scholarly journals Carbon black nanoparticles induce biphasic gene expression changes associated with inflammatory responses in the lungs of C57BL/6 mice following a single intratracheal instillation

2015 ◽  
Vol 289 (3) ◽  
pp. 573-588 ◽  
Author(s):  
Mainul Husain ◽  
Zdenka O. Kyjovska ◽  
Julie Bourdon-Lacombe ◽  
Anne T. Saber ◽  
Keld A. Jensen ◽  
...  
Author(s):  
Qianghu Tang ◽  
Baijie Tu ◽  
Xuejun Jiang ◽  
Jun Zhang ◽  
Lulu Bai ◽  
...  

Carbon black nanoparticles (CBNPs) are one of the most frequently used nanoparticles. Exposure to CBNPs during pregnancy (PrE to CBNPs) can directly induce inflammation, lung injury and genotoxicity in dams, and results in abnormalities in offspring. However, whether exposure to CBNPs during pregnancy enhances the susceptibility of offspring to environmental stimuli remains unknown. To address this issue, in this study, we intranasally treated pregnant mice with mock or CBNPs from gestational day (GD) 9 to GD18, and F1 and F2 offspring were normally obtained. By intratracheal instillation of mice with lipopolysaccharide (LPS) to trigger a classic animal model for acute lung injury, we intriguingly found that after LPS treatment, F1 and F2 offspring after exposure during pregnancy to CBNPs both exhibited more pronounced lung injury symptoms, including more degenerative histopathological changes, vascular leakage, elevated MPO activity and activation of inflammation-related signaling transduction, compared to F1 and F2 offspring in the mock treatment group, suggesting PrE to CBNPs would aggravate LPS-induced lung injury in offspring, and this effect is intergenerational. We also observed that PrE to CBNPs upregulated the mRNA expression of DNA methyltransferases (Dnmt) 1/3a/3b and DNA hypermethylation in both F1 and F2 offspring, which might partially account for the intergenerational effect. Together, our study demonstrates for the first time that PrE to CBNPs can enhance sensitivity to LPS in both F1 and F2 offspring, and this intergenerational effect may be related to DNA hypermethylation caused by CBNPs.


2013 ◽  
Vol 13 (4) ◽  
pp. 406-412 ◽  
Author(s):  
Julie A. Bourdon ◽  
Anne T. Saber ◽  
Nicklas R. Jacobsen ◽  
Andrew Williams ◽  
Ulla Vogel ◽  
...  

2020 ◽  
Vol 36 (10) ◽  
pp. 759-768 ◽  
Author(s):  
Hui Lin ◽  
Guoqing Fu ◽  
Qimei Yu ◽  
Zhenyu Wang ◽  
Yao Zuo ◽  
...  

Long-term inhalation of carbon black nanoparticles (CBNPs) leads to pulmonary inflammatory diseases. Histone deacetylase 6 (HDAC6) has been identified as an important regulator in the development of inflammatory disorders. However, the direct involvement of HDAC6 in CBNPs-induced pulmonary inflammatory responses remains unclear. To explore whether HDAC6 participates in CBNPs-induced pulmonary inflammation, human bronchial epithelial cell line (16HBE cells) was transfected with HDAC6 small interference RNA (siRNA) and then exposed to CBNPs at concentrations of 0, 25, and 50 µg/ml for 24 h. Intracellular HDAC6 and intraflagellar transport protein 88 (IFT88) mRNA and protein were determined by real-time polymerase chain reaction and Western blot, respectively. The secretions of inflammatory cytokines including interleukin (IL)-8, tumor necrosis factor (TNF)-α, IL-6, and IL-1β were measured by enzyme-linked immunosorbent assay. CBNPs induced a significant increase in the expressions of IL-8 and IL-6, accompanied by a high level of intracellular HDAC6 mRNA when compared with a blank control group ( p < 0.05). However, there were no significant changes in the levels of TNF-α secretion, intracellular HDAC6 and IFT88 protein induced by CBNPs ( p > 0.05). The HDAC6 mRNA expression was significantly suppressed in HDAC6 siRNA-transfected cells ( p < 0.05). The secretions of IL-8, TNF-α, and IL-6 were significantly less in HDAC6 siRNA-transfected cells than that in normal 16HBE cells with exposure to 25 or 50 µg/ml of CBNPs, but intracellular IFT88 mRNA expression was markedly increased in HDAC6 siRNA-transfected cells when compared with normal 16HBE cells exposed to 50 µg/ml of CBNPs (all p < 0.05). Downregulation of the HDAC6 gene inhibits CBNPs-induced inflammatory responses in bronchial epithelial cells, partially through regulating IFT88 expression. It is suggested that CBNPs may trigger inflammatory responses in bronchial epithelial cells by an HDAC6/IFT88-dependent pathway.


Cellulose ◽  
2021 ◽  
Author(s):  
Katsuhide Fujita ◽  
Sawae Obara ◽  
Junko Maru ◽  
Shigehisa Endoh

Abstract Safety assessment of cellulose nanofibrils (CNFs) is required to accelerate the utilization of these materials in industrial applications. The present study aimed to characterize the effects on rat pulmonary inflammation over a period of 90 days following intratracheal instillation of three types of CNFs or multi-walled carbon nanotubes (MWCNTs) at doses of 0.5, 1.0, or 2.0 mg/kg. The pulmonary inflammatory responses induced by phosphorylated CNFs (CNF1), 2,2,6,6-tetramethylpiperidine-1-oxyl radical (TEMPO)-oxidized CNFs (CNF2), CNFs produced via mechanical defibrillation (CNF3), and MWCNTs were investigated using bronchoalveolar lavage fluid analysis, histopathological findings, and comprehensive gene expression profiling of rat lungs. CNF1 and CNF2 with approximately equal diameter (7.0–8.0 nm) and length (0.8–1.0 µm) distributions induced inflammation after dosing, which was attenuated 90 days post-instillation. CNF3 of relatively greater thickness (21.2 nm) and longer length (1.7 μm) deposited around the terminal bronchioles were observed after instillation. Acute inflammatory responses in the alveoli induced by CNF3 were mild compared with those induced by other materials and attenuated 90 days post-instillation. MWCNTs induced severe pulmonary inflammatory responses that continued during the test period. The inflammation failed to resolve within 90 days post-instillation. A hierarchical cluster analysis revealed comparable gene expression profiles for CNF1, CNF2, and CNF3, whereas profiles of MWCNTs were different from those of other test substances. This study suggests that pulmonary inflammation is associated with the diameter and length distributions of CNFs and that the pulmonary inflammation caused by CNFs is mild compared with that caused by MWCNTs. Graphic abstract


Applied Nano ◽  
2021 ◽  
Vol 2 (3) ◽  
pp. 222-241
Author(s):  
Justyna Modrzynska ◽  
Alicja Mortensen ◽  
Trine Berthing ◽  
Gitte Ravn-Haren ◽  
Józef Szarek ◽  
...  

Exposure to nanoparticles by various routes results in size-dependent translocation of nanoparticles to the systemic circulation and subsequent accumulation in the liver. The purpose of this study was to determine possible adverse effects in the liver of long-lasting nanoparticle presence in the organ. Mice exposed to a single dose (162 µg/animal equivalent to 9 mg/kg body weight) of TiO2, CeO2 or carbon black nanoparticles by intratracheal instillation or intravenous injection, resulting in relatively low or high liver burdens, were followed for 1, 28 or 180 days. Clinical appearance, feed intake, body and liver weights, hematological indices, and transaminases and alkaline phosphatase activities were unaffected by exposure. Exposure-related foreign material persisted in the liver up to 180 days after intratracheal and intravenous exposure, mainly in sinusoids, near Kupffer cells, or around blood vessels. Increased incidences of histological findings after intratracheal or intravenous exposure included: initially, prominent nuclei of Kupffer cells, the apparent increase in binucleate hepatocytes (TiO2 and carbon black) and inflammatory infiltrations (CeO2); later, cytoplasmic vacuolation, pyknosis and necrosis, especially for CeO2. Thus, neither low nor high nanoparticle burden in the liver affected enzymatic markers of liver injury, but indications of exposure-related necrotic changes, particularly for CeO2 nanoparticles, were noted.


Author(s):  
Nisha Verma ◽  
Mario Pink ◽  
Simone Schmitz-Spanke

AbstractToxicological studies propose that exposure to carbon black nanoparticles induces organ injuries and inflammatory responses. Besides, current understanding of the molecular mechanisms implies that carbon black nanoparticles (CBNP) exposure induces the production of reactive oxygen species (ROS) causing inflammation, mitochondrial dysfunction or disturbance in calcium homeostasis. However, the precise mechanisms whereby CBNP exert these effects in the lung are still not fully understood. To gain insight into the possible mechanism of CBNP exerted toxicity, human alveolar epithelial cells (A549) were exposed to different concentrations of CBNP and for different timepoints. The reaction of the cells was monitored by the systematic use of cell-based measurements of calcium and ROS, in the presence and absence of calcium (Ca2+) pump inhibitors/chelators and antioxidants. Followed by an in-depth PCR analysis of 84 oxidative stress-related genes. The measurements revealed, as compared to the control, that exposure to CBNP nanoparticles leads to the generation of high ROS levels, as well as a disturbance in calcium homeostasis, which remained primarily unchanged even after 24 h of exposure. Nevertheless, in presence of antioxidants N-acetylcysteine (NAC) and Trolox, ROS formation was considerably reduced without affecting the intracellular calcium concentration. On the other hand, Ca2+ pump inhibitors/chelators, BAPTA (1,2-bis(o-amino phenoxy)ethane-N, N, N′, N′-tetraacetic acid) and verapamil not only decreased the Ca2+ overload, but also further decreased the ROS formation, indicating its role in CBNP-induced oxidative stress. Further, a PCR array analysis of A549 cells in presence and absence of the calmodulin (CaM) antagonist W7, indicated toward nine altered oxidative stress-related genes which further confirmed our cytotoxicity results. Obtained data suggested that CBNP exposure elevates calcium ion concentration, which further contributes to oxidative stress, via the calcium-binding protein CaM. Its inhibition with W7 leads to downregulation in gene expression of nine oxidative stress-related genes, which otherwise, as compared to control, show increased gene expression. The results of the study thus confirm that exposure of lung epithelial cells to CBNP leads to oxidative stress; however, the oxidative stress itself is a result of a disturbance in both calcium and ROS homeostasis, and should be considered while searching for a new strategy for prevention of CBNP-induced lung toxicity.


Sign in / Sign up

Export Citation Format

Share Document