Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Toxicol Appl Pharmacol ; 461: 116388, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36690086

RESUMO

Chlorine (Cl2) gas is a highly toxic and oxidizing irritant that causes life-threatening lung injuries. Herein, we investigated the impact of Cl2-induced injury and oxidative stress on lung macrophage phenotype and function. Spontaneously breathing male C57BL/6J mice were exposed to air or Cl2 (300 ppm, 25 min) in a whole-body exposure chamber. Bronchoalveolar lavage (BAL) fluid and cells, and lung tissue were collected 24 h later and analyzed for markers of injury, oxidative stress and macrophage activation. Exposure of mice to Cl2 resulted in increases in numbers of BAL cells and levels of IgM, total protein, and fibrinogen, indicating alveolar epithelial barrier dysfunction and inflammation. BAL levels of inflammatory proteins including surfactant protein (SP)-D, soluble receptor for glycation end product (sRAGE) and matrix metalloproteinase (MMP)-9 were also increased. Cl2 inhalation resulted in upregulation of phospho-histone H2A.X, a marker of double-strand DNA breaks in the bronchiolar epithelium and alveolar cells; oxidative stress proteins, heme oxygenase (HO)-1 and catalase were also upregulated. Flow cytometric analysis of BAL cells revealed increases in proinflammatory macrophages following Cl2 exposure, whereas numbers of resident and antiinflammatory macrophages were not altered. This was associated with increases in numbers of macrophages expressing cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS), markers of proinflammatory activation, with no effect on mannose receptor (MR) or Ym-1 expression, markers of antiinflammatory activation. Metabolic analysis of lung cells showed increases in glycolytic activity following Cl2 exposure in line with proinflammatory macrophage activation. Mechanistic understanding of Cl2-induced injury will be useful in the identification of efficacious countermeasures for mitigating morbidity and mortality of this highly toxic gas.


Assuntos
Cloro , Lesão Pulmonar , Camundongos , Masculino , Animais , Cloro/toxicidade , Camundongos Endogâmicos C57BL , Pulmão , Macrófagos , Líquido da Lavagem Broncoalveolar , Estresse Oxidativo , Metabolismo Energético
2.
Drug Chem Toxicol ; 42(3): 321-327, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30426789

RESUMO

Methyl isocyanate (MIC) is a highly toxic industrial chemical causing acute lethality after inhalation. The objective of this study was to determine whether alterations in hemostasis also occur in the immediate hours after exposure. Male rats were exposed to MIC (125-500 ppm) by nose-only vapor inhalation for 30 min. Arterial O2 saturation was monitored prior to exposure, and hourly thereafter. Rats were euthanized at 1, 2, 4, and 8 hr and plasma analyzed for recalcification clotting time, tissue factor (TF) activity, and protein levels. Hypoxemia, as assessed by pulse oximetry, was an early feature of MIC inhalation. In contrast to sham or low (125 ppm) concentrations, 250 and 500 ppm MIC caused significant declines in blood oxygen saturation (% SpO2) at 1 hr, which remained at deficit during the postexposure period. Commensurate with hypoxemia, plasma clotting time was significantly accelerated 1 hr after MIC inhalation (sham treatment: 955 ± 62.8 s; 125 ppm MIC: 790 ± 62 s; 250 ppm: 676 ± 28.0 s; 500 ppm: 581 ± 175 s). This procoagulant effect was transient, with no difference observed between sham and all MIC groups by 8 hr. Similarly, elevated TF activity and protein were detected in plasma 1 hr after MIC inhalation, each of which showed a progressive decline back to control levels at later timepoints. This study demonstrates that MIC inhalation resulted in hypoxemia and transient hypercoagulability of blood. Accelerated clotting occurred rapidly and was likely due to intravascular TF, which initiates the extrinsic coagulation pathway.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Exposição por Inalação/efeitos adversos , Isocianatos/toxicidade , Tromboplastina/metabolismo , Animais , Relação Dose-Resposta a Droga , Hipóxia/sangue , Hipóxia/induzido quimicamente , Masculino , Oxigênio/sangue , Ratos , Ratos Sprague-Dawley
3.
J Immunol ; 195(8): 3866-79, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26342031

RESUMO

Elevated serum concentrations of the vasoactive protein endothelin-1 (ET-1) occur in the setting of systemic inflammatory response syndrome and contribute to distal organ hypoperfusion and pulmonary hypertension. Thus, understanding the cellular source and transcriptional regulation of systemic inflammatory stress-induced ET-1 expression may reveal therapeutic targets. Using a murine model of LPS-induced septic shock, we demonstrate that the hepatic macrophage is the primary source of elevated circulating ET-1, rather than the endothelium as previously proposed. Using pharmacologic inhibitors, ET-1 promoter luciferase assays, and by silencing and overexpressing NF-κB inhibitory protein IκB expression, we demonstrate that LPS-induced ET-1 expression occurs via an NF-κB-dependent pathway. Finally, the specific role of the cRel/p65 inhibitory protein IκBß was evaluated. Although cytoplasmic IκBß inhibits activity of cRel-containing NF-κB dimers, nuclear IκBß stabilizes NF-κB/DNA binding and enhances gene expression. Using targeted pharmacologic therapies to specifically prevent IκBß/NF-κB signaling, as well as mice genetically modified to overexpress IκBß, we show that nuclear IκBß is both necessary and sufficient to drive LPS-induced ET-1 expression. Together, these results mechanistically link the innate immune response mediated by IκBß/NF-κB to ET-1 expression and potentially reveal therapeutic targets for patients with Gram-negative septic shock.


Assuntos
Endotelina-1/imunologia , Endotoxemia/imunologia , Regulação da Expressão Gênica/imunologia , Proteínas I-kappa B/imunologia , Fígado/imunologia , Macrófagos/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular , Endotelina-1/genética , Endotoxemia/genética , Endotoxemia/patologia , Proteínas I-kappa B/genética , Fígado/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , NF-kappa B/genética , Transdução de Sinais/genética
4.
Am J Respir Cell Mol Biol ; 51(4): 559-67, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24796565

RESUMO

Acute lung injury in response to mustard gas (sulfur mustard [SM]) inhalation results in formation of fibrin casts, which obstruct the airway. The objective of this study was to identify fibrinolytic pathways that could be contributing to the persistence of airway casts after SM exposure. Rats were exposed to the SM analog, 2-chloroethyl ethyl sulfide, via nose-only aerosol inhalation. At 4 and 18 hours after exposure, animals were killed and airway-capillary leak estimated by measuring bronchoalveolar lavage fluid (BALF) protein and IgM content. The fibrin clot-degrading and plasminogen-activating capabilities of BALF were also assessed by activity assays, whereas Western blotting was used to determine the presence and activities of plasminogen activator inhibitor-1, thrombin activatable fibrinolytic inhibitor and α2-antiplasmin. Measurement of tissue-specific steady-state mRNA levels was also conducted for each fibrinolytic inhibitor to assess whether its synthesis occurs in lung or at extrapulmonary sites. The results of this study demonstrate that fibrin-degrading and plasminogen-activating capabilities of the airways become impaired during the onset of 2-chloroethyl ethyl sulfide-induced vascular leak. Findings of functionally active reservoirs of plasminogen activator inhibitor-1, thrombin activatable fibrinolysis inhibitor, and α2-antiplasmin in BALF indicate that airway fibrinolysis is inhibited at multiple levels in response to SM.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Antifibrinolíticos/toxicidade , Substâncias para a Guerra Química/toxicidade , Fibrinólise/efeitos dos fármacos , Exposição por Inalação , Pulmão/efeitos dos fármacos , Gás de Mostarda/análogos & derivados , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Obstrução das Vias Respiratórias/induzido quimicamente , Obstrução das Vias Respiratórias/metabolismo , Obstrução das Vias Respiratórias/patologia , Animais , Barreira Alveolocapilar/efeitos dos fármacos , Barreira Alveolocapilar/metabolismo , Líquido da Lavagem Broncoalveolar/química , Permeabilidade Capilar/efeitos dos fármacos , Carboxipeptidase B2/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Gás de Mostarda/toxicidade , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo , alfa 2-Antiplasmina/metabolismo
5.
Am J Respir Cell Mol Biol ; 48(1): 94-104, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23065128

RESUMO

Tissue factor (TF) initiates the extrinsic coagulation cascade and is a high-affinity receptor for coagulation factor VII. TF also participates in protease-activated receptor (PAR)1 and PAR2 activation. Human epithelial basal cells were previously purified on the basis of TF expression. The purpose of this study was to determine if tracheobronchial epithelial basal cell-associated TF drives coagulation and/or activates PARs to promote basal cell functions. We used human tracheobronchial tissues to isolate human airway epithelial cells using specific cell surface markers by flow cytometry and studied TF expression by immunostaining. TF-dependent fibrin network formation was observed by confocal and scanning electron microscopy. TF knockdown was done using short hairpin RNA, and TF mRNA was measured using quantitative RT-PCR. We found that 97 ± 5% of first-passage human tracheobronchial epithelial cells were basal cells, and 100% of these basal cells expressed TF. Basal cell-associated TF was active, but TF activity was dependent on added extrinsic coagulation cascade factors. TF inhibition caused basal cell apoptosis and necrosis. This was due to two parallel but interdependent TF-regulated processes: failure to generate a basal cell-associated fibrin network and suboptimal PAR1 and PAR2 activity. The data indicate that membrane surface TF mediates airway epithelial basal cell attachment, which maintains cell survival and mitotic potential. The implications of these findings are discussed in the context of basal cell-associated TF activity in normal and injured tissues and of the potential for repair of airway epithelium in lung disease.


Assuntos
Fator VII/fisiologia , Receptor PAR-1/fisiologia , Receptor PAR-2/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , Tromboplastina/fisiologia , Coagulação Sanguínea/fisiologia , Brônquios/citologia , Brônquios/fisiologia , Adesão Celular/fisiologia , Morte Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Fibrina/fisiologia , Fibrina/ultraestrutura , Técnicas de Silenciamento de Genes , Humanos , RNA Interferente Pequeno/genética , Transdução de Sinais , Tromboplastina/antagonistas & inibidores , Tromboplastina/genética , Traqueia/citologia , Traqueia/fisiologia , Cicatrização/fisiologia
6.
Toxicol Appl Pharmacol ; 272(1): 86-95, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23727623

RESUMO

UNLABELLED: Sulfur mustard (SM) inhalation causes airway injury, with enhanced vascular permeability, coagulation, and airway obstruction. The objective of this study was to determine whether recombinant tissue factor pathway inhibitor (TFPI) could inhibit this pathogenic sequence. METHODS: Rats were exposed to the SM analog 2-chloroethyl ethyl sulfide (CEES) via nose-only aerosol inhalation. One hour later, TFPI (1.5mg/kg) in vehicle, or vehicle alone, was instilled into the trachea. Arterial O2 saturation was monitored using pulse oximetry. Twelve hours after exposure, animals were euthanized and bronchoalveolar lavage fluid (BALF) and plasma were analyzed for prothrombin, thrombin-antithrombin complex (TAT), active plasminogen activator inhibitor-1 (PAI-1) levels, and fluid fibrinolytic capacity. Lung steady-state PAI-1 mRNA was measured by RT-PCR analysis. Airway-capillary leak was estimated by BALF protein and IgM, and by pleural fluid measurement. In additional animals, airway cast formation was assessed by microdissection and immunohistochemical detection of airway fibrin. RESULTS: Airway obstruction in the form of fibrin-containing casts was evident in central conducting airways of rats receiving CEES. TFPI decreased cast formation, and limited severe hypoxemia. Findings of reduced prothrombin consumption, and lower TAT complexes in BALF, demonstrated that TFPI acted to limit thrombin activation in airways. TFPI, however, did not appreciably affect CEES-induced airway protein leak, PAI-1 mRNA induction, or inhibition of the fibrinolytic activity present in airway surface liquid. CONCLUSIONS: Intratracheal administration of TFPI limits airway obstruction, improves gas exchange, and prevents mortality in rats with sulfur mustard-analog-induced acute lung injury.


Assuntos
Obstrução das Vias Respiratórias/induzido quimicamente , Obstrução das Vias Respiratórias/prevenção & controle , Substâncias para a Guerra Química/toxicidade , Lipoproteínas/farmacologia , Gás de Mostarda/análogos & derivados , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/prevenção & controle , Administração por Inalação , Obstrução das Vias Respiratórias/patologia , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Substâncias para a Guerra Química/farmacocinética , Ensaio de Imunoadsorção Enzimática , Fibrina/metabolismo , Fibrinólise/efeitos dos fármacos , Imunoglobulina M/metabolismo , Imuno-Histoquímica , Indicadores e Reagentes , Masculino , Microdissecção , Gás de Mostarda/farmacocinética , Gás de Mostarda/toxicidade , Proteínas/farmacologia , Protrombina/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Insuficiência Respiratória/patologia
7.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L82-92, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21964405

RESUMO

Acute lung injury is a principal cause of morbidity and mortality in response to mustard gas (SM) inhalation. Obstructive, fibrin-containing airway casts have recently been reported in a rat inhalation model employing the SM analog 2-chloroethyl ethyl sulfide (CEES). The present study was designed to identify the mechanism(s) causing activation of the coagulation cascade after CEES-induced airway injury. Here we report that CEES inhalation elevates tissue factor (TF) activity and numbers of detached epithelial cells present in lavage fluid (BALF) from rats after exposure (18 h). In vitro studies using 16HBE cells, or with rat BALF, indicated that detached epithelial cells could convert factor X (FX) to the active form FXa when incubated with factor VII and could elicit rapid clotting of plasma. In addition, immunocytochemical analysis demonstrated elevated cell surface (TF) expression on CEES-exposed 16HBE cells as a function of time. However, total cell TF expression did not increase. Since membrane surfaces bearing TF are important determinants of clot initiation, anticoagulants directed against these entities were tested for ability to limit plasma clotting or FX activation capacity of BALF or culture media. Addition of tifacogin, a TF pathway inhibitor, effectively blocked either activity, demonstrating that the procoagulant actions of CEES were TF pathway dependent. Lactadherin, a protein capable of competing with clotting factors for phospholipid-binding sites, was partially effective in limiting these procoagulant actions. These findings indicate that TF pathway inhibition could be an effective strategy to prevent airway obstruction after SM or CEES inhalation.


Assuntos
Obstrução das Vias Respiratórias/induzido quimicamente , Brônquios/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Gás de Mostarda/análogos & derivados , Tromboplastina/metabolismo , Animais , Antígenos de Superfície/metabolismo , Brônquios/citologia , Brônquios/enzimologia , Líquido da Lavagem Broncoalveolar/citologia , Linhagem Celular Transformada , Substâncias para a Guerra Química/toxicidade , Modelos Animais de Doenças , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Fator VII/metabolismo , Fator Xa/metabolismo , Humanos , Exposição por Inalação , Masculino , Proteínas do Leite/metabolismo , Gás de Mostarda/toxicidade , Proteínas/farmacologia , Ratos , Ratos Sprague-Dawley , Tromboplastina/antagonistas & inibidores , Fatores de Tempo
8.
Toxicol Sci ; 187(1): 162-174, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35201360

RESUMO

Ozone is a ubiquitous air pollutant that causes lung damage and altered functioning. Evidence suggests that proinflammatory macrophages contribute to ozone toxicity. Herein, we analyzed the role of extracellular vesicles (EVs) and microRNA (miRNA) cargo in ozone-induced macrophage activation. Exposure of mice to ozone (0.8 ppm, 3 h) resulted in increases in bronchoalveolar lavage fluid EVs, which were comprised predominantly of microvesicles (MVs). NanoFACS analysis revealed that MVs generated following both air and ozone exposure was largely from CD45+ myeloid cells; these MVs were readily taken up by macrophages. Functionally, MVs from ozone, but not air treated mice, upregulated mRNA expression of inflammatory proteins in macrophages including inducible nitric oxide synthase (iNOS), CXCL-1, CXCL-2, and interleukin (IL)-1ß. The miRNA profile of MVs in bronchoalveolar lavage fluid (BALF) was altered after ozone exposure; thus, increases in miR-21, miR-145, miR320a, miR-155, let-7b, miR744, miR181, miR-17, miR-92a, and miR-199a-3p were observed, whereas miR-24-3p and miR-20 were reduced. Ingenuity pathway analysis revealed that these miRNAs regulate pathways that promote inflammatory macrophage activation, and predicted that let-7a-5p/let-7b, miR-24-3p, miR-21-5p, miR-17, and miR-181a-5p are key upstream regulators of inflammatory proteins. After ozone exposure, miR-199a-3p, but not precursor miR-199a-3p, was increased in lung macrophages, indicating that it is derived from MV-mediated delivery. Furthermore, lung macrophage mRNA expression of IL-1ß was upregulated after administration of MVs containing miR-199a-3p mimic but downregulated by miR-199a-3p inhibitor. Collectively, these data suggest that MVs generated following ozone exposure contribute to proinflammatory macrophage activation via MV-derived miRNAs including miR-199a-3p. These findings identify a novel pathway regulating macrophage inflammatory responses to inhaled ozone.


Assuntos
MicroRNAs , Ozônio , Animais , Pulmão/metabolismo , Ativação de Macrófagos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Ozônio/toxicidade , RNA Mensageiro/metabolismo
9.
Am J Respir Crit Care Med ; 182(11): 1352-61, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20639443

RESUMO

RATIONALE: Sulfur mustard (SM) is a frequently used chemical warfare agent, even in modern history. SM inhalation causes significant respiratory tract injury, with early complications due to airway obstructive bronchial casts, akin to those seen after smoke inhalation and in single-ventricle physiology. This process with SM is poorly understood because animal models are unavailable. OBJECTIVES: To develop a rat inhalation model for airway obstruction with the SM analog 2-chloroethyl ethyl sulfide (CEES), and to investigate the pathogenesis of bronchial cast formation. METHODS: Adult rats were exposed to 0, 5, or 7.5% CEES in ethanol via nose-only aerosol inhalation (15 min). Airway microdissection and confocal microscopy were used to assess cast formation (4 and 18 h after exposure). Bronchoalveolar lavage fluid (BALF) retrieval and intravascular dye injection were done to evaluate vascular permeability. MEASUREMENTS AND MAIN RESULTS: Bronchial casts, composed of abundant fibrin and lacking mucus, occluded dependent lobar bronchi within 18 hours of CEES exposure. BALF contained elevated concentrations of IgM, protein, and fibrin. Accumulation of fibrin-rich fluid in peribronchovascular regions (4 h) preceded cast formation. Monastral blue dye leakage identified bronchial vessels as the site of leakage. CONCLUSIONS: After CEES inhalation, increased permeability from damaged bronchial vessels underlying damaged airway epithelium leads to the appearance of plasma proteins in both peribronchovascular regions and BALF. The subsequent formation of fibrin-rich casts within the airways then leads to airways obstruction, causing significant morbidity and mortality acutely after exposure.


Assuntos
Obstrução das Vias Respiratórias/induzido quimicamente , Brônquios/irrigação sanguínea , Brônquios/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Gás de Mostarda/toxicidade , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar , Permeabilidade Capilar/efeitos dos fármacos , Modelos Animais de Doenças , Fibrina/efeitos dos fármacos , Imunoglobulina M/efeitos dos fármacos , Exposição por Inalação , Masculino , Microdissecção , Microscopia Confocal , Ratos , Ratos Sprague-Dawley
10.
Toxicol Sci ; 178(2): 358-374, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33002157

RESUMO

Sulfur mustard (SM) inhalation causes debilitating pulmonary injury in humans which progresses to fibrosis. Herein, we developed a rat model of SM toxicity which parallels pathological changes in the respiratory tract observed in humans. SM vapor inhalation caused dose (0.2-0.6 mg/kg)-related damage to the respiratory tract within 3 days of exposure. At 0.4-0.6 mg/kg, ulceration of the proximal bronchioles, edema and inflammation were observed, along with a proteinaceous exudate containing inflammatory cells in alveolar regions. Time course studies revealed that the pathologic response was biphasic. Thus, changes observed at 3 days post-SM were reduced at 7-16 days; this was followed by more robust aberrations at 28 days, including epithelial necrosis and hyperplasia in the distal bronchioles, thickened alveolar walls, enlarged vacuolated macrophages, and interstitial fibrosis. Histopathologic changes were correlated with biphasic increases in bronchoalveolar lavage (BAL) cell and protein content and proliferating cell nuclear antigen expression. Proinflammatory proteins receptor for advanced glycation end product (RAGE), high-mobility group box protein (HMGB)-1, and matrix metalloproteinase (MMP)-9 also increased in a biphasic manner following SM inhalation, along with surfactant protein-D (SP-D). Tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS), inflammatory proteins implicated in mustard lung toxicity, and the proinflammatory/profibrotic protein, galectin (Gal)-3, were upregulated in alveolar macrophages and in bronchiolar regions at 3 and 28 days post-SM. Inflammatory changes in the lung were associated with oxidative stress, as reflected by increased expression of heme oxygenase (HO)-1. These data demonstrate a similar pathologic response to inhaled SM in rats and humans suggesting that this rodent model can be used for mechanistic studies and for the identification of efficacious therapeutics for mitigating toxicity.


Assuntos
Substâncias para a Guerra Química , Lesão Pulmonar , Gás de Mostarda , Animais , Substâncias para a Guerra Química/toxicidade , Fibrose , Inflamação/patologia , Pulmão/efeitos dos fármacos , Lesão Pulmonar/patologia , Gás de Mostarda/toxicidade , Estresse Oxidativo , Ratos
11.
Antioxid Redox Signal ; 10(2): 277-85, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18158760

RESUMO

Lipoic acid (LA) and its reduced product dihydrolipoic acid (DHLA) are potent antioxidants with capacity to scavenge reactive oxygen species (ROS) and recycle endogenous antioxidants. LA may increase cellular glutathione (GSH), an antioxidant lacking in the lung's epithelial lining fluid in lung disorders such as idiopathic pulmonary fibrosis (IPF). Neutrophils (PMN) are key innate responders and are pivotal in clearing bacterial infection, therefore it is crucial to understand the impact LA may have on their function. Circulating neutrophils were isolated from healthy volunteers and pretreated with LA or diluent. Cells were subsequently activated with phorbol 12-myristate 13-acetate (PMA, 100 ng/ml) to induce ROS production. SOD-inhibitable reduction of acetylated cytochrome c demonstrated the PMA-dependent respiratory burst was suppressed by LA. Oxygen consumption also was diminished when PMA-stimulated cells were pretreated with LA. PMN respiratory burst was partially restored by addition of NADPH but not other pyridine nucleotides. LA did not inhibit glucose-6-phosphate dehydrogenase activity of PMN. These data together suggest that the reduction of LA to DHLA using cellular NADPH may limit the capacity of the PMN NADPH oxidase to produce superoxide. Further studies will be required to determine if LA can diminish excessive superoxide produced by PMN and/or alveolar macrophages in IPF or relevant disease models in vivo.


Assuntos
Ácido Fólico/farmacologia , NADP/farmacologia , Neutrófilos/fisiologia , Explosão Respiratória/fisiologia , Ácido Tióctico/farmacologia , Infecções Bacterianas/prevenção & controle , Glucosefosfato Desidrogenase/sangue , Humanos , Cinética , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Consumo de Oxigênio , Peroxidase/sangue , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória/efeitos dos fármacos , Superóxidos/sangue
12.
Free Radic Biol Med ; 42(9): 1441-53, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17395017

RESUMO

Thioredoxin (Trx) decreases viscosity of cystic fibrosis (CF) sputum. In this study reduced Trx increased the solubility and decreased the size of MUC5B glycoprotein while reducing disulfide bonds in sputum. Because Trx used as a mucolytic would enter airways, this study determined the effects of intratracheal instillation of reduced recombinant human thioredoxin (rhTrx) in naïve rat airways. Reduced rhTrx increased neutrophils and the cytokines TNFalpha, CINC2beta, and MIP3alpha in airways after 4 h. The effect of rhTrx was concentration-dependent. Exposure to saline, human serum albumin, or oxidized rhTrx at equal molarities did not increase airway neutrophils or cytokines. Instilling CF sputum (50 microl) into the lung before reduced rhTrx delivery attenuated these responses. This suggests that rhTrx reduces disulfide bonds present in CF sputum, limiting the reduction of other lung constituents. Together these findings indicate that the chemotactic and cytokine responses are due to the reducing potential of rhTrx and that the potential for inflammation in non-CF and CF patients given aerosolized rhTrx may differ. In parallel studies, increased amounts of the p65 subunit of NF-kappaB were present in nuclear extracts from rat lungs administered reduced rhTrx, suggesting a role for NF-kappaB in these proinflammatory responses.


Assuntos
Citocinas/biossíntese , Inflamação/fisiopatologia , Neutrófilos/fisiologia , Mucosa Respiratória/fisiopatologia , Tiorredoxinas/metabolismo , Animais , Arabidopsis , Proteínas de Arabidopsis/metabolismo , Clonagem Molecular , Humanos , Cinética , Modelos Animais , Mucina-5B , Mucinas/metabolismo , Oxirredução , Ratos , Proteínas Recombinantes/metabolismo , Solubilidade , Tiorredoxina Dissulfeto Redutase/genética , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxinas/genética
13.
Toxicol Sci ; 157(2): 399-409, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28402575

RESUMO

Sulfur mustard (SM) is a chemical warfare agent that causes chronic airway remodeling. This study's objective was to assess for changes to the bronchiolar epithelium after SM exposure to explain its contribution to chronic airway remodeling. Materials and methods: Adult male rats were exposed to a sublethal dose of SM inhalation (1.0-1.2 mg/kg) for 50 min. Histological sections of the bronchiolar epithelium were analyzed for changes using hematoxylin and eosin, trichrome, and immunofluorescent staining for acetylated tubulin (AT) and club cell secretory protein (CCSP). CCSP in bronchoalveolar lavage fluid was assessed using western blot. A bromodeoxyuridine (BRDU) assay was used to assess for epithelial proliferation, and real-time PCR measured changes in Notch mRNA expression. Results: SM caused significant proximal bronchiolar epithelial injury with epithelial denudation, loss of acetylated tubulin and CCSP staining, and reduced bronchoalveolar lavage fluid CCSP levels. bromodeoxyuridine (BRDU) + staining of proximal bronchiolar epithelial cells was not increased, but staining was increased in the distal bronchiolar epithelium. One month after injury, the proximal bronchiolar epithelium was not fully repaired. Significant collagen deposition surrounded proximal bronchioles with luminal obstruction, consistent with bronchiolitis obliterans. These changes corresponded with a downregulation of Notch1, Notch3, and Hes1 mRNA expressions. Conclusions: This study demonstrates that SM exposure resulted in severe proximal airway epithelial injury, persistent morphological changes, impaired epithelial proliferation and, ultimately, bronchiolitis obliterans. These changes occurred at the same time that the Notch signaling genes were downregulated. Thus, the lung epithelium and the Notch signaling pathway may be worthy targets for the prevention of chronic airway remodeling after SM inhalation injury.


Assuntos
Bronquiolite Obliterante/induzido quimicamente , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Gás de Mostarda/toxicidade , Mucosa Respiratória/efeitos dos fármacos , Remodelação das Vias Aéreas/efeitos dos fármacos , Animais , Bronquiolite Obliterante/metabolismo , Bronquiolite Obliterante/patologia , Exposição por Inalação/efeitos adversos , Masculino , Ratos Sprague-Dawley , Receptores Notch/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
14.
Toxicol Sci ; 159(2): 461-469, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28962529

RESUMO

Sulfur mustard (SM) is a chemical warfare agent. When inhaled, SM causes significant injury to the respiratory tract. Although the mechanism involved in acute airway injury after SM inhalation has been well described previously, the mechanism of SM's contribution to distal lung vascular injury is not well understood. We hypothesized that acute inhalation of vaporized SM causes activated systemic coagulation with subsequent pulmonary vascular thrombi formation after SM inhalation exposure. Sprague Dawley rats inhaled SM ethanolic vapor (3.8 mg/kg). Barium/gelatin CT pulmonary angiograms were performed to assess for pulmonary vascular thrombi burden. Lung immunohistochemistry was performed for common procoagulant markers including fibrin(ogen), von Willebrand factor, and CD42d in control and SM-exposed lungs. Additionally, systemic levels of d-dimer and platelet aggregometry after adenosine diphosphate- and thrombin-stimulation were measured in plasma after SM exposure. In SM-exposed lungs, chest CT angiography demonstrated a significant decrease in the distal pulmonary vessel density assessed at 6 h postexposure. Immunohistochemistry also demonstrated increased intravascular fibrin(ogen), vascular von Willebrand factor, and platelet CD42d in the distal pulmonary vessels (<200 µm diameter). Circulating d-dimer levels were significantly increased (p < .001) at 6, 9, and 12 h after SM inhalation versus controls. Platelet aggregation was also increased in both adenosine diphosphate - (p < .01) and thrombin- (p < .001) stimulated platelet-rich plasma after SM inhalation. Significant pulmonary vascular thrombi formation was evident in distal pulmonary arterioles following SM inhalation in rats assessed by CT angiography and immunohistochemistry. Enhanced systemic platelet aggregation and activated systemic coagulation with subsequent thrombi formation likely contributed to pulmonary vessel occlusion.


Assuntos
Arteríolas/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Pulmão/efeitos dos fármacos , Gás de Mostarda/toxicidade , Trombose/induzido quimicamente , Animais , Arteríolas/patologia , Angiografia por Tomografia Computadorizada , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Exposição por Inalação , Pulmão/irrigação sanguínea , Pneumopatias/induzido quimicamente , Masculino , Gás de Mostarda/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
15.
Ann N Y Acad Sci ; 1378(1): 87-95, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27384912

RESUMO

Acute lung injury due to sulfur mustard (SM) inhalation causes the formation of airway fibrin casts that obstruct airways at multiple levels, leading to acute respiratory failure and death. These pathophysiological effects are seen in rodent models of acute SM vapor inhalation, as well as in human victims of acute SM inhalation. In rat models, the initial steps in activation of the coagulation system at extravascular sites depend on tissue factor (TF) expression by airway cells, especially in the microparticle fraction, and these effects can be inhibited by TF pathway inhibitor protein. Not only does the procoagulant environment of the acutely injured lung contribute to airway cast formation, but these lesions persist in airways because of the activation of multiple antifibrinolytic pathways, including plasminogen activator inhibitor-1, thrombin-activatable fibrinolysis inhibitor, and α2-antiplasmin. Airway administration of tissue plasminogen activator can overwhelm these effects and save lives by preventing fibrin-dependent airway obstruction, gas-exchange abnormalities, and respiratory failure. In human survivors of SM inhalation, fibrotic processes, including bronchiolitis obliterans and interstitial fibrosis of the lung, are among the most disabling chronic lesions. Antifibrotic therapies may prove useful in preventing either or both of these forms of chronic lung damage.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Coagulação Sanguínea/efeitos dos fármacos , Fibrinolíticos/uso terapêutico , Exposição por Inalação/efeitos adversos , Gás de Mostarda/toxicidade , Lesão Pulmonar Aguda/sangue , Animais , Coagulação Sanguínea/fisiologia , Humanos , Exposição por Inalação/prevenção & controle , Gás de Mostarda/administração & dosagem
16.
Pediatr Pulmonol ; 50(2): 118-26, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24692161

RESUMO

BACKGROUND: Inhalation of sulfur mustard (SM) and SM analog, 2-chloroethyl ethyl sulfide (CEES), cause fibrinous cast formation that occludes the conducting airways, similar to children with Fontan physiology-induced plastic bronchitis. These airway casts cause significant mortality and morbidity, including hypoxemia and respiratory distress. Our hypothesis was that intratracheal heparin, a highly cost effective and easily preserved rescue therapy, could reverse morbidity and mortality induced by bronchial cast formation. METHODS: Sprague-Dawley rats were exposed to 7.5% CEES via nose-only aerosol inhalation to produce extensive cast formation and mortality. The rats were distributed into three groups: non-treated, phosphate-buffered saline (PBS)-treated, and heparin-treated groups. Morbidity was assessed with oxygen saturations and clinical distress. Blood and bronchoalveolar lavage fluid (BALF) were obtained for analysis, and lungs were fixed for airway microdissection to quantify the extent of airway cast formation. RESULTS: Heparin, given intratracheally, improved survival (100%) when compared to non-treated (75%) and PBS-treated (90%) controls. Heparin-treated rats also had improved oxygen saturations, clinical distress and airway cast scores. Heparin-treated rats had increased thrombin clotting times, factor Xa inhibition and activated partial thromboplastin times, indicating systemic absorption of heparin. There were also increased red blood cells (RBCs) in the BALF in 2/6 heparin-treated rats compared to PBS-treated control rats. CONCLUSIONS: Intratracheal heparin 1 hr after CEES inhalation improved survival, oxygenation, airway obstruction, and clinical distress. There was systemic absorption of heparin in rats treated intratracheally. Some rats had increased RBCs in BALF, suggesting a potential for intrapulmonary bleeding if used chronically after SM inhalation.


Assuntos
Bronquite/tratamento farmacológico , Substâncias para a Guerra Química/toxicidade , Fibrinolíticos/administração & dosagem , Heparina/administração & dosagem , Gás de Mostarda/análogos & derivados , Animais , Testes de Coagulação Sanguínea , Líquido da Lavagem Broncoalveolar/citologia , Vias de Administração de Medicamentos , Eritrócitos/metabolismo , Modelos Animais , Gás de Mostarda/toxicidade , Oxigênio/sangue , Ratos Sprague-Dawley , Traqueia
17.
Exp Toxicol Pathol ; 67(2): 161-70, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25481215

RESUMO

Vesicating agents sulfur mustard (SM) and nitrogen mustard (NM) are reported to be easily absorbed by skin upon exposure causing severe cutaneous injury and blistering. Our studies show that topical exposure of NM (3.2mg) onto SKH-1 hairless mouse skin, not only caused skin injury, but also led to significant body weight loss and 40-80% mortality (120 h post-exposure), suggesting its systemic effects. Accordingly, further studies herein show that NM exposure initiated an increase in circulating white blood cells by 24h (neutrophils, eosinophils and basophils) and thereafter a decrease (neutrophils, lymphocytes and monocytes). NM exposure also reduced both white and red pulp areas of the spleen. In the small intestine, NM exposure caused loss of membrane integrity of the surface epithelium, abnormal structure of glands and degeneration of villi. NM exposure also resulted in the dilation of glomerular capillaries of kidneys, and an increase in blood urea nitrogen/creatinine ratio. Our results here with NM are consistent with earlier reports that exposure to higher SM levels can cause damage to the hematopoietic system, and kidney, spleen and gastrointestinal tract toxicity. These outcomes will add to our understanding of the toxic effects of topical vesicant exposure, which might be helpful towards developing effective countermeasures against injuries from acute topical exposures.


Assuntos
Substâncias para a Guerra Química/toxicidade , Sistema Hematopoético/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Rim/efeitos dos fármacos , Mecloretamina/toxicidade , Pele/efeitos dos fármacos , Baço/efeitos dos fármacos , Administração Cutânea , Animais , Apoptose/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Intestino Delgado/patologia , Rim/patologia , Contagem de Leucócitos , Masculino , Camundongos Pelados , Tamanho do Órgão/efeitos dos fármacos , Pele/lesões , Baço/patologia , Análise de Sobrevida
18.
Toxicol Sci ; 143(1): 178-84, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25331496

RESUMO

RATIONALE: Sulfur mustard (SM) is a chemical weapon stockpiled today in volatile regions of the world. SM inhalation causes a life-threatening airway injury characterized by airway obstruction from fibrin casts, which can lead to respiratory failure and death. Mortality in those requiring intubation is more than 80%. No therapy exists to prevent mortality after SM exposure. Our previous work using the less toxic analog of SM, 2-chloroethyl ethyl sulfide, identified tissue plasminogen activator (tPA) an effective rescue therapy for airway cast obstruction (Veress, L. A., Hendry-Hofer, T. B., Loader, J. E., Rioux, J. S., Garlick, R. B., and White, C. W. (2013). Tissue plasminogen activator prevents mortality from sulfur mustard analog-induced airway obstruction. Am. J. Respir. Cell Mol. Biol. 48, 439-447). It is not known if exposure to neat SM vapor, the primary agent used in chemical warfare, will also cause death due to airway casts, and if tPA could be used to improve outcome. METHODS: Adult rats were exposed to SM, and when oxygen saturation reached less than 85% (median: 6.5 h), intratracheal tPA or placebo was given under isoflurane anesthesia every 4 h for 48 h. Oxygen saturation, clinical distress, and arterial blood gases were assessed. Microdissection was done to assess airway obstruction by casts. RESULTS: Intratracheal tPA treatment eliminated mortality (0% at 48 h) and greatly improved morbidity after lethal SM inhalation (100% death in controls). tPA normalized SM-associated hypoxemia, hypercarbia, and lactic acidosis, and improved respiratory distress. Moreover, tPA treatment resulted in greatly diminished airway casts, preventing respiratory failure from airway obstruction. CONCLUSIONS: tPA given via airway more than 6 h after exposure prevented death from lethal SM inhalation, and normalized oxygenation and ventilation defects, thereby rescuing from respiratory distress and failure. Intra-airway tPA should be considered as a life-saving rescue therapy after a significant SM inhalation exposure incident.


Assuntos
Obstrução das Vias Respiratórias/tratamento farmacológico , Substâncias para a Guerra Química , Fibrinolíticos/administração & dosagem , Exposição por Inalação , Pulmão/efeitos dos fármacos , Gás de Mostarda , Insuficiência Respiratória/prevenção & controle , Terapia Trombolítica , Ativador de Plasminogênio Tecidual/administração & dosagem , Acidose/induzido quimicamente , Acidose/prevenção & controle , Administração por Inalação , Obstrução das Vias Respiratórias/induzido quimicamente , Obstrução das Vias Respiratórias/patologia , Obstrução das Vias Respiratórias/fisiopatologia , Animais , Modelos Animais de Doenças , Esquema de Medicação , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Oxigênio/sangue , Ventilação Pulmonar/efeitos dos fármacos , Ratos Sprague-Dawley , Respiração/efeitos dos fármacos , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/patologia , Insuficiência Respiratória/fisiopatologia , Fatores de Tempo
19.
Antioxid Redox Signal ; 6(1): 129-36, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14713344

RESUMO

The effect of hyperoxia on levels of DNA damage and global DNA methylation was examined in lung epithelial-like A549 cells. DNA damage was assessed by the single-cell gel electrophoresis (comet assay) and DNA methylation status by the cytosine extension assays. Cells exposed to ionizing radiation (0, 1, 2, 4, or 8 Gy) showed increasing rates of percentage of DNA in the tail and tail length with increasing radiation dose. When cells were exposed to room air (normoxia) for 1 day and 95% O2 (hyperoxia) for 1, 2, 3, 4, and 5 days, data indicated that hyperoxia caused time-dependent increases in levels of (a) single strand breaks, (b) double strand breaks, and (c) 8-oxoguanine. Decreased DNA methylation also was observed at day 5 of hyperoxic exposure, suggesting that hyperoxia-induced DNA damage can influence patterns of DNA methylation in a lung-derived cell line.


Assuntos
Dano ao DNA , Metilação de DNA/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Guanina/análogos & derivados , Hiperóxia/fisiopatologia , Oxigênio/toxicidade , Linhagem Celular Tumoral , Ensaio Cometa/métodos , DNA/análise , DNA/efeitos dos fármacos , DNA/efeitos da radiação , Metilação de DNA/efeitos da radiação , Reparo do DNA , DNA de Cadeia Simples/efeitos dos fármacos , DNA de Cadeia Simples/efeitos da radiação , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Guanina/metabolismo , Humanos , Neoplasias Pulmonares , Fatores de Tempo
20.
PLoS One ; 6(11): e27451, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22096575

RESUMO

Calcium mobilization can regulate a wide range of essential functions of respiratory epithelium, including ion transport, ciliary beat frequency, and secretion of mucus, all of which are modified in cystic fibrosis (CF). SERCA2, an important controller of calcium signaling, is deficient in CF epithelium. We conducted this study to determine whether SERCA2 deficiency can modulate airway epithelial responses to environmental oxidants such as ozone. This could contribute to the pathogenesis of pulmonary exacerbations, which are important and frequent clinical events in CF. To address this, we used air-liquid interface (ALI) cultures of non-CF and CF cell lines, as well as differentiated cultures of cells derived from non-CF and CF patients. We found that ozone exposure caused enhanced membrane damage, mitochondrial dysfunction and apoptotic cell death in CF airway epithelial cell lines relative to non-CF. Ozone exposure caused increased proinflammatory cytokine production in CF airway epithelial cell lines. Elevated proinflammatory cytokine production also was observed in shRNA-mediated SERCA2 knockdown cells. Overexpression of SERCA2 reversed ozone-induced proinflammatory cytokine production. Ozone-induced proinflammatory cytokine production was NF-κB- dependent. In a stable NF-κB reporter cell line, SERCA2 inhibition and knockdown both upregulated cytomix-induced NF-κB activity, indicating importance of SERCA2 in modulating NF-κB activity. In this system, increased NF-κB activity was also accompanied by increased IL-8 production. Ozone also induced NF-κB activity and IL-8 release, an effect that was greater in SERCA2-silenced NF-κB-reporter cells. SERCA2 overexpression reversed cytomix-induced increased IL-8 release and total nuclear p65 in CFTR-deficient (16HBE-AS) cells. These studies suggest that SERCA2 is an important regulator of the proinflammatory response of airway epithelial cells and could be a potential therapeutic target.


Assuntos
Fibrose Cística/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Ozônio/toxicidade , Western Blotting , Células Cultivadas , Imunofluorescência , Humanos , Interleucina-8/metabolismo , Pulmão/citologia , Pulmão/patologia , NF-kappa B/metabolismo , RNA Interferente Pequeno , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA