Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Respir Cell Mol Biol ; 58(6): 696-705, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29314868

RESUMO

Inhalation of powerful chemical agents, such as sulfur mustard (SM), can have debilitating pulmonary consequences, such as bronchiolitis obliterans (BO) and parenchymal fibrosis (PF). The underlying pathogenesis of disorders after SM inhalation is not clearly understood, resulting in a paucity of effective therapies. In this study, we evaluated the role of profibrotic pathways involving transforming growth factor-ß (TGF-ß) and platelet-derived growth factor (PDGF) in the development of BO and PF after SM inhalation injury using a rat model. Adult Sprague-Dawley rats were intubated and exposed to SM (1.0 mg/kg), then monitored daily for respiratory distress, oxygen saturation changes, and weight loss. Rats were killed at 7, 14, 21, or 28 days, and markers of injury were determined by histopathology; pulmonary function testing; and assessment of TGF-ß, PDGF, and PAI-1 concentrations. Respiratory distress developed over time after SM inhalation, with progressive hypoxemia, respiratory distress, and weight loss. Histopathology confirmed the presence of both BO and PF, and both gradually worsened with time. Pulmonary function testing demonstrated a time-dependent increase in lung resistance, as well as a decrease in lung compliance. Concentrations of TGF-ß, PDGF, and PAI-1 were elevated at 28 days in lung, BAL fluid, and/or plasma. Time-dependent development of BO and PF occurs in lungs of rats exposed to SM inhalation, and the elevated concentrations of TGF-ß, PDGF, and PAI-1 suggest involvement of these profibrotic pathways in the aberrant remodeling after injury.


Assuntos
Bronquiolite Obliterante/induzido quimicamente , Gás de Mostarda/administração & dosagem , Gás de Mostarda/toxicidade , Fibrose Pulmonar/induzido quimicamente , Administração por Inalação , Animais , Bronquiolite Obliterante/metabolismo , Bronquiolite Obliterante/mortalidade , Bronquiolite Obliterante/patologia , Líquido da Lavagem Broncoalveolar , Substâncias para a Guerra Química/toxicidade , Relação Dose-Resposta a Droga , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/mortalidade , Ratos Sprague-Dawley , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Testes de Função Respiratória , Fator de Crescimento Transformador beta1/metabolismo , Redução de Peso/efeitos dos fármacos
2.
Int J Toxicol ; 33(4): 271-281, 2014 07.
Artigo em Inglês | MEDLINE | ID: mdl-24801489

RESUMO

Mustard gas (sulfur mustard [SM], bis-[2-chloroethyl] sulfide) is a vesicating chemical warfare agent and a potential chemical terrorism agent. Exposure of SM causes debilitating skin blisters (vesication) and injury to the eyes and the respiratory tract; of these, the respiratory injury, if severe, may even be fatal. Therefore, developing an effective therapeutic strategy to protect against SM-induced respiratory injury is an urgent priority of not only the US military but also the civilian antiterrorism agencies, for example, the Homeland Security. Toward developing a respiratory medical countermeasure for SM, four different classes of therapeutic compounds have been evaluated in the past: anti-inflammatory compounds, antioxidants, protease inhibitors and antiapoptotic compounds. This review examines all of these different options; however, it suggests that preventing cell death by inhibiting apoptosis seems to be a compelling strategy but possibly dependent on adjunct therapies using the other drugs, that is, anti-inflammatory, antioxidant, and protease inhibitor compounds.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Antioxidantes/uso terapêutico , Substâncias para a Guerra Química/toxicidade , Intoxicação por Gás/tratamento farmacológico , Modelos Biológicos , Gás de Mostarda/toxicidade , Inibidores de Proteases/uso terapêutico , Animais , Antídotos/uso terapêutico , Apoptose/efeitos dos fármacos , Quimioterapia Combinada , Intoxicação por Gás/imunologia , Intoxicação por Gás/metabolismo , Intoxicação por Gás/patologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
3.
J Pharmacol Exp Ther ; 344(1): 308-16, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23129783

RESUMO

Sulfur mustard (SM) is a vesicant chemical warfare and terrorism agent. Besides skin and eye injury, respiratory damage has been mainly responsible for morbidity and mortality after SM exposure. Previously, it was shown that suppressing the death receptor (DR) response by the dominant-negative Fas-associated death domain protein prior to SM exposure blocked apoptosis and microvesication in skin. Here, we studied whether antagonizing the Fas receptor (FasR) pathway by small-interfering RNA (siRNA) applied after SM exposure would prevent apoptosis and, thus, airway injury. Normal human bronchial/tracheal epithelial (NHBE) cells were used as an in vitro model with FasR siRNA, FasR agonistic antibody CH11, and FasR antagonistic antibody ZB4 as investigative tools. In NHBE cells, both SM (300 µM) and CH11 (100 ng/ml) induced caspase-3 activation, which was inhibited by FasR siRNA and ZB4, indicating that SM-induced apoptosis was via the Fas response. FasR siRNA inhibited SM-induced caspase-3 activation when added to NHBE cultures up to 8 hours after SM. Results using annexin V/propidium iodide-stained cells showed that both apoptosis and necrosis were involved in cell death due to SM; FasR siRNA decreased both apoptotic and necrotic cell populations. Bronchoalveolar lavage fluid (BALF) of rats exposed to SM (1 mg/kg, 50 minutes) revealed a significant (P < 0.05) increase in soluble Fas ligand and active caspase-3 in BALF cells. These findings suggest an intervention of Fas-mediated apoptosis as a postexposure therapeutic strategy with a therapeutic window for SM inhalation injury and possibly other respiratory diseases involving the Fas response.


Assuntos
Apoptose/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Células Epiteliais/efeitos dos fármacos , Gás de Mostarda/toxicidade , RNA Interferente Pequeno/farmacologia , Receptor fas/antagonistas & inibidores , Receptor fas/genética , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Queimaduras por Inalação/tratamento farmacológico , Caspase 3/metabolismo , Linhagem Celular , Células Cultivadas , Ativação Enzimática/fisiologia , Ensaio de Imunoadsorção Enzimática , Proteína Ligante Fas/análise , Proteína Ligante Fas/metabolismo , Citometria de Fluxo , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Sistema Respiratório/citologia , Sistema Respiratório/efeitos dos fármacos , Transfecção
4.
Toxicol Pathol ; 39(7): 1056-64, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21934141

RESUMO

Macrolide antibiotics have been shown to protect airway epithelial cells and macrophages from sulfur mustard (SM)-induced cytotoxicity. In the current study, the efficacy of roxithromycin in ameliorating SM-induced respiratory injury was further evaluated in a rat model. Anesthetized rats (N = 8/group) were intratracheally exposed to SM by vapor inhalation. For the drug treatment groups, rats were orally given 10, 20, or 40 mg/kg roxithromycin one hr prior to exposure and every twenty-four hr thereafter. After one, three, or seven days of treatment, sections of the lung were examined and scored for histopathological parameters. Treatment with roxithromycin ameliorated many of the symptoms caused by SM in some animals. In particular, treatment at 40 mg/kg for three days showed significant improvements (p < .05) over the untreated group. When the evaluation was focused on trachea, treatment with roxithromycin for three days showed a trend of dose-dependent protection; moreover, the groups treated with 20 or 40 mg/kg of roxithromycin were statistically different (p < .001 and p < .05, respectively) from the untreated group. These results suggest that roxithromycin protects against some damages associated with SM injury in the lung, particularly in the upper respiratory tract.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Gás de Mostarda/toxicidade , Substâncias Protetoras/farmacologia , Roxitromicina/farmacologia , Animais , Brônquios/química , Brônquios/efeitos dos fármacos , Brônquios/patologia , Modelos Animais de Doenças , Histocitoquímica , Exposição por Inalação , Masculino , Gás de Mostarda/administração & dosagem , Alvéolos Pulmonares/química , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Ratos , Projetos de Pesquisa , Traqueia/química , Traqueia/efeitos dos fármacos , Traqueia/patologia
5.
Toxicol Appl Pharmacol ; 248(2): 89-99, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20659490

RESUMO

Inhalation of sulfur mustard (SM), a bifunctional alkylating agent that causes severe lung damage, is a significant threat to both military and civilian populations. The mechanisms mediating its cytotoxic effects are unknown and were investigated in the present studies. Male rats Crl:CD(SD) were anesthetized, and then intratracheally intubated and exposed to 0.7-1.4mg/kg SM by vapor inhalation. Animals were euthanized 6, 24, 48h or 7days post-exposure and bronchoalveolar lavage fluid (BAL) and lung tissue collected. Exposure of rats to SM resulted in rapid pulmonary toxicity, including focal ulceration and detachment of the trachea and bronchial epithelia from underlying mucosa, thickening of alveolar septal walls and increased numbers of inflammatory cells in the tissue. There was also evidence of autophagy and apoptosis in the tissue. This was correlated with increased BAL protein content, a marker of injury to the alveolar epithelial lining. SM exposure also resulted in increased expression of markers of inflammation including cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNFα), inducible nitric oxide synthase (iNOS), and matrix metalloproteinase-9 (MMP-9), each of which has been implicated in pulmonary toxicity. Whereas COX-2, TNFα and iNOS were mainly localized in alveolar regions, MMP-9 was prominent in bronchial epithelium. In contrast, expression of the anti-oxidant hemeoxygenase, and the anti-inflammatory collectin, surfactant protein-D, decreased in the lung after SM exposure. These data demonstrate that SM-induced oxidative stress and injury are associated with the generation of cytotoxic inflammatory proteins which may contribute to the pathogenic response to this vesicant.


Assuntos
Pulmão/efeitos dos fármacos , Gás de Mostarda/toxicidade , Pneumonia/induzido quimicamente , Animais , Antioxidantes/metabolismo , Apoptose , Autofagia , Líquido da Lavagem Broncoalveolar , Substâncias para a Guerra Química , Pulmão/enzimologia , Pulmão/metabolismo , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Pneumonia/enzimologia , Pneumonia/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
6.
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
7.
Mil Med ; 171(5): 448-53, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16761898

RESUMO

Although best known as a blistering agent, sulfur mustard (HD) can also induce neutropenia in exposed individuals, increasing their susceptibility to infection. Granulocyte colony-stimulating factor (G-CSF) and pegylated G-CSF (peg-G-CSF) have been approved by the U.S. Food and Drug Administration as hematopoietic growth factors to treat chemotherapy-induced neutropenia. The goal of this study was to determine the effectiveness of G-CSF and peg-G-CSF in ameliorating HD-induced neutropenia. African green monkeys (Chlorocebus aethiops) were challenged with HD and, at 1, 3, 5, or 7 days after exposure, G-CSF therapy (10 microg/kg per day for 21 days) was initiated. Peg-G-CSF (300 microg/kg, single treatment) was similarly tested, with treatment given at 3 days after exposure. Untreated HD-exposed animals recovered from neutropenia 28 days after exposure, whereas G-CSF- or peg-G-CSF-treated animals recovered 8 to 19 days after exposure (p < 0.05). These results indicate that G-CSF or peg-G-CSF may provide Food and Drug Administration-approved treatments that will reduce the duration of HD-induced neutropenia.


Assuntos
Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Gás de Mostarda/efeitos adversos , Neutropenia/induzido quimicamente , Animais , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Haplorrinos , Masculino , Estados Unidos
8.
Toxicol Lett ; 244: 8-20, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26562770

RESUMO

Toxic industrial chemicals are used throughout the world to produce everyday products such as household and commercial cleaners, disinfectants, pesticides, pharmaceuticals, plastics, paper, and fertilizers. These chemicals are produced, stored, and transported in large quantities, which poses a threat to the local civilian population in cases of accidental or intentional release. Several of these chemicals have no known medical countermeasures for their toxic effects. Phosgene is a highly toxic industrial chemical which was used as a chemical warfare agent in WWI. Exposure to phosgene causes latent, non-cardiogenic pulmonary edema which can result in respiratory failure and death. The mechanisms of phosgene-induced pulmonary injury are not fully identified, and currently there is no efficacious countermeasure. Here, we provide a proposed mechanism of phosgene-induced lung injury based on the literature and from studies conducted in our lab, as well as provide results from studies designed to evaluate survival efficacy of potential therapies following whole-body phosgene exposure in mice. Several therapies were able to significantly increase 24h survival following an LCt50-70 exposure to phosgene; however, no treatment was able to fully protect against phosgene-induced mortality. These studies provide evidence that mortality following phosgene toxicity can be mitigated by neuro- and calcium-regulators, antioxidants, phosphodiesterase and endothelin receptor antagonists, angiotensin converting enzymes, and transient receptor potential cation channel inhibitors. However, because the mechanism of phosgene toxicity is multifaceted, we conclude that a single therapeutic is unlikely to be sufficient to ameliorate the multitude of direct and secondary toxic effects caused by phosgene inhalation.


Assuntos
Antídotos/uso terapêutico , Substâncias para a Guerra Química , Lesão Pulmonar/tratamento farmacológico , Pulmão/efeitos dos fármacos , Fosgênio , Animais , Modelos Animais de Doenças , Exposição por Inalação , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/diagnóstico , Lesão Pulmonar/metabolismo , Lesão Pulmonar/fisiopatologia , Masculino , Camundongos , Terapia de Alvo Molecular , Transdução de Sinais/efeitos dos fármacos
9.
Toxicol Sci ; 154(2): 341-353, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27605419

RESUMO

Sulfur mustard (bis 2-chloroethyl ethyl sulfide, SM) is a powerful bi-functional vesicating chemical warfare agent. SM tissue injury is partially mediated by the overproduction of reactive oxygen species resulting in oxidative stress. We hypothesized that using a catalytic antioxidant (AEOL 10150) to alleviate oxidative stress and secondary inflammation following exposure to SM would attenuate the toxic effects of SM inhalation. Adult male rats were intubated and exposed to SM (1.4 mg/kg), a dose that produces an LD50 at approximately 24 h. Rats were randomized and treated via subcutaneous injection with either sterile PBS or AEOL 10150 (5 mg/kg, sc, every 4 h) beginning 1 h post-SM exposure. Rats were euthanized between 6 and 48 h after exposure to SM and survival and markers of injury were determined. Catalytic antioxidant treatment improved survival after SM inhalation in a dose-dependent manner, up to 52% over SM PBS at 48 h post-exposure. This improvement was sustained for at least 72 h after SM exposure when treatments were stopped after 48 h. Non-invasive monitoring throughout the duration of the studies also revealed blood oxygen saturations were improved by 10% and clinical scores were reduced by 57% after SM exposure in the catalytic antioxidant treatment group. Tissue analysis showed catalytic antioxidant therapy was able to decrease airway cast formation by 69% at 48 h post-exposure. To investigate antioxidant induced changes at the peak of injury, several biomarkers of oxidative stress and inflammation were evaluated at 24 h post-exposure. AEOL 10150 attenuated SM-mediated lung lipid oxidation, nitrosative stress and many proinflammatory cytokines. The findings indicate that catalytic antioxidants may be useful medical countermeasure against inhaled SM exposure.


Assuntos
Antídotos/farmacologia , Antioxidantes/farmacologia , Substâncias para a Guerra Química/toxicidade , Lesão Pulmonar/prevenção & controle , Pulmão/efeitos dos fármacos , Metaloporfirinas/farmacologia , Gás de Mostarda/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Pneumonia/prevenção & controle , Animais , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Mediadores da Inflamação/metabolismo , Exposição por Inalação , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Masculino , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Pneumonia/patologia , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
10.
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
11.
Toxicology ; 263(1): 41-6, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18852015

RESUMO

Sulfur mustard (SM)-induced lung injury has been associated with protease activation, oxidative injury and inflammatory response culminating in tissue necrosis. The protease inhibitors aprotinin and ilomastat and the antioxidant trolox were evaluated for efficacy in ameliorating SM-induced lung injury. Anesthetized spontaneously breathing rats (N=6-8/group) were intratracheally intubated and exposed to 1.4 mg/kg SM (0.35 mg SM in 0.1 ml of ethanol) or ethanol alone by vapor inhalation for 50 min. At 1 min before the exposure rats were treated with one of the following: intravenous aprotinin, 4.4 mg/kg; intraperitoneal (ip) ilomastat, 25mg/kg; or ip trolox, 500 microg/kg. Aprotinin-treated animals received supplemental 2.2mg/kg doses at 1 min and 6h post-exposure (PE). A whole body plethysmograph system was used to monitor pulmonary function (PF) parameters for 1h before exposure (baseline), and from 5-6 and 23-24h post-exposure. SM inhalation caused significant increases in several PF parameters, including tidal volume, peak inspiratory flow, peak expiratory flow, end expiratory pause and enhanced pause. Consistent with the reported development of SM-induced pathology, these changes were minimal at the 5-6-h time and significant at the 23-24-h timepoint. At the later time it is known from previous work that airways are becoming obstructed with loose cellular debris, damaged cells and exudate, which contributed to the changes in PF parameters. Treatment with aprotinin or ilomastat eliminated these PF changes, yielding results comparable with controls for each of these parameters. Lung lavage fluid analysis showed that SM caused a significant increase in total protein (TP) and in the cytokines IL-1alpha and IL-13. Aprotinin treatment prevented the increases in TP and IL-1alpha production, ilomastat prevented the increased production of IL-13, and trolox treatment did not significantly prevent the SM-related increases in TP, IL-1alpha or IL-13. Histopathologic examination of lung tissue 24h post-exposure showed minimal alveolar effects caused by SM, while damage to bronchiolar regions was much more severe due to the highly reactive nature of SM. While aprotinin and ilomastat both alleviated the PF perturbations, surprisingly only aprotinin reduced the observed pathology, both grossly and histologically. These early results indicate that treatment with aprotinin and to a lesser extent ilomastat reduces some of the direct inflammatory response and damage associated with SM-induced lung injury. This research was supported by the Defense Threat Reduction Agency - Joint Science and Technology Office, Medical S&T Division.


Assuntos
Antioxidantes/farmacologia , Aprotinina/farmacologia , Substâncias para a Guerra Química/toxicidade , Cromanos/farmacologia , Indóis/farmacologia , Pneumopatias/prevenção & controle , Gás de Mostarda/toxicidade , Inibidores de Serina Proteinase/farmacologia , Animais , Líquido da Lavagem Broncoalveolar/química , Citocinas/análise , Quimioterapia Combinada , Ácidos Hidroxâmicos , Injeções Intraperitoneais , Injeções Intravenosas , Intubação Intratraqueal , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/fisiopatologia , Pneumopatias/induzido quimicamente , Pneumopatias/fisiopatologia , Masculino , Proteínas/análise , Ratos , Ratos Sprague-Dawley , Testes de Função Respiratória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA