Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
J Neurol Sci ; 318(1-2): 146-54, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22537900

ABSTRACT

Blast-induced neurotrauma (BINT) is a major medical concern yet its etiology is largely undefined. Complement activation may play a role in the development of secondary injury following traumatic brain injury; however, its role in BINT is still undefined. The present study was designed to characterize the complement system and adaptive immune-inflammatory responses in a rat model of moderate BINT. Anesthetized rats were exposed to a moderate blast (120 kPa) using an air-driven shock tube. Brain tissue injury, systemic and local complement, cerebral edema, inflammatory cell infiltration, and pro-inflammatory cytokine production were measured at 0.5, 3, 48, 72, 120, and 168 h. Injury to brain tissue was evaluated by histological evaluation. Systemic complement was measured via ELSIA. The remaining measurements were determined by immunohistoflourescent staining. Moderate blast triggers moderate brain injuries, elevated levels of local brain C3/C5b-9 and systemic C5b-9, increased leukocyte infiltration, unregulated tumor necrosis factor alpha (TNFα), and aquaporin-4 in rat brain cortex at 3- and 48-hour post blast. Early immune-inflammatory response to BINT involves complement and TNFα, which correlates with hippocampus and cerebral cortex damage. Complement and TNFα activation may be a novel therapeutic target for reducing the damaging effects of BINT inflammation.


Subject(s)
Blast Injuries/physiopathology , Brain Injuries/physiopathology , Complement Activation/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Blast Injuries/immunology , Blast Injuries/pathology , Brain/immunology , Brain/metabolism , Brain/physiopathology , Brain Injuries/immunology , Brain Injuries/pathology , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Time Factors
2.
J Neurotrauma ; 28(11): 2307-18, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22082449

ABSTRACT

A high incidence of blast exposure is a 21st century reality in counter-insurgency warfare. However, thresholds for closed-head blast-induced traumatic brain injury (bTBI) remain unknown. Moreover, without objective information about relative blast exposure, warfighters with bTBI may not receive appropriate medical care and may remain in harm's way. Accordingly, we have engineered a blast injury dosimeter (BID) using a photonic crystalline material that changes color following blast exposure. The photonic crystals are fabricated using SU-8 via multi-beam interference laser lithography. The final BID is similar in appearance to an array of small colored stickers that may be affixed to uniforms or helmets in multiple locations. Although durable under normal conditions, the photonic crystalline micro- and nano-structure are precisely altered by blast to create a color change. These BIDs were evaluated using a rat model of bTBI, for which blast shockwave exposure was generated via a compressed air-driven shock tube. With prototype BID arrays affixed to the animals, we found that BID color changes corresponded with subtle brain pathologies, including neuronal degeneration and reactive astrocytosis. These subtle changes were most notable in the dentate gyrus of the hippocampus, cerebral cortex, and cerebellum. These data demonstrate the feasibility of using a materials-based, power-free colorimetric BID as the first self-contained blast sensor calibrated to correspond with brain pathology.


Subject(s)
Blast Injuries/pathology , Brain Injuries/pathology , Optical Phenomena , Animals , Blast Injuries/complications , Brain Diseases/etiology , Brain Diseases/pathology , Brain Injuries/etiology , Colorimetry/methods , Crystallization , Rats , Rats, Sprague-Dawley
3.
J Neurosci Methods ; 195(1): 61-6, 2011 Jan 30.
Article in English | MEDLINE | ID: mdl-21129403

ABSTRACT

Exposure to a blast wave generated during an explosion may result in brain damage and related neurological impairments. Several mechanisms by which the primary blast wave can damage the brain have been proposed, including: (1) a direct effect of the shock wave on the brain causing tissue damage by skull flexure and propagation of stress and shear forces; and (2) an indirect transfer of kinetic energy from the blast, through large blood vessels and cerebrospinal fluid (CSF), to the central nervous system. To address a basic question related to the mechanisms of blast brain injury, pressure was measured inside the brains of rats exposed to a low level of blast (~35kPa), while positioned in three different orientations with respect to the primary blast wave; head facing blast, right side exposed to blast and head facing away from blast. Data show different patterns and durations of the pressure traces inside the brain, depending on the rat orientation to blast. Frontal exposures (head facing blast) resulted in pressure traces of higher amplitude and longer duration, suggesting direct transmission and reflection of the pressure inside the brain (dynamic pressure transfer). The pattern of the pressure wave inside the brain in the head facing away from blast exposures assumes contribution of the static pressure, similar to hydrodynamic pressure to the pressure wave inside the brain.


Subject(s)
Blast Injuries/complications , Brain Injuries/etiology , High-Energy Shock Waves/adverse effects , Pressure/adverse effects , Animals , Blast Injuries/physiopathology , Brain Injuries/physiopathology , Male , Posture , Rats , Rats, Sprague-Dawley
4.
J Neurosci Res ; 88(16): 3530-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20882564

ABSTRACT

Traumatic brain injury (TBI) as a consequence of exposure to blast is increasingly prevalent in military populations, with the underlying pathophysiological mechanisms mostly unknown. In the present study, we utilized an air-driven shock tube to investigate the effects of blast exposure (120 kPa) on rat brains. Immediately following exposure to blast, neurological function was reduced. BBB permeability was measured using IgG antibody and evaluating its immunoreactivity in the brain. At 3 and 24 hr postexposure, there was a transient significant increase in IgG staining in the cortex. At 3 days postexposure, IgG immunoreactivity returned to control levels. Quantitative immunostaining was employed to determine the temporal course of brain oxidative stress following exposure to blast. Levels of 4-hydroxynonenal (4-HNE) and 3-nitrotyrosine (3-NT) were significantly increased at 3 hr postexposure and returned to control levels at 24 hr postexposure. The response of microglia to blast exposure was determined by autoradiographic localization of (3) H-PK11195 binding. At 5 days postexposure, increased binding was observed in the contralateral and ipsilateral dentate gyrus. These regions also displayed increased binding at 10 days postexposure; in addition to these regions there was increased binding in the contralateral ventral hippocampus and substantia nigra at this time point. By using antibodies against CD11b/c, microglia morphology characteristic of activated microglia was observed in the hippocampus and substantia nigra of animals exposed to blast. These results indicate that BBB breakdown, oxidative stress, and microglia activation likely play a role in the neuropathology associated with TBI as a result of blast exposure.


Subject(s)
Blast Injuries/pathology , Blood-Brain Barrier/metabolism , Brain Injuries/pathology , Microglia/immunology , Analysis of Variance , Animals , Blast Injuries/complications , Blast Injuries/immunology , Blast Injuries/metabolism , Blood-Brain Barrier/physiopathology , Brain Injuries/etiology , Brain Injuries/immunology , Brain Injuries/metabolism , Disease Models, Animal , Glasgow Coma Scale , Hippocampus/immunology , Hippocampus/pathology , Male , Microglia/metabolism , Oxidative Stress/immunology , Permeability , Random Allocation , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Substantia Nigra/immunology , Substantia Nigra/pathology
5.
Shock ; 32(3): 325-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19174737

ABSTRACT

Lung contusion is a common problem from blunt chest trauma caused by mechanical forces and by exposure to blast overpressure, often with fatal consequences. Lung contusion is also a risk factor for the development of pneumonia, severe clinical acute lung injury (ALI), and acute respiratory distress syndrome (ARDS). Infiltrating neutrophils are considered to be central mediators of lung injuries after blunt trauma. Recent studies have demonstrated that antioxidants reduced pulmonary inflammation in different models of lung damage. This study examined the effect of antioxidant N-acetylcysteine amide (NACA) on the progression of lung inflammation after exposure to a moderate level of blast overpressure (140 kPa). Rats were administered with NACA (i.p. 100 mg/kg) or placebo (PBS) 30, 60 min and 24 h after exposure. Nonblasted sham-injected animals served as controls. Neutrophil infiltration measured by myeloperoxidase (MPO) activity in the lung was significantly increased at 2 days after blast and returned to controls at 8 days. This increase corresponded with activation of integrin CD11b mRNA and lung inflammatory chemokine mRNA expression; macrophage inflammatory protein-1 (MIP-1), monocyte chemotactic peptide-1 (MCP-1), and cytokine-induced neutrophil chemoattractant-1 (CINC-1). At 8 days, all inflammatory mediators returned to control levels. In addition, expression of heme oxygenase-1 (HO-1) mRNA increased at 2 days after exposure. No changes were detected in the lung manganase superoxide dismutase (MnSOD) or glutathione reductase (GR) mRNA expression after blast. N-Acetylcysteine amide significantly reduced infiltration of neutrophils and CD11b mRNA activation in lungs, and completely blocked activation of MIP-1, MCP-1 and CINC-1 mRNA. The relatively higher inhibition of chemokine mRNAs compared with reduction in MPO activity and CD11b is in accordance with an antioxidant effect of NACA on reactive oxygen species (ROS) accumulation, rather than by an effect on neutrophil sequestration. The inhibition of HO-1 mRNA activation after blast was likely also related to the drug antioxidant effect.


Subject(s)
Acetylcysteine/analogs & derivatives , Antioxidants/therapeutic use , Lung Injury/drug therapy , Lung Injury/immunology , Pneumonia/drug therapy , Pneumonia/immunology , Acetylcysteine/therapeutic use , Animals , CD11b Antigen/genetics , Chemokine CCL2/genetics , Chemokine CXCL1/genetics , Glutathione Reductase/genetics , Heme Oxygenase-1/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Macrophage Inflammatory Proteins/genetics , Male , Neutrophil Infiltration/immunology , Neutrophil Infiltration/physiology , Peroxidase/metabolism , Pneumonia/metabolism , Pneumonia/pathology , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL