Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Cell Mol Biol ; 58(6): 696-705, 2018 06.
Article in English | MEDLINE | ID: mdl-29314868

ABSTRACT

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.


Subject(s)
Bronchiolitis Obliterans/chemically induced , Mustard Gas/administration & dosage , Mustard Gas/toxicity , Pulmonary Fibrosis/chemically induced , Administration, Inhalation , Animals , Bronchiolitis Obliterans/metabolism , Bronchiolitis Obliterans/mortality , Bronchiolitis Obliterans/pathology , Bronchoalveolar Lavage Fluid , Chemical Warfare Agents/toxicity , Dose-Response Relationship, Drug , Plasminogen Activator Inhibitor 1/metabolism , Platelet-Derived Growth Factor/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/mortality , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Respiratory Function Tests , Transforming Growth Factor beta1/metabolism , Weight Loss/drug effects
2.
Toxicol Sci ; 159(2): 461-469, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28962529

ABSTRACT

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.


Subject(s)
Arterioles/drug effects , Chemical Warfare Agents/toxicity , Lung/drug effects , Mustard Gas/toxicity , Thrombosis/chemically induced , Animals , Arterioles/pathology , Computed Tomography Angiography , Fibrin Fibrinogen Degradation Products/metabolism , Inhalation Exposure , Lung/blood supply , Lung Diseases/chemically induced , Male , Mustard Gas/administration & dosage , Platelet Aggregation/drug effects , Rats , Rats, Sprague-Dawley
3.
Toxicol Sci ; 154(2): 341-353, 2016 12.
Article in English | MEDLINE | ID: mdl-27605419

ABSTRACT

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.


Subject(s)
Antidotes/pharmacology , Antioxidants/pharmacology , Chemical Warfare Agents/toxicity , Lung Injury/prevention & control , Lung/drug effects , Metalloporphyrins/pharmacology , Mustard Gas/toxicity , Oxidative Stress/drug effects , Pneumonia/prevention & control , Animals , Cytokines/metabolism , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Inhalation Exposure , Lung/metabolism , Lung/pathology , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Male , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Time Factors
4.
Toxicol Lett ; 244: 21-27, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26562769

ABSTRACT

Phosgene (CG), a toxic inhalation and industrial hazard, causes bronchoconstriction, vasoconstriction and associated pathological effects that could be life threatening. Ion channels of the transient receptor potential (TRP) family have been identified to act as specific chemosensory molecules in the respiratory tract in the detection, control of adaptive responses and initiation of detrimental signaling cascades upon exposure to various toxic inhalation hazards (TIH); their activation due to TIH exposure may result in broncho- and vasoconstriction. We studied changes in the regulation of intracellular free Ca(2+) concentration ([Ca(2+)]i) in cultures of human bronchial smooth muscle cells (BSMC) and human pulmonary microvascular endothelial cells (HPMEC) exposed to CG (16ppm, 8min), using an air/liquid interface exposure system. CG increased [Ca(2+)]i (p<0.05) in both cell types, The CG-induced [Ca(2+)]i was blocked (p<0.05) by two types of TRP channel blockers, SKF-96365, a general TRP channel blocker, and RR, a general TRPV (vanilloid type) blocker, in both BSMC and HPMEC. These effects correlate with the in vivo efficacies of these compounds to protect against lung injury and 24h lethality from whole body CG inhalation exposure in mice (8-10ppm×20min). Thus the TRP channel mechanism appears to be a potential target for intervention in CG toxicity.


Subject(s)
Antidotes/pharmacology , Bronchi/drug effects , Chemical Warfare Agents/toxicity , Endothelial Cells/drug effects , Membrane Transport Modulators/pharmacology , Myocytes, Smooth Muscle/drug effects , Phosgene/toxicity , Transient Receptor Potential Channels/antagonists & inhibitors , Animals , Bronchi/metabolism , Bronchi/pathology , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Imidazoles/pharmacology , Inhalation Exposure , Male , Mice , Molecular Targeted Therapy , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/metabolism
5.
Toxicol Lett ; 244: 8-20, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26562770

ABSTRACT

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.


Subject(s)
Antidotes/therapeutic use , Chemical Warfare Agents , Lung Injury/drug therapy , Lung/drug effects , Phosgene , Animals , Disease Models, Animal , Inhalation Exposure , Lung/metabolism , Lung/pathology , Lung/physiopathology , Lung Injury/chemically induced , Lung Injury/diagnosis , Lung Injury/metabolism , Lung Injury/physiopathology , Male , Mice , Molecular Targeted Therapy , Signal Transduction/drug effects
6.
Toxicol Sci ; 143(1): 178-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25331496

ABSTRACT

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.


Subject(s)
Airway Obstruction/drug therapy , Chemical Warfare Agents , Fibrinolytic Agents/administration & dosage , Inhalation Exposure , Lung/drug effects , Mustard Gas , Respiratory Insufficiency/prevention & control , Thrombolytic Therapy , Tissue Plasminogen Activator/administration & dosage , Acidosis/chemically induced , Acidosis/prevention & control , Administration, Inhalation , Airway Obstruction/chemically induced , Airway Obstruction/pathology , Airway Obstruction/physiopathology , Animals , Disease Models, Animal , Drug Administration Schedule , Lung/pathology , Lung/physiopathology , Male , Oxygen/blood , Pulmonary Ventilation/drug effects , Rats, Sprague-Dawley , Respiration/drug effects , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/pathology , Respiratory Insufficiency/physiopathology , Time Factors
7.
Int J Toxicol ; 33(4): 271-281, 2014 07.
Article in English | MEDLINE | ID: mdl-24801489

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , Chemical Warfare Agents/toxicity , Gas Poisoning/drug therapy , Models, Biological , Mustard Gas/toxicity , Protease Inhibitors/therapeutic use , Animals , Antidotes/therapeutic use , Apoptosis/drug effects , Drug Therapy, Combination , Gas Poisoning/immunology , Gas Poisoning/metabolism , Gas Poisoning/pathology , Humans , Lung/drug effects , Lung/immunology , Lung/metabolism , Lung/pathology , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
8.
J Pharmacol Exp Ther ; 344(1): 308-16, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23129783

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Chemical Warfare Agents/toxicity , Epithelial Cells/drug effects , Mustard Gas/toxicity , RNA, Small Interfering/pharmacology , fas Receptor/antagonists & inhibitors , fas Receptor/genetics , Animals , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Burns, Inhalation/drug therapy , Caspase 3/metabolism , Cell Line , Cells, Cultured , Enzyme Activation/physiology , Enzyme-Linked Immunosorbent Assay , Fas Ligand Protein/analysis , Fas Ligand Protein/metabolism , Flow Cytometry , Humans , Male , Rats , Rats, Sprague-Dawley , Respiratory System/cytology , Respiratory System/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...