Nanoparticulate carbon black in cigarette smoke induces DNA cleavage and Th17-mediated emphysema

Ran You, Wen Lu, Ming Shan, Jacob M Berlin, Errol Lg Samuel, Daniela C Marcano, Zhengzong Sun, William Ka Sikkema, Xiaoyi Yuan, Lizhen Song, Amanda Y Hendrix, James M Tour, David B Corry, Farrah Kheradmand, Ran You, Wen Lu, Ming Shan, Jacob M Berlin, Errol Lg Samuel, Daniela C Marcano, Zhengzong Sun, William Ka Sikkema, Xiaoyi Yuan, Lizhen Song, Amanda Y Hendrix, James M Tour, David B Corry, Farrah Kheradmand

Abstract

Chronic inhalation of cigarette smoke is the major cause of sterile inflammation and pulmonary emphysema. The effect of carbon black (CB), a universal constituent of smoke derived from the incomplete combustion of organic material, in smokers and non-smokers is less known. In this study, we show that insoluble nanoparticulate carbon black (nCB) accumulates in human myeloid dendritic cells (mDCs) from emphysematous lung and in CD11c(+) lung antigen presenting cells (APC) of mice exposed to smoke. Likewise, nCB intranasal administration induced emphysema in mouse lungs. Delivered by smoking or intranasally, nCB persisted indefinitely in mouse lung, activated lung APCs, and promoted T helper 17 cell differentiation through double-stranded DNA break (DSB) and ASC-mediated inflammasome assembly in phagocytes. Increasing the polarity or size of CB mitigated many adverse effects. Thus, nCB causes sterile inflammation, DSB, and emphysema and explains adverse health outcomes seen in smokers while implicating the dangers of nCB exposure in non-smokers.

Keywords: Th17; emphysema; human; human biology; immunology; inflammation; medicine; mouse.

Conflict of interest statement

The authors declare that no competing interests exist.

Figures

Figure 1.. Carbon black (CB) deposition in…
Figure 1.. Carbon black (CB) deposition in the lungs of patients with emphysema.
(A) Representative images of lung CD1a+ cells from a smoker with emphysema and a control subject. Scale bar: 10 μm. (B) Lung CD1a+ cells from a patient with emphysema, detected by transmission electron microscopy (TEM). Arrow indicates black substance in the vesicles. Scale bar: 1 μm. (C) Structure of the residual black material from digested human emphysema lung tissue, detected by high-resolution transmission electronic microscopy (HRTEM). Scale bar: 10 nm. (D) Raman spectrum yielded by the black material in the cells. The bifid spectral peaks between 1000 and 2000 cm−1 are the typical Raman signature for CB. Representative hyperspectral image of lung CD1a+ cells from a patient with emphysema (EH): a reference sample of nanoparticulate carbon black (nCB) was used to generate a signature spectral library (E) using CytoViva Hyperspectral Imaging System. Each colored spectra represents the spectral profile of a distinct area of the nCB sample, which were used in combination to map nCB present in cells. (F) Bright field (BF), (G) dark field (DF), and (H) overlay CB signature spectrum of lung CD1a+ cells. Positive signals were pseudo-colored red to aid visualization. Scale bar: 20 μm. (I) Raman spectrum yielded in lung CD11c+ and macrophages isolated from lungs of mice exposed to smoke for 4 months; CB reference (CB Ref) signal indicates solid CB sample. SMK: 4 months of cigarette smoke. Inset images for cell type correspond to Raman spectra indicating the subcellular localization of CB. The brightness of each 2 µm × 2 µm pixel, representing one spectrum, indicates the height of the graphitic band of CB at 1600 cm−1 compared to the background, such that brighter pixels indicate more CB. DOI:http://dx.doi.org/10.7554/eLife.09623.003
Figure 2.. Carbon black-induced emphysema mouse model.
Figure 2.. Carbon black-induced emphysema mouse model.
(A) Representative image of fresh lungs harvested from mice exposed to vehicle (PBS) or nanoparticulate carbon black (nCB) as described in Figure 2—figure supplement 1. (B) Representative Hematoxylin and eosin (H&E) staining of formalin-fixed lung sections. Scale bar: 100 μm. (C) Micro-CT quantification of lung volume. (D) MLI measurement was done on the same groups of mice. (E) Total and differential cell count in bronchoalveolar (BAL) fluid: macrophages (Mac), neutrophils (Neu), and lymphocytes (Lym). Quantitative PCR of Mmp9 and Mmp12 (F) gene expression in BAL cells isolated from PBS- or CB-challenged mice. Representative lung CD11c+ cells isolated from mice challenged with nCB under bright field (BF) (G), dark field (H), and overlap images (pseudo-red area) (I) signifying nCB signature spectrum. Scale bar: 20 μm. Data are mean ± SEM and representative of three independent experiments; ***p < 0.001, **p < 0.01 as determined by the Student's t-test; n = 5 per group. DOI:http://dx.doi.org/10.7554/eLife.09623.004
Figure 2—figure supplement 1.. Schematic representation of…
Figure 2—figure supplement 1.. Schematic representation of nCB-induced lung inflammation and emphysema protocol.
Mice were lightly anesthetized with isoflurane and challenged with 50 µl of 107 ng/ml of CB or vehicle (PBS with 1% sucrose) twice weekly for 6 weeks; 4 weeks following the last challenge, mice underwent CT scan of chest and were euthanized. DOI:http://dx.doi.org/10.7554/eLife.09623.005
Figure 2—figure supplement 2.. nCB induces pro-inflammatory…
Figure 2—figure supplement 2.. nCB induces pro-inflammatory cytokines and chemokines in the lung.
Concentration of pro-inflammatory cytokines and chemokines detected via MILLIPLEX Assay (Millipore, Billerica, MA) in the lung homogenate collected from mice in each group. n = 5 per group. ***p t-test and data are mean ± SEM and representative of three independent experiments. DOI:http://dx.doi.org/10.7554/eLife.09623.006
Figure 2—figure supplement 3.. nCB persists in…
Figure 2—figure supplement 3.. nCB persists in the lungs 18 months after the last challenge.
Representative image of left and right lobes of the lungs harvested from mice 18 months after the last nCB challenge. DOI:http://dx.doi.org/10.7554/eLife.09623.007
Figure 2—figure supplement 4.. nCB-induced emphysema persists…
Figure 2—figure supplement 4.. nCB-induced emphysema persists in the lungs.
Micro-CT quantification of lung volume in mice rested for 7 month after the last nCB or PBS challenge. ***p t-test, and data are mean ± SEM. DOI:http://dx.doi.org/10.7554/eLife.09623.008
Figure 2—figure supplement 5.. nCB-induced immune cell…
Figure 2—figure supplement 5.. nCB-induced immune cell infiltration persists in the lungs.
BAL fluid analysis of the mice rested for 7 month after the last nCB or PBS challenge showing the cell number of macrophages (Mac) and neutrophils (Neu). ***p t-test, and data are mean ± SEM. DOI:http://dx.doi.org/10.7554/eLife.09623.009
Figure 3.. nCB promotes Th17 responses.
Figure 3.. nCB promotes Th17 responses.
Representative staining (A) and cumulative analysis (B) of the percentage of CD11c+CD11bhigh cells in lung B220− cell subset. Representative intracellular staining (C) and cumulative analysis (D) of IL-17A+ cells expressing lung CD4+ T cell (Th17) subset. (E) Micro-CT quantification of lung volume in WT and Il-17a−/− mice. (F) Lung MLI was determined in the same group of mice. (G) BAL fluid analysis of the indicated groups of mice showing the total cells including macrophages (Mac), neutrophils (Neu), and lymphocytes (Lym). ***p < 0.001, **p < 0.01, *p < 0.05 as determined by the one-way ANOVA and Bonferroni's multiple comparison test. N = 4 to 6 per group. Data are mean ± SEM. (H) Representative H&E staining of formalin-fixed, 5-μm lung sections in indicated groups of mice. Scale bar: 100 μm. DOI:http://dx.doi.org/10.7554/eLife.09623.010
Figure 3—figure supplement 1.. nCB did not…
Figure 3—figure supplement 1.. nCB did not induce Th1 responses.
Cumulative analysis of intracellular cytokine staining of IFNγ in lung CD4+ T cell subsets in PBS or nCB-challenged mice. Data are mean ± SEM and representative of three independent experiments. DOI:http://dx.doi.org/10.7554/eLife.09623.011
Figure 3—figure supplement 2.. Lung APCs of…
Figure 3—figure supplement 2.. Lung APCs of nCB-challenged mice secrete Th17 cell-specific pro-inflammatory cytokines and chemokines.
Concentration of pro-inflammatory cytokines and chemokines detected by Multiplex system in the supernatant of overnight cultured of lung CD11c+ cells isolated from indicated groups.*p < 0.05 as determined by the Student's t-test. n = 3 per group. Data are mean ± SEM and representative of three independent experiments. DOI:http://dx.doi.org/10.7554/eLife.09623.012
Figure 3—figure supplement 3.. Lung APCs of…
Figure 3—figure supplement 3.. Lung APCs of nCB-challenged mice-induced Th17 responses.
Concentration of IL-17A, IFN-γ, and IL-4 expressed in the supernatant of lung CD11c+ cells isolated from CB- or PBS-challenged mice and co-cultured with splenic CD4+ T cells in the presence of anti-CD3 (1 μg/ml). *p < 0.05 as determined by the Student's t-test. n = 5 per group. Data are mean ± SEM and representative of three independent experiments. DOI:http://dx.doi.org/10.7554/eLife.09623.013
Figure 3—figure supplement 4.. nCB-induced Th17 responses…
Figure 3—figure supplement 4.. nCB-induced Th17 responses persist in the lungs.
Cumulative analysis of intracellular cytokine staining of IL-17A in lung CD4+ T cell subsets in the mice rested for 7 month after the last nCB or PBS. ***p < 0.001 as determined by the Student's t-test, and data are mean ± SEM. DOI:http://dx.doi.org/10.7554/eLife.09623.014
Figure 3—figure supplement 5.. Direct effect of…
Figure 3—figure supplement 5.. Direct effect of nCB on T helper cell differentiation in vitro.
(A) Flow cytometric analysis of intracellular cytokine staining of IFN-γ (Th1), IL-17A (Th17), and Foxp3/CD25 surface expression (Tregs). Diff. is the differentiation conditions for Th1, Th17, and Tregs (as described in the methods). Y-axis of both Th1 and Th17 panel is empty channel. (B) Cumulative summary of four independent experiments for Treg differentiation. **p < 0.01 as determined by the one-way ANOVA and Bonferroni's multiple comparison test. Data are mean ± SEM. DOI:http://dx.doi.org/10.7554/eLife.09623.015
Figure 4.. Hydrophobicity of nCB is important…
Figure 4.. Hydrophobicity of nCB is important for its pathogenesis.
Micro-CT quantification of lung volume (A) and MLI measurement of lung morphometry (B) in vehicle (PBS), nCB, and PEG-nCB treated mice. (C) Representative H&E staining of lung sections Scale bar: 100 μm. (D) Total and differential cell count in bronchoalveolar (BAL) fluid; macrophages (Mac), neutrophils (Neu), and lymphocytes (Lym). Quantitative PCR of Mmp9 (E) and Mmp12 (F) gene expression in BAL cells isolated from the above group of mice. Lung homogenate collected from indicated groups of mice were measured for IL-6 (G) and IL-1β (H) by ELISA. Representative intracellular staining (I) or cumulative analysis (J) of Th17 cells in the lungs. ***p < 0.001, **p < 0.01, *p < 0.05 as determined by the one-way ANOVA and Bonferroni's multiple comparison test. n = 4 to 6 per group, and data are mean ± SEM and representative of two independent studies. DOI:http://dx.doi.org/10.7554/eLife.09623.016
Figure 4—figure supplement 1.. nCB-induced cell damage…
Figure 4—figure supplement 1.. nCB-induced cell damage compared with PEG-nCB.
Representative image of H&E stained cytospin preparation of BAL cells isolated from indicated groups of mice. Scale bar: 50 μm. Data are representative of two independent studies. DOI:http://dx.doi.org/10.7554/eLife.09623.017
Figure 4—figure supplement 2.. nCB-induced cell death…
Figure 4—figure supplement 2.. nCB-induced cell death compared with PEG-nCB.
Lactate dehydrogenase (LDH) release from RAW 264.7 cells after 24 hr of the indicated treatment. Maximum LDH release was the amount of LDH released from lysed cells. ***p DOI: http://dx.doi.org/10.7554/eLife.09623.018
Figure 4—figure supplement 3.. nCB-induced strong lung…
Figure 4—figure supplement 3.. nCB-induced strong lung inflammation compared with PEG-nCB.
Multiplex analysis of pro-inflammatory cytokines and chemokines in lung homogenate of indicated groups. ***p DOI: http://dx.doi.org/10.7554/eLife.09623.019
Figure 5.. nCB activates APCs by the…
Figure 5.. nCB activates APCs by the induction of DNA damage and Erk signaling.
(A) Heat map (reverse phase protein array) of protein expression and phosphorylation level in RAW 264.7 cells stimulated with vehicle (PBS), nCB (105 ng/ml), and PEG-CB (105 ng/ml). p: phosphorylated. Blue is relatively low (−0.5) and yellow high (0.5) based on log2 ratio of the value for expression level. (B) RAW 264.7 cells under indicated conditions immunostained for nuclear DNA (DAPI, blue) and anti-γH2AX (green) to detect double strand break (DSB). Scale bar: 50 μm. (C) Quantitative summary of panel B indicating the percentage γH2AX positive RAW cells in indicated groups. (D) IL-6 concentration detected by ELISA after 48 hr in the supernatant of MDDC treated with CB or LPS in the presence of increasing dose of Nu7026 or vehicle (DMSO). (E) IL-17A concentration detected by ELISA after 72 hr co-culture of splenic CD4 T cells and lung CD11c+ cell isolated from the mice after challenged with PBS or nCB and anti-CD3 (1 μg/ml) in the presence of Nu7026 (100 nM), Ku55933 (100 nM), or vehicle control (DMSO). (F) Western blot of protein extracted from BMDC treated with different concentration of nCB targeting phosphorylated-Erk. Data are representative of two independent experiments. (G) IL-6 concentration detected by ELISA in the supernatant of MDDC treated with nCB in the presence of increasing dose of U0126 (MEK1/2 inhibitor) for 48 hr. n = 4 to 7 per group and data are mean ± SEM and representative of two independent experiments (C, D, E, G). ***p < 0.001, **p < 0.01 as determined by the one-way ANOVA and Bonferroni's multiple comparison test. DOI:http://dx.doi.org/10.7554/eLife.09623.020
Figure 5—figure supplement 1.. Heat map depicting…
Figure 5—figure supplement 1.. Heat map depicting molecules whose expression and phosphorylation level differed when RAW 264.7 cells were treated with nCB compared with PBS or PEG-nCB treated groups detected by reverse phase protein array.
DOI:http://dx.doi.org/10.7554/eLife.09623.021
Figure 5—figure supplement 2.. Larger nCB size…
Figure 5—figure supplement 2.. Larger nCB size correlates with weak induction of DNA double strand breaks (DSB).
(A) RAW 264.7 cells were treated with vehicle (PBS), nCB (105 ng/ml) with different size in diameter for overnight and immunostained for nuclear (DAPI) (blue) and anti-γH2AX (green) to detect double strand break (DSB). Scale bar: 100 μm. (B) Quantitative summary of panel A indicating the percentage γH2AX-positive RAW cells in indicated groups. ***p < 0.001 as determined by the one-way ANOVA and Bonferroni's multiple comparison test. n = 3 fields per group. Data are mean ± SEM. DOI:http://dx.doi.org/10.7554/eLife.09623.022
Figure 5—figure supplement 3.. ATM is required…
Figure 5—figure supplement 3.. ATM is required for nCB-induced inflammatory factor upregulation in RAW cells.
(A) Atm mRNA expression in RAW 264.7 cells transfected with scramble or Atm siRNA. (B) Il6 and Tnf-α mRNA expression in RAW cells transfected with scramble or Atm siRNA followed by overnight treatment with nCB or vehicle (PBS). ***p < 0.001, **p < 0.01, *p < 0.1 as determined by One-way ANOVA and Bonferroni's multiple comparison test. n = 4 per group. Data are mean ± SEM and representative of two independent studies. DOI:http://dx.doi.org/10.7554/eLife.09623.023
Figure 5—figure supplement 4.. Inhibition of DNA…
Figure 5—figure supplement 4.. Inhibition of DNA damage does not affect Th1 or Th2 responses.
IFN-γ (A) and IL-4 (B) concentrations were measured using ELISA in the supernatant of anti-CD3 (1 μg/ml) treated CD4+ T cells co-cultured with lung CD11c+ cell isolated from the mice challenged with PBS or nCB in the presence of vehicle (DMSO) or inhibitors of DNA damage (Nu7026 or Ku55933 at 100 nM). n = 5 per group, and data are mean ± SEM. DOI:http://dx.doi.org/10.7554/eLife.09623.024
Figure 6.. ASC-mediated inflammasome pathway is required…
Figure 6.. ASC-mediated inflammasome pathway is required for nCB-induced Th17 responses and emphysema.
(A) Representative H&E staining of lung sections from WT and Pycard−/− mice exposed to nCB or vehicle (PBS) as described in Figure 2—figure supplement 1. Scale bar: 100 μm. (B) Micro-CT quantification of lung volume in indicated groups of mice. (C) Lung MLI measurement in the same group of mice. (D) Total and differential cell count in bronchoalveolar (BAL) fluid: macrophages (Mac), neutrophils (Neu), and lymphocytes (Lym). (E) Relative abundance of lung mDCs (CD11c+CD11bhigh) isolated from whole lung tissue in the same group of mice. IL-6 (F) and IL-1β (G) concentrations detected by ELISA in the supernatant of lung CD11c+ cells isolated from indicated group of mice after overnight culture. (H) IL-17A concentration detected by ELISA in the supernatant of splenic CD4+ T cells co-cultured with lung CD11c+ cells isolated from indicated group of mice for 3 days in the presence of anti-CD3 (1 μg/ml). ***p < 0.001, **p < 0.01, *p < 0.05 as determined by the one-way ANOVA and Bonferroni's multiple comparison test; n = 3 to 7 per group, and data are mean ± SEM and representative of two independent studies. DOI:http://dx.doi.org/10.7554/eLife.09623.025
Figure 6—figure supplement 1.. Pycard −/− mice…
Figure 6—figure supplement 1.. Pycard−/− mice produce less pro-inflammatory chemokines in the lungs in response to nCB challenge.
(A) Multiplex detection of indicated chemokines in freshly harvested lung homogenate from different groups of mice. ***p < 0.001, *p < 0.05 as determined by the one-way ANOVA and Bonferroni's multiple comparison test. n = 5 per group. Data are mean ± SEM and representative of two independent studies. DOI:http://dx.doi.org/10.7554/eLife.09623.026
Author response image 1.
Author response image 1.
Lung volume and MLI figures without zero suppression. DOI:http://dx.doi.org/10.7554/eLife.09623.029
Author response image 2.. Inhibition of Erk…
Author response image 2.. Inhibition of Erk phosphorylation reduces IL-17A and IL-6.
CD11c+ lung cells were isolated from lungs of mice exposed to PBS or nCB as described in the manuscript and treated with increasing concentration of U0126 for two days (left panel). Using the same conditions cells were co-cultured with congenic splenic T cells (1:10 ratio) and antiCD3 (1μg/ml) in the presence of increasing concentration of U0126 for three days (right panel). Concentration of IL-17A, and IL-6 were measured using ELISA. ** p<0.01, *** p<0.001 as determined by the one-way ANOVA and Bonferroni’s Multiple Comparison test. DOI:http://dx.doi.org/10.7554/eLife.09623.030
Author response image 3.. Nu7026 and U0126…
Author response image 3.. Nu7026 and U0126 inhibit Erk phosphorylation.
BMDCs were treated with PBS or nCB in the presence of different inhibitors as indicated (Ku: Ku55933, 3μM; Nu: Nu7026, 3μM; U0: U0126, 0.1μM) for 24 hours and phosphorylated Erk (p-Erk), total Erk and β-actin were measured using western blot of cell lysates (left panel). Inhibition of p-Erk was examined using the same system in the presence of increasing concentration of U0126 (right panel). DOI:http://dx.doi.org/10.7554/eLife.09623.031

References

    1. Adam T, Mitschke S, Streibel T, Baker RR, Zimmermann R. Puff-by-puff resolved characterisation of cigarette mainstream smoke by single photon ionisation (SPI)-time-of-flight mass spectrometry (TOFMS): comparison of the 2R4F research cigarette and pure Burley, Virginia, Oriental and Maryland tobacco cigarettes. Analytica Chimica Acta. 2006;572:219–229. doi: 10.1016/j.aca.2006.05.043.
    1. Arif JM, Khan SG, Ashquin M, Rahman Q. Modulation of macrophage-mediated cytotoxicity by kerosene soot: possible role of reactive oxygen species. Environmental Research. 1993;61:232–238. doi: 10.1006/enrs.1993.1067.
    1. Baker RR. Temperature distribution inside a burning cigarette. Nature. 1974;247:405–406. doi: 10.1038/247405a0.
    1. Baker RR, Dixon M. The retention of tobacco smoke constituents in the human respiratory tract. Inhalation Toxicology. 2006;18:255–294. doi: 10.1080/08958370500444163.
    1. Barnes PJ. Cellular and molecular mechanisms of chronic obstructive pulmonary disease. Clinics in Chest Medicine. 2014;35:71–86. doi: 10.1016/j.ccm.2013.10.004.
    1. Bleck B, Tse DB, Jaspers I, Curotto de Lafaille MA, Reibman J. Diesel exhaust particle-exposed human bronchial epithelial cells induce dendritic cell maturation. The Journal of Immunology. 2006;176:7431–7437. doi: 10.4049/jimmunol.176.12.7431.
    1. Carter BD, Abnet CC, Feskanich D, Freedman ND, Hartge P, Lewis CE, Ockene JK, Prentice RL, Speizer FE, Thun MJ, Jacobs EJ. Smoking and mortality—beyond established causes. New England Journal of Medicine. 2015;372s:631–640. doi: 10.1056/NEJMc1503675#SA2.
    1. Chang Y, Al-Alwan L, Audusseau S, Chouiali F, Carlevaro-Fita J, Iwakura Y, Baglole CJ, Eidelman DH, Hamid Q. Genetic deletion of IL-17A reduces cigarette smoke-induced inflammation and alveolar type II cell apoptosis. American Journal of Physiology. 2014;306:L132–L143. doi: 10.1152/ajplung.00111.2013.
    1. Churg A, Myers JL, Tazelaar H, Wright JL. Thurlbeck's pathology of the lung. 3rd edition. New York, NY: Thieme; 2005.
    1. Churg A, Marshall CV, Sin DD, Bolton S, Zhou S, Thain K, Cadogan EB, Maltby J, Soars MG, Mallinder PR, Wright JL. Late intervention with a myeloperoxidase inhibitor stops progression of experimental chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine. 2012a;185:34–43. doi: 10.1164/rccm.201103-0468OC.
    1. Churg A, Zhou S, Wright JL. Series ‘matrix metalloproteinases in lung health and disease’: matrix metalloproteinases in COPD. European Respiratory Journal. 2012b;39:197–209. doi: 10.1183/09031936.00121611.
    1. Corry D, Kulkarni P, Lipscomb MF. The migration of bronchoalveolar macrophages into hilar lymph nodes. American Journal of Pathology. 1984;115:321–328.
    1. Cosio MG, Saetta M, Agusti A. Immunologic aspects of chronic obstructive pulmonary disease. New England Journal of Medicine. 2009;360:2445–2454. doi: 10.1056/NEJMra0804752.
    1. Dadvand P, Nieuwenhuijsen MJ, Agustí À, de Batlle J, Benet M, Beelen R, Cirach M, Martinez D, Hoek G, Basagaña X, Ferrer A, Ferrer J, Rodriguez-Roisin R, Sauleda J, Guerra S, Antó JM, Garcia-Aymerich J. Air pollution and biomarkers of systemic inflammation and tissue repair in COPD patients. European Respiratory Journal. 2014;44:603–613. doi: 10.1183/09031936.00168813.
    1. Eppert BL, Wortham BW, Flury JL, Borchers MT. Functional characterization of T cell populations in a mouse model of chronic obstructive pulmonary disease. The Journal of Immunology. 2013;190:1331–1340. doi: 10.4049/jimmunol.1202442.
    1. Eriksen M, Mackay J, Ross H. The Tobacco Atlas. 2014
    1. Franchi L, Nunez G. Immunology. Orchestrating inflammasomes. Science. 2012;337:1299–1300. doi: 10.1126/science.1229010.
    1. Furlaneto JA, Anderson AE, Jr, Foraker AG. Soot emphysema in a locomotive engineer. Archives of Environmental Health. 1969;18:1008–1013. doi: 10.1080/00039896.1969.10665527.
    1. Garza KM, Soto KF, Murr LE. Cytotoxicity and reactive oxygen species generation from aggregated carbon and carbonaceous nanoparticulate materials. International Journal of Nanomedicine. 2008;3:83–94. doi: 10.2217/17435889.3.1.83.
    1. Goswami S, Angkasekwinai P, Shan M, Greenlee KJ, Barranco WT, Polikepahad S, Seryshev A, Song LZ, Redding D, Singh B, Sur S, Woodruff P, Dong C, Corry DB, Kheradmand F. Divergent functions for airway epithelial matrix metalloproteinase 7 and retinoic acid in experimental asthma. Nature Immunology. 2009;10:496–503. doi: 10.1038/ni.1719.
    1. Guo Z, Deshpande R, Paull TT. ATM activation in the presence of oxidative stress. Cell Cycle. 2010;9:4805–4811. doi: 10.4161/cc.9.24.14323.
    1. Harwood LM, Moody CJ. Experimental organic chemistry: principles and practice. Hoboken, NJ: Wiley-Blackwell; 1989.
    1. Hwang CC, Ruan G, Wang L, Zheng H, Samuel EL, Xiang C, Lu W, Kasper W, Huang K, Peng Z, Schaefer Z, Kan AT, Martí AA, Wong MS, Tomson MB, Tour JM. Carbon-based nanoreporters designed for subsurface hydrogen sulfide detection. ACS Applied Materials and Interfaces. 2014;6:7652–7658. doi: 10.1021/am5009584.
    1. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans . IARC monographs on the evaluation of carcinogenic risks to humans. Volume 93. Lyon, France: 2010. pp. 1–466.
    1. Kheradmand F, Shan M, Xu C, Corry D. Autoimmunity in chronic obstructive pulmonary disease: clinical and experimental evidence. Expert Review of Clinical Immunology. 2012;8:285–292. doi: 10.1586/eci.12.7.
    1. Kono H, Orlowski GM, Patel Z, Rock KL. The IL-1-dependent sterile inflammatory response has a substantial caspase-1-independent component that requires cathepsin C. Journal of Immunology. 2012;189:3734–3740. doi: 10.4049/jimmunol.1200136.
    1. Kurimoto E, Miyahara N, Kanehiro A, Waseda K, Taniguchi A, Ikeda G, Koga H, Nishimori H, Tanimoto Y, Kataoka M, Iwakura Y, Gelfand EW, Tanimoto M. IL-17A is essential to the development of elastase-induced pulmonary inflammation and emphysema in mice. Respiratory Research. 2013;14:5. doi: 10.1186/1465-9921-14-5.
    1. Li Y, Yang D. The ATM inhibitor KU-55933 suppresses cell proliferation and induces apoptosis by blocking Akt in cancer cells with overactivated Akt. Molecular Cancer Therapeutics. 2010;1:113–125. doi: 10.1158/1535-7163.MCT-08-1189.
    1. Lu W, You R, Yuan X, Yang T, Samuel EL, Marcano DC, Sikkema WK, Tour JM, Rodriguez A, Kheradmand F, Corry DB. MicroRNA-22 Inhibits histone Deacytylase 4 to promote t Helper-17 cell-dependent emphysema. Nature Immunology. 2015 doi: 10.1038/ni.3292.
    1. Lutz MB, Kukutsch N, Ogilvie AL, Rössner S, Koch F, Romani N, Schuler G. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. Journal of Immunological Methods. 1999;223:77–92. doi: 10.1016/S0022-1759(98)00204-X.
    1. Mariathasan S, Newton K, Monack DM, Vucic D, French DM, Lee WP, Roose-Girma M, Erickson S, Dixit VM. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature. 2004;430:213–218. doi: 10.1038/nature02664.
    1. Merget R, Bauer T, Küpper HU, Philippou S, Bauer HD, Breitstadt R, Bruening T. Health hazards due to the inhalation of amorphous silica. Archives of Toxicology. 2002;75:625–634. doi: 10.1007/s002040100266.
    1. Mitchev K, Dumortier P, De Vuyst P. ‘Black Spots’ and hyaline pleural plaques on the parietal pleura of 150 urban necropsy cases. American Journal of Surgical Pathology. 2002;26:1198–1206. doi: 10.1097/00000478-200209000-00010.
    1. Newton R, Cambridge L, Hart LA, Stevens DA, Lindsay MA, Barnes PJ. The MAP kinase inhibitors, PD098059, UO126 and SB203580, inhibit IL-1beta-dependent PGE(2) release via mechanistically distinct processes. British Journal of Pharmacology. 2000;130:1353–1361. doi: 10.1038/sj.bjp.0703431.
    1. Oberdörster G, Oberdörster E, Oberdörster J. Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles. Environmental Health Perspectives. 2005;113:823–839. doi: 10.1289/ehp.7339.
    1. Perfetti TA, Rodgman A. The chemical components of tobacco and tobacco smoke. 2nd edition. Boca Raton, FL: CRC Press; 2013.
    1. Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediators of Inflammation. 2010 doi: 10.1155/2010/672395.
    1. Pilla D, Kavadi AKM, Gurijala P, Masuram S, Delaney MS, Merchant ME, Sneddon J. Determination of selected chlorohydrocarbons and polyaromatic hydrocarbons by gas chromatography–mass spectrometry in soils in Southwest Louisiana. Microchemical Journal. 2009;91:13–15. doi: 10.1016/j.microc.2008.06.002.
    1. Pope CA, III, Burnett RT, Turner MC, Cohen A, Krewski D, Jerrett M, Gapstur SM, Thun MJ. Lung cancer and cardiovascular disease mortality associated with ambient air pollution and cigarette smoke: shape of the exposure-response relationships. Environmental Health Perspectives. 2011;119:1616–1621. doi: 10.1289/ehp.1103639.
    1. Reisetter AC, Stebounova LV, Baltrusaitis J, Powers L, Gupta A, Grassian VH, Monick MM. Induction of inflammasome-dependent pyroptosis by carbon black nanoparticles. Journal of Biological Chemistry. 2011;286:21844–21852. doi: 10.1074/jbc.M111.238519.
    1. Reynolds ES. The use of lead citrate at high pH as an electron-opaque stain in electron microscopy. The Journal of Cell Biology. 1963;17:208–212. doi: 10.1083/jcb.17.1.208.
    1. Sahu S, Timari M, Bhangare R, Pandit G. Particle size distribution of mainstream and Exhaled cigarette smoke and Predictive deposition in human respiratory tract. Aerosol and Air Quality Research. 2013;13:324–332.
    1. Salvi S. Tobacco smoking and environmental risk factors for chronic obstructive pulmonary disease. Clinics in Chest Medicine. 2014;35:17–27. doi: 10.1016/j.ccm.2013.09.011.
    1. Shan M, Cheng HF, Song LZ, Roberts L, Green L, Hacken-Bitar J, Huh J, Bakaeen F, Coxson HO, Storness-Bliss C, Ramchandani M, Lee SH, Corry DB, Kheradmand F. Lung myeloid dendritic cells coordinately induce TH1 and TH17 responses in human emphysema. Science Translational Medicine. 2009;1:4ra10. doi: 10.1126/scitranlsmed.3000154.
    1. Shan M, You R, Yuan X, Frazier MV, Porter P, Seryshev A, Hong JS, Song LZ, Zhang Y, Hilsenbeck S, Whitehead L, Zarinkamar N, Perusich S, Corry DB, Kheradmand F. Agonistic induction of PPAR reverses cigarette smoke-induced emphysema. Journal of Clinical Investigation. 2014;124:1371–1381. doi: 10.1172/JCI70587.
    1. Shan M, Yuan X, Song LZ, Roberts L, Zarinkamar N, Seryshev A, Zhang Y, Hilsenbeck S, Chang SH, Dong C, Corry DB, Kheradmand F. Cigarette smoke induction of osteopontin (SPP1) mediates T(H)17 inflammation in human and experimental emphysema. Science Translational Medicine. 2012;4:117ra119. doi: 10.1126/scitranslmed.3003041.
    1. Spurr AR. A low-viscosity epoxy resin embedding medium for electron microscopy. Journal of Ultrastructure Research. 1969;26:31–43. doi: 10.1016/S0022-5320(69)90033-1.
    1. Wang B, Ho SSH, Ho KF, Huang Y, Chan CS, Feng NSY. An environmental Chamber study of the characteristics of air Pollutants released from environmental tobacco smoke. Aerosol and Air Quality Research. 2012;12:1269–1281.
    1. Watson J, Chow J, Chen L. Summary of organic and elemental Carbon/Black carbon analysis methods and Intercomparisons. Aerosol and Air Quality Research. 2005;5:65–102.
    1. Watson ML. Staining of tissue sections for electron microscopy with heavy metals. The Journal of Biophysical and Biochemical Cytology. 1958;4:475–478. doi: 10.1083/jcb.4.4.475.
    1. Wilmore E, de Caux S, Sunter NJ, Tilby MJ, Jackson GH, Austin CA, Durkacz BW. A novel DNA-dependent protein kinase inhibitor, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia. Blood. 2004;103:4659–4665. doi: 10.1182/blood-2003-07-2527.
    1. Zhang J, Chu S, Zhong X, Lao Q, He Z, Liang Y. Increased expression of CD4+IL-17+ cells in the lung tissue of patients with stable chronic obstructive pulmonary disease (COPD) and smokers. International Immunopharmacology. 2013;15:58–66. doi: 10.1016/j.intimp.2012.10.018.
    1. Zhou Y, Caron P, Legube G, Paull TT. Quantitation of DNA double-strand break resection intermediates in human cells. Nucleic Acids Research. 2014;42:e19. doi: 10.1093/nar/gkt1309.

Source: PubMed

3
Sottoscrivi