting mice of CD4+ T cells and then injecting them with the anti-MHC class I monoclonal antibodies (34-1-2s+AF6-88.5.5.3) and we examined the effects of the anti-inflammatory cytokine IL-10 on the antibody-mediated TRALI reaction. IL-10 (45 µg/kg iv) or volume-matched PBS was injected 15 minutes after the administration of anti-MHC antibodies when the onset of TRALI symptoms (e.g. a 2 degree drop in rectal temperature indicative of systemic shock) began. Results show that 90 minutes after anti-MHC class I antibody injection, control mice injected with PBS exhibited a high degree of pulmonary edema as assessed by significantly elevated lung wet-to-dry weight ratios (W/D: 5.84 ± 1.02). Pulmonary neutrophil levels were also found to be increased and lung tissue histology confirmed severe signs of acute lung injury. In contrast, mice injected with IL-10 completely recovered from TRALI; after 90 minutes post-antibody injection they displayed no signs of pulmonary edema (W/D: 4.76 ± 0.04, ** p<0.004 compared to mice injected with PBS) and no signs of severe acute lung injury as assessed by lung tissue histology. Pulmonary neutrophil levels, however, were equally increased in both groups indicating that although IL-10 rescues the mice from acute lung injury, it does not interfere with pulmonary neutrophil recruitment. Preliminary data suggests that IL-10 administration interferes with the ability of neutrophils to generate reactive oxygen species (ROS) that mediate lung injury. Our results suggest that IL-10 therapy significantly rescues an ongoing severe TRALI reaction and this may prove to be an effective and feasible therapeutic strategy in combating human TRALI. Disclosures No relevant conflicts of interest to declare.


2008 ◽  
Vol 81 (1) ◽  
pp. 9-15 ◽  
Author(s):  
Gautam K. Sahu ◽  
David Paar ◽  
Simon D.W. Frost ◽  
Melissa M. Smith ◽  
Scott Weaver ◽  
...  

2021 ◽  
Author(s):  
Irina N. Baranova ◽  
Alexander V. Bocharov ◽  
Tatyana G. Vishnyakova ◽  
Zhigang Chen ◽  
Anna A. Birukova ◽  
...  

Recent studies suggest an anti-inflammatory protective role for class B scavenger receptor BI (SR-BI) in endotoxin-induced inflammation and sepsis. Other data, including ours, provide evidence for an alternative role of SR-BI, facilitating bacterial and endotoxin uptake, and contributing to inflammation and bacterial infection. Enhanced endotoxin susceptibility of SR-BI deficient mice due to their anti-inflammatory glucocorticoid deficiency complicates understanding SR-BI’s role in endotoxemia/sepsis, calling for use of alternative models. In this study, using hSR-BI and hSR-BII transgenic mice, we found that SR-BI and to a lesser extent its splicing variant SR-BII, protects against LPS-induced lung damage. At 20 hours after intratracheal LPS instillation the extent of pulmonary inflammation and vascular leakage was significantly lower in hSR-BI and hSR-BII transgenic mice compared to wild type mice. Higher bronchoalveolar lavage fluid (BALF) inflammatory cell count and protein content as well as lung tissue neutrophil infiltration found in wild type mice was associated with markedly (2-3 times) increased pro-inflammatory cytokine production as compared to transgenic mice following LPS administration. Markedly lower endotoxin levels detected in BALF of transgenic vs. wild type mice along with the significantly increased BODIPY-LPS uptake observed in lungs of hSR-BI and hSR-BII mice 20 hours after the IT LPS injection suggest that hSR-BI and hSR-BII-mediated enhanced LPS clearance in the airways could represent the mechanism of their protective role against LPS-induced acute lung injury.


2018 ◽  
Vol 51 (6) ◽  
pp. 2776-2793 ◽  
Author(s):  
Yung-Fong Tsai ◽  
Shun-Chin Yang ◽  
Wen-Yi Chang ◽  
Jih-Jung Chen ◽  
Chun-Yu Chen ◽  
...  

Background/Aims: Formyl peptide receptors (FPRs) recognize different endogenous and exogenous molecular stimuli and mediate neutrophil activation. Dysregulation of excessive neutrophil activation and the resulting immune responses can induce acute lung injury (ALI) in the host. Accordingly, one promising approach to the treatment of neutrophil-dominated inflammatory diseases involves therapeutic FPR1 inhibition. Methods: We extracted a potent FPR1 antagonist from Garcinia multiflora Champ. (GMC). The inhibitory effects of GMC on superoxide anion release and elastase degranulation from activated human neutrophils were determined with spectrophotometric analysis. Reactive oxygen species (ROS) production and the FPR1 binding ability of neutrophils were assayed by flow cytometry. Signaling transduction mediated by GMC in response to chemoattractants was assessed with a calcium influx assay and western blotting. A lipopolysaccharide (LPS)-induced ALI mouse model was used to determine the therapeutic effects of GMC in vivo. Results: GMC significantly reduced superoxide anion release, the reactive oxidants derived therefrom, and elastase degranulation mediated through selective, competitive FPR1 blocking in N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLF)-stimulated human neutrophils. In cell-free systems, GMC was unable to scavenge superoxide anions or suppress elastase activity. GMC produced a right shift in fMLF-activated concentration-response curves and was confirmed to be a competitive FPR1 antagonist. GMC binds to FPR1 not only in neutrophils, but also FPR1 in neutrophil-like THP-1 and hFPR1-transfected HEK293 cells. Furthermore, the mobilization of calcium and phosphorylation of mitogen-activated protein kinases and Akt, which are involved in FPR1-mediated downstream signaling, was competitively blocked by GMC. In an in vivo study, GMC significantly reduced pulmonary edema, neutrophil infiltration, and alveolar damage in LPS-induced ALI mice. Conclusion: Our findings demonstrate that GMC is a natural competitive FPR1 inhibitor, which makes it a possible anti-inflammatory treatment option for patients critically inflicted with FPR1-mediated neutrophilic lung damage.


2018 ◽  
Vol 88 (5) ◽  
pp. e12715 ◽  
Author(s):  
Linlin Wang ◽  
Xiaocen Wang ◽  
Lin Tong ◽  
Jian Wang ◽  
Maosen Dou ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document