Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Front Neurol ; 10: 650, 2019.
Article in English | MEDLINE | ID: mdl-31297080

ABSTRACT

Traumatic brain injury resulting from exposure to blast overpressure (BOP) is associated with neuropathology including impairment of the blood-brain barrier (BBB). This study examined the effects of repeated exposure to primary BOP and post-blast treatment with an antioxidant, N-acetylcysteine amide (NACA) on the integrity of BBB. Anesthetized rats were exposed to three 110 kPa BOPs separated by 0.5 h. BBB integrity was examined in vivo via a cranial window allowing imaging of pial microcirculation by intravital microscopy. Tetramethylrhodamine isothiocyanate Dextran (TRITC-Dextran, mw = 40 kDa or 150 kDa) was injected intravenously 2.5 h after the first BOP exposure and the leakage of TRITC-Dextran from pial microvessels into the brain parenchyma was assessed. The animals were randomized into 6 groups (n = 5/group): four groups received 40 kDa TRITC-Dextran (BOP-40, sham-40, BOP-40 NACA, and sham-40 NACA), and two groups received 150 kDa TRITC-Dextran (BOP-150 and sham-150). NACA treated groups were administered NACA 2 h after the first BOP exposure. The rate of TRITC-Dextran leakage was significantly higher in BOP-40 than in sham-40 group. NACA treatment significantly reduced TRITC-Dextran leakage in BOP-40 NACA group and sham-40 NACA group presented the least amount of leakage. The rate of leakage in BOP-150 and sham-150 groups was comparable to sham-40 NACA and thus these groups were not assessed for the effects of NACA. Collectively, these data suggest that BBB integrity is compromised following BOP exposure and that NACA treatment at a single dose may significantly protect against blast-induced BBB breakdown.

2.
Mil Med ; 184(Suppl 1): 265-272, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30901455

ABSTRACT

OBJECTIVE: Mild blast traumatic brain injury is commonly prevalent in modern combat casualty care and has been associated with the development of neurodegenerative conditions. However, whether primary lower level blast overpressure (LBOP) causes neurodegeneration and neuroinflammation remains largely unknown. The aim of our present study was to determine whether LBOP can cause neuroinflammation and neurodegeneration. METHODS: Anesthetized rats were randomly assigned to LBOP group (70 kPa, n = 5) or sham group (without blast, n = 5). Histopathological and cytokine changes in brain tissue at 5 days post-injury were evaluated by hematoxylin-eosin staining and Bioplex assay, respectively. RESULTS: Histopathological assessment revealed neuronal degeneration and increased density of inflammatory cells in frontal and parietal cortex, hippocampus and thalamus in rats exposed to LBOP. LBOP exposure significantly elevated levels of pro-inflammatory cytokines (EPO, IL-1ß, IL-6, IL-12, IL-18, and TNF-α) and chemokines (GRO and RANTES) as well as of an anti-inflammatory cytokine (IL-13) in the frontal cortex. CONCLUSIONS: This study reveals a role of neuroinflammation in neurodegeneration after mild blast traumatic brain injury. Therapies that target this process might in warfighters might function either by attenuating the development of post-traumatic stress disorder, chronic traumatic encephalopathy and Alzheimer's disease, or by slowing their progression.


Subject(s)
Encephalitis/pathology , Explosions/statistics & numerical data , Nerve Degeneration/pathology , Animals , Biomarkers/analysis , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/pathology , Chemokine CCL5/analysis , Chemokine CXCL1/analysis , Chemokines/analysis , Cytokines/analysis , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Interleukin-12/analysis , Interleukin-18/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Rats/injuries , Tumor Necrosis Factor-alpha/analysis
3.
PLoS One ; 13(8): e0202594, 2018.
Article in English | MEDLINE | ID: mdl-30133517

ABSTRACT

BACKGROUND AND OBJECTIVE: Complement activation as an early and important inflammatory process contributes to multiple organ dysfunction after trauma. We have recently shown that complement inhibition by decay-accelerating factor (DAF) protects brain from blast-overpressure (BOP)-induced damage. This study was conducted to determine the effect of DAF on acute lung injury induced by BOP exposure and to elucidate its possible mechanisms of action. METHODS: Anesthetized adult male Sprague-Daley rats were exposed to BOP (120 kPa) from a compressed air-driven shock tube. Rats were randomly assigned to three experimental groups: 1) Control (no BOP and no DAF treatment), 2) BOP (120 kPa BOP exposure), and 3) BOP followed by treatment with rhDAF (500µg/kg, i.v) at 30 minutes after blast. After a recovery period of 3, 24, or 48 hours, animals were euthanized followed by the collection of blood and tissues at each time point. Samples were subjected to the assessment of cytokines and histopathology as well as for the interaction of high-mobility-group box 1 (HMGB1) protein, NF-κB, receptor for advanced glycation end products (RAGE), C3a, and C3aR. RESULTS: BOP exposure significantly increased in the production of systemic pro- and anti-inflammatory cytokines, and obvious pathological changes as characterized by pulmonary edema, inflammation, endothelial damage and hemorrhage in the lungs. These alterations were ameliorated by early administration of rhDAF. The rhDAF treatment not only significantly reduced the expression levels of HMGB1, RAGE, NF-κB, C3a, and C3aR, but also reversed the interaction of C3a-C3aR and nuclear translocation of HMGB1 in the lungs. CONCLUSIONS: Our findings indicate that early administration of DAF efficiently inhibits systemic and local inflammation, and mitigates blast-induced lung injury. The underlying mechanism might be attributed to its potential modulation of C3a-C3aR-HMGB1-transcriptional factor axis. Therefore, complement and/or HMGB1 may be potential therapeutic targets in amelioration of acute lung injury after blast injury.


Subject(s)
Acute Lung Injury/drug therapy , Blast Injuries/drug therapy , CD55 Antigens/administration & dosage , HMGB1 Protein/genetics , Inflammation/drug therapy , Acute Lung Injury/genetics , Acute Lung Injury/physiopathology , Animals , Blast Injuries/genetics , Blast Injuries/pathology , Complement Activation/drug effects , Complement C3a/antagonists & inhibitors , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/physiopathology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , NF-kappa B/genetics , Pressure/adverse effects , Rats , Rats, Sprague-Dawley
4.
Front Neurol ; 8: 219, 2017.
Article in English | MEDLINE | ID: mdl-28634463

ABSTRACT

Blast-induced traumatic brain injury is associated with acute and possibly chronic elevation of intracranial pressure (ICP). The outcome after TBI is dependent on the progression of complex processes which are mediated by oxidative stress. So far, no effective pharmacological protection against TBI exists. In this study, rats were exposed to a single or repetitive blast overpressure (BOP) at moderate intensities of 72 or 110 kPa in a compressed air-driven shock tube. The degree and duration of the increase in ICP were proportional to the intensity and frequency of the blast exposure(s). In most cases, a single dose of antioxidant N-acetylcysteine amide (NACA) (500 mg/kg) administered intravenously 2 h after exposure to BOP significantly attenuated blast-induced increase in ICP. A single dose of NACA was not effective in improving the outcome in the group of animals that were subjected to repetitive blast exposures at 110 kPa on the same day. In this group, two treatments with NACA at 2 and 4 h post-BOP exposure resulted in significant attenuation of elevated ICP. Treatment with NACA prior to BOP exposure completely prevented the elevation of ICP. The findings indicate that oxidative stress plays an important role in blast-induced elevated ICP as treatment with NACA-ameliorated ICP increase, which is frequently related to poor functional recovery after TBI.

5.
Biomed Res Int ; 2017: 2075463, 2017.
Article in English | MEDLINE | ID: mdl-28553646

ABSTRACT

Blast-induced traumatic brain injury (bTBI) is a leading cause of injuries in recent military conflicts and it is responsible for an increased number of civilian casualties by terrorist attacks. bTBI includes a variety of neuropathological changes depending on the intensity of blast overpressure (BOP) such as brain edema, neuronal degeneration, diffuse axonal damage, and vascular dysfunction with neurological manifestations of psychological and cognitive abnormalities. Internal jugular vein (IJV) compression is known to reduce intracranial compliance by causing an increase in brain volume and was shown to reduce brain damage during closed impact-induced TBI. We investigated whether IJV compression can attenuate signs of TBI in rats after exposure to BOP. Animals were exposed to three 110 ± 5 kPa BOPs separated by 30 min intervals. Exposure to BOP resulted in a significant decrease of neuronal nuclei (NeuN) together with upregulation of aquaporin-4 (AQP-4), 3-nitrotyrosine (3-NT), and endothelin 1 receptor A (ETRA) expression in frontal cortex and hippocampus one day following exposures. IJV compression attenuated this BOP-induced increase in 3-NT in cortex and ameliorated the upregulation of AQP-4 in hippocampus. These results suggest that elevated intracranial pressure and intracerebral volume have neuroprotective potential in blast-induced TBI.


Subject(s)
Blast Injuries/therapy , Brain Injuries, Traumatic/prevention & control , Frontal Lobe/physiopathology , Hippocampus/physiopathology , Intracranial Pressure , Animals , Blast Injuries/complications , Blast Injuries/metabolism , Blast Injuries/physiopathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/physiopathology , Frontal Lobe/metabolism , Frontal Lobe/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Organ Size , Rats , Rats, Sprague-Dawley
6.
PLoS One ; 11(12): e0167510, 2016.
Article in English | MEDLINE | ID: mdl-27907158

ABSTRACT

Exposure to blast overpressure (BOP) activates a cascade of pathological processes including changes in intracranial pressure (ICP) and blood-brain barrier (BBB) permeability resulting in traumatic brain injury (TBI). In this study the effect of single and multiple exposures at two intensities of BOP on changes in ICP and BBB permeability in Sprague-Dawley rats was evaluated. Animals were exposed to a single or three repetitive (separated by 0.5 h) BOPs at 72 kPa or 110 kPa. ICP was monitored continuously via telemetry for 6 days after exposure to BOP. The alteration in the permeability of BBB was determined by extravasation of Evans Blue (EB) into brain parenchyma. A significant increase in ICP was observed in all groups except the single 72 kPa BOP group. At the same time a marked increase in BBB permeability was also seen in various parts of the brain. The extent of ICP increase as well as BBB permeability change was dependent on intensity and frequency of blast.


Subject(s)
Blast Injuries/metabolism , Blast Injuries/physiopathology , Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Intracranial Pressure , Animals , Brain Injuries/pathology , Disease Models, Animal , Explosions , Fluorescent Dyes/metabolism , Male , Permeability , Rats , Time Factors
7.
Int J Mol Sci ; 16(12): 28979-97, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26690122

ABSTRACT

Intracranial pressure (ICP) measurements are essential in evaluation and treatment of neurological disorders such as subarachnoid and intracerebral hemorrhage, ischemic stroke, hydrocephalus, meningitis/encephalitis, and traumatic brain injury (TBI). The techniques of ICP monitoring have evolved from invasive to non-invasive-with both limitations and advantages. Some limitations of the invasive methods include short-term monitoring, risk of infection, restricted mobility of the subject, etc. The invasiveness of a method limits the frequency of ICP evaluation in neurological conditions like hydrocephalus, thus hampering the long-term care of patients with compromised ICP. Thus, there has been substantial interest in developing noninvasive techniques for assessment of ICP. Several approaches were reported, although none seem to provide a complete solution due to inaccuracy. ICP measurements are fundamental for immediate care of TBI patients in the acute stages of severe TBI injury. In severe TBI, elevated ICP is associated with mortality or poor clinical outcome. ICP monitoring in conjunction with other neurological monitoring can aid in understanding the pathophysiology of brain damage. This review article presents: (a) the significance of ICP monitoring; (b) ICP monitoring methods (invasive and non-invasive); and (c) the role of ICP monitoring in the management of brain damage, especially TBI.


Subject(s)
Brain Injuries/diagnosis , Brain Injuries/physiopathology , Brain/physiopathology , Intracranial Pressure , Animals , Diagnostic Techniques and Procedures , Disease Management , Humans , Telemetry/methods
8.
Clin Exp Ophthalmol ; 43(3): 239-46, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25112787

ABSTRACT

BACKGROUND: The incidence of blast-induced ocular injury has dramatically increased due to advances in weaponry and military tactics. A single exposure to blast overpressure (BOP) has been shown to cause damage to the eye in animal models; however, on the battlefield, military personnel are exposed to BOP multiple times. The effects of repeated exposures to BOP on ocular tissues have not been investigated. The purpose of this study is to characterize the effects of single or repeated exposure on ocular tissues. METHODS: A compressed air shock tube was used to deliver 70 ± 7 KPa BOP to rats, once (single blast overpressure [SBOP]) or once daily for 5 days (repeated blast overpressure [RBOP]). Immunohistochemistry was performed to characterize the pathophysiology of ocular injuries induced by SBOP and RBOP. Apoptosis was determined by quantification activated caspase 3. Gliosis was examined by detection of glial fibrillary acidic protein (GFAP). Inflammation was examined by detection of CD68. RESULTS: Activated caspase 3 was detected in ocular tissues from all animals subjected to BOP, while those exposed to RBOP had more activated caspase 3 in the optic nerve than those exposed to SBOP. GFAP was detected in the retinas from all animals subjected to BOP. CD68 was detected in optic nerves from all animals exposed to BOP. CONCLUSION: SBOP and RBOP induced retinal damage. RBOP caused more apoptosis in the optic nerve than SBOP, suggesting that RBOP causes more severe optic neuropathy than SBOP. SBOP and RBOP caused gliosis in the retina and increased inflammation in the optic nerve.


Subject(s)
Air Pressure , Blast Injuries/physiopathology , Disease Models, Animal , Eye Injuries/physiopathology , Gliosis/physiopathology , Optic Nerve Injuries/physiopathology , Retina/injuries , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoptosis , Blast Injuries/metabolism , Caspase 3/metabolism , Eye Injuries/metabolism , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Immunoenzyme Techniques , Male , Optic Nerve Injuries/metabolism , Rats , Rats, Long-Evans
9.
Mil Med ; 179(8 Suppl): 34-40, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25102547

ABSTRACT

BACKGROUND: Blast-induced ocular trauma is a frequent cause of morbidity for survivors of improvised explosive devices. Blast overpressure (BOP) of 120 ± 7 KPa has been shown to cause damage to lungs, brain, and gut in a rat model; however, the effects of BOP on ocular tissues have not been characterized. To elucidate the pathophysiology of blast-induced ocular trauma, ocular tissues from rats subjected to blast were examined for evidence of apoptosis by the detection of activated caspase 3 and TUNEL assay in their ocular tissues. METHODS: A compressed air shock tube was used to deliver 120 ± 7 KPa of BOP for duration of 2 msec to the right side of the rats. Rats were then euthanized at specific time points after blast exposure (3 hours, 24 hours, 48 hours). Ocular tissues were processed for immunohistochemistry to detect activated caspase 3 and TUNEL assay. Tissues were evaluated for relative levels of positive signal as compared to nonblast exposed controls. RESULTS: Activated caspase 3 was detected in the optic nerve, ganglion layer, and inner nuclear layer post blast exposure. At 24 and 48 hours, the inner nuclear layer from the right side had more cells with activated caspase 3. In the optic nerve, the highest levels of activated caspase 3 were detected on the right side at 24 hours post blast. CONCLUSION: BOP of 120 ± 7 KPa induces optic neuropathy and retinal damage. In both the optic nerve and retina, caspase 3 was activated in the right and left sides following blast exposure. The results of this study reveal that blast exposure induces apoptosis in both the optic nerve and retinal tissues.


Subject(s)
Blast Injuries/physiopathology , Eye Injuries/physiopathology , Optic Nerve Injuries/physiopathology , Retina/injuries , Animals , Apoptosis , Caspase 3/analysis , Male , Optic Nerve Injuries/metabolism , Rats , Rats, Sprague-Dawley , Retina/chemistry
10.
Acta Neuropathol Commun ; 2: 67, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24938728

ABSTRACT

BACKGROUND: Blast-related traumatic brain injury (TBI) is a common cause of injury in the military operations in Iraq and Afghanistan. How the primary blast wave affects the brain is not well understood. The aim of the present study was to examine whether blast exposure affects the cerebral vasculature in a rodent model. We analyzed the brains of rats exposed to single or multiple (three) 74.5 kPa blast exposures, conditions that mimic a mild TBI. Rats were sacrificed 24 hours or between 6 and 10 months after exposure. Blast-induced cerebral vascular pathology was examined by a combination of light microscopy, immunohistochemistry, and electron microscopy. RESULTS: We describe a selective vascular pathology that is present acutely at 24 hours after injury. The vascular pathology is found at the margins of focal shear-related injuries that, as we previously showed, typically follow the patterns of penetrating cortical vessels. However, changes in the microvasculature extend beyond the margins of such lesions. Electron microscopy revealed that microvascular pathology is found in regions of the brain with an otherwise normal neuropil. This initial injury leads to chronic changes in the microvasculature that are still evident many months after the initial blast exposure. CONCLUSIONS: These studies suggest that vascular pathology may be a central mechanism in the induction of chronic blast-related injury.


Subject(s)
Blast Injuries/complications , Brain Injuries/etiology , Brain Injuries/pathology , Cerebral Cortex/pathology , Cerebral Hemorrhage/etiology , Vasculitis, Central Nervous System/etiology , Animals , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Cerebral Hemorrhage/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Male , Microscopy, Electron , Microvessels/metabolism , Microvessels/pathology , Microvessels/ultrastructure , Rats , Rats, Long-Evans , Time Factors , Vasculitis, Central Nervous System/pathology
11.
IEEE Trans Biomed Eng ; 61(3): 841-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24557686

ABSTRACT

The long-term monitoring of intracranial pressure (ICP) is important for the management of acute and chronic neuropathological conditions which include head injury, traumatic brain injury, hydrocephalus, etc. In this study, we developed an implantable device for measuring ICP over long periods of time in an animal model of blast-induced brain injury. The performance of the device was first evaluated in vitro and subsequently utilized to measure ICP in rats exposed to blast overpressures. The effects of blast-induced brain injury on ICP were measured for six days. A significant difference was observed between the injured group and the nonexposed control group. ICP in injured animals showed a biphasic transient increase; an immediate increase within the first 1-3 h and a more gradual elevation occurring two days after the blast. The ability to monitor changes of ICP continuously over long periods after brain injury and during the course of treatment may improve the prognosis after injury and can also serve as a tool in determining the therapeutic effectiveness of new drugs.


Subject(s)
Brain Injuries/physiopathology , Intracranial Pressure/physiology , Monitoring, Physiologic/instrumentation , Telemetry/instrumentation , Animals , Equipment Design , Male , Monitoring, Physiologic/methods , Rats , Rats, Sprague-Dawley
12.
Acta Neuropathol Commun ; 1: 51, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-24252601

ABSTRACT

BACKGROUND: Blast-related traumatic brain injury (TBI) has been a significant cause of injury in the military operations of Iraq and Afghanistan, affecting as many as 10-20% of returning veterans. However, how blast waves affect the brain is poorly understood. To understand their effects, we analyzed the brains of rats exposed to single or multiple (three) 74.5 kPa blast exposures, conditions that mimic a mild TBI. RESULTS: Rats were sacrificed 24 hours or between 4 and 10 months after exposure. Intraventricular hemorrhages were commonly observed after 24 hrs. A screen for neuropathology did not reveal any generalized histopathology. However, focal lesions resembling rips or tears in the tissue were found in many brains. These lesions disrupted cortical organization resulting in some cases in unusual tissue realignments. The lesions frequently appeared to follow the lines of penetrating cortical vessels and microhemorrhages were found within some but not most acute lesions. CONCLUSIONS: These lesions likely represent a type of shear injury that is unique to blast trauma. The observation that lesions often appeared to follow penetrating cortical vessels suggests a vascular mechanism of injury and that blood vessels may represent the fault lines along which the most damaging effect of the blast pressure is transmitted.


Subject(s)
Blast Injuries/physiopathology , Brain Injuries/physiopathology , Brain/physiopathology , Animals , Apoptosis/physiology , Blast Injuries/complications , Blast Injuries/pathology , Blast Injuries/psychology , Brain/pathology , Brain Hemorrhage, Traumatic/etiology , Brain Hemorrhage, Traumatic/pathology , Brain Hemorrhage, Traumatic/physiopathology , Brain Hemorrhage, Traumatic/psychology , Brain Injuries/etiology , Brain Injuries/pathology , Brain Injuries/psychology , Dendrites/pathology , Dendrites/physiology , Disease Models, Animal , Exploratory Behavior/physiology , Gliosis/etiology , Gliosis/pathology , Gliosis/physiopathology , Male , Microglia/pathology , Microglia/physiology , Neurons/pathology , Neurons/physiology , Pressure , Random Allocation , Rats , Rats, Long-Evans , Spatial Learning/physiology , Time Factors
13.
Acta Neuropathol Commun ; 1: 52, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-24252631

ABSTRACT

BACKGROUND: Blast-induced neurotrauma (BINT) is the signature life threatening injury of current military casualties. Neuroinflammation is a key pathological occurrence of secondary injury contributing to brain damage after blast injury. We have recently demonstrated that blast-triggered complement activation and cytokine release are associated with BINT. Here, we evaluated if administration of the complement inhibitor recombinant human decay-accelerating factor (rhDAF) is beneficial on neuroinflammation and neurodegeneration in a rat model of moderate BINT. Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI. RESULT: Administration of rhDAF after exposure to moderate blast overpressure (BOP, 120 kPa) mitigated brain injury characterized by neuronal degeneration. rhDAF treatment reduced complement hemolytic activity at 3 hours and tissue complement deposition at 3, 24, and 48 hours as well as systemic and local cytokine release at 24 hours post BOP. Furthermore, rhDAF protected blood-brain barrier (BBB) integrity and reduced cytotoxic edema. Interaction between complement cleavage component, C3a and C3a receptor and tau phosphorylation were also attenuated in rhDAF treated animals at 3 and 24 hours after BOP. CONCLUSION: These novel findings suggest early complement targeted inhibition as a new therapeutic strategy to decrease neuroinflammation and neurodegeneration after blast TBI.


Subject(s)
Blast Injuries/complications , Brain Injuries/drug therapy , Brain Injuries/etiology , CD55 Antigens/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain Edema/drug therapy , Brain Edema/etiology , Brain Edema/physiopathology , Brain Injuries/physiopathology , Cytokines/metabolism , Disease Models, Animal , Humans , Male , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Neuroimmunomodulation/drug effects , Pressure , Random Allocation , Rats, Sprague-Dawley , Time Factors , Treatment Outcome
14.
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
15.
Front Neurol ; 3: 32, 2012.
Article in English | MEDLINE | ID: mdl-22403572

ABSTRACT

Mild traumatic brain injury (mTBI) resulting from exposure to improvised explosive devices (IEDs) has fueled a requirement to develop animals models that mirror this condition using exposure to blast overpressure (BOP). En route to developing a model of repeated exposure to BOP we sought to initially characterize the effects of acute BOP exposure in rodents, focusing specifically on the levels of BOP exposure that produced clinical mTBI symptoms. We first measured BOP effects on gross motor function on a balance beam. Separate groups of unanesthetized rats were exposed (in different orientations) to 36.6, 74.5, and 116.7 kPa BOP exposure inside a pneumatically driven shock tube. Results demonstrated that rats exposed to 116.7 kPa demonstrated transient alterations or loss of consciousness indicated by a transient loss of righting and by increased latencies on the balance beam. The 116.7 kPa exposure was the threshold for overt pathology for acute BOP exposure with approximately 30% of rats presenting with evidence of subdural hemorrhage and cortical contusions. All animals exposed to 116.7 kPa BOP manifested evidence of significant pulmonary hemorrhage. Anterograde memory deficits were observed in rats exposed to 74.5 kPa facing the BOP wave and rats exposed to 116.7 kPa in the lateral (side) orientation. We next assessed repeated exposure to either lateral or frontal 36.6 kPa BOP in anesthetized rats, once per day for 12 days. Results showed that repeated exposure in the frontal, but not side, orientation to the BOP wave produced a transitory learning deficit on a Morris water maze task as shown by significantly longer latencies to reach the submerged platform in the second and third blocks of a four block session. Implications of these data are discussed in relation to the manifestation of mTBI in military personnel exposed to IEDs. Finally, we suggest that there are multiple types of long-term brain injury from blast exposure.

16.
J Neurotrauma ; 29(7): 1379-87, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22352906

ABSTRACT

Blast-induced traumatic brain injury (TBI) is of significant concern in soldiers returning from the current conflicts in Iraq and Afghanistan. Incidents of TBI have increased significantly in the current conflicts compared to previous wars, and a majority of these injuries are caused by improvised explosive devices. Currently, no specific technique or biomarker is available for diagnosing TBI when no obvious clinical symptoms are present. Micro-RNAs are small RNA (~ 22nts) molecules that are expressed endogenously and play an important role in regulating gene expression. MicroRNAs have emerged as novel serum diagnostic biomarkers for various diseases. In this study, we studied the effect of blast overpressure injury on the microRNA signatures in the serum of rats. Rats were exposed to three serial 120-kPa blast overpressure exposures through a shockwave tube. Blood and cerebrospinal fluid were collected at various time points after injury, and microRNA modulation was analyzed using real-time PCR. Five microRNAs were significantly modulated in the serum samples of these animals at three time points post-injury. Further, we also found that the levels of microRNA let-7i are also elevated in cerebrospinal fluid post-blast wave exposure. The presence of microRNA in both serum and cerebrospinal fluid immediately after injury makes microRNA let-7i an ideal candidate for further studies of biomarkers in TBI.


Subject(s)
Blast Injuries/diagnosis , Blast Injuries/genetics , Brain Injuries/diagnosis , MicroRNAs/blood , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Blast Injuries/blood , Brain Injuries/blood , Brain Injuries/cerebrospinal fluid , Disease Models, Animal , Male , MicroRNAs/biosynthesis , MicroRNAs/cerebrospinal fluid , Rats , Rats, Sprague-Dawley
17.
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
18.
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
19.
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
20.
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
...