Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 19(1): 288, 2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36463243

ABSTRACT

BACKGROUND: Adverse pathophysiological and behavioral outcomes related to mild traumatic brain injury (mTBI), posttraumatic stress disorder (PTSD), and chronic pain are common following blast exposure and contribute to decreased quality of life, but underlying mechanisms and prophylactic/treatment options remain limited. The dynorphin/kappa opioid receptor (KOR) system helps regulate behavioral and inflammatory responses to stress and injury; however, it has yet to be investigated as a potential mechanism in either humans or animals exposed to blast. We hypothesized that blast-induced KOR activation mediates adverse outcomes related to inflammation and affective behavioral response. METHODS: C57Bl/6 adult male mice were singly or repeatedly exposed to either sham (anesthesia only) or blast delivered by a pneumatic shock tube. The selective KOR antagonist norBNI or vehicle (saline) was administered 72 h prior to repetitive blast or sham exposure. Serum and brain were collected 10 min or 4 h post-exposure for dynorphin A-like immunoreactivity and cytokine measurements, respectively. At 1-month post-exposure, mice were tested in a series of behavioral assays related to adverse outcomes reported by humans with blast trauma. RESULTS: Repetitive but not single blast exposure resulted in increased brain dynorphin A-like immunoreactivity. norBNI pretreatment blocked or significantly reduced blast-induced increase in serum and brain cytokines, including IL-6, at 4 h post exposure and aversive/anxiety-like behavioral dysfunction at 1-month post-exposure. CONCLUSIONS: Our findings demonstrate a previously unreported role for the dynorphin/KOR system as a mediator of biochemical and behavioral dysfunction following repetitive blast exposure and highlight this system as a potential prophylactic/therapeutic treatment target.


Subject(s)
Blast Injuries , Dynorphins , Receptors, Opioid, kappa , Animals , Male , Mice , Blast Injuries/complications , Blast Injuries/genetics , Blast Injuries/immunology , Brain/immunology , Brain/physiology , Dynorphins/genetics , Dynorphins/immunology , Quality of Life , Receptors, Opioid, kappa/genetics , Receptors, Opioid, kappa/immunology
2.
J Neurotrauma ; 37(12): 1463-1480, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32056479

ABSTRACT

The purpose of this study was to characterize acute changes in inflammatory pathways in the mouse eye after blast-mediated traumatic brain injury (bTBI) and to determine whether modulation of these pathways could protect the structure and function of retinal ganglion cells (RGC). The bTBI was induced in C57BL/6J male mice by exposure to three 20 psi blast waves directed toward the head with the body shielded, with an inter-blast interval of one hour. Acute cytokine expression in retinal tissue was measured through reverse transcription-quantitative polymerase chain reaction (RT-qPCR) four hours post-blast. Increased retinal expression of interleukin (lL)-1ß, IL-1α, IL-6, and tumor necrosis factor (TNF)α was observed in bTBI mice exposed to blast when compared with shams, which was associated with activation of microglia and macroglia reactivity, assessed via immunohistochemistry with ionized calcium binding adaptor molecule 1 and glial fibrillary acidic protein, respectively, one week post-blast. Blockade of the IL-1 pathway was accomplished using anakinra, an IL-1RI antagonist, administered intra-peritoneally for one week before injury and continuing for three weeks post-injury. Retinal function and RGC layer thickness were evaluated four weeks post-injury using pattern electroretinogram (PERG) and optical coherence tomography (OCT), respectively. After bTBI, anakinra treatment resulted in a preservation of RGC function and RGC structure when compared with saline treated bTBI mice. Optic nerve integrity analysis demonstrated a trend of decreased damage suggesting that IL-1 blockade also prevents axonal damage after blast. Blast exposure results in increased retinal inflammation including upregulation of pro-inflammatory cytokines and activation of resident microglia and macroglia. This may explain partially the RGC loss we observed in this model, as blockade of the acute inflammatory response after injury with the IL-1R1 antagonist anakinra resulted in preservation of RGC function and RGC layer thickness.


Subject(s)
Brain Injuries, Traumatic/immunology , Immunity/immunology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Receptors, Interleukin-1/antagonists & inhibitors , Retina/immunology , Visual Perception/immunology , Animals , Blast Injuries/diagnostic imaging , Blast Injuries/drug therapy , Blast Injuries/immunology , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/drug therapy , Electroretinography/methods , Immunity/drug effects , Interleukin 1 Receptor Antagonist Protein/pharmacology , Male , Mice , Mice, Inbred C57BL , Retina/diagnostic imaging , Retina/drug effects , Tomography, Optical Coherence/methods , Treatment Outcome , Visual Perception/drug effects
3.
J Neuroinflammation ; 17(1): 20, 2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31931830

ABSTRACT

BACKGROUND: There is a paucity of human data on exposure to blast traumatic brain injury (bTBI) and the corresponding systemic cytokine immune response at later time points (i.e., months, years) post-injury. METHODS: We conducted a repeated measures, case-control study, examining associations of serum levels of pro- and anti-inflammatory cytokines, measured both pre- and post-deployment with having mild and moderate/severe bTBI. Utilizing serum from the Department of Defense Serum Repository cytokines were measured via an ELISA-based array for 15 cytokines. We compared pre- vs. post-levels among mild cases, moderate/severe cases, and controls and carried out case-control comparisons, using paired t- tests and generalized linear models. RESULTS: The average time between bTBI and post-deployment/bTBI serum among cases was 315.8 days. From pre- to post-deployment/bTBI, levels of interleukin 8 (IL-8) were decreased among both mild cases (µ = - 83.43 pg/ml; s.e. = 21.66) and moderate/severe cases (µ = - 107.67 pg/ml; s.e. = 28.74 pg/ml), while levels increased among controls (µ = 32.86 pg/ml; s.e. = 30.29). The same pattern occurred for matrix metallopeptidase 3 (MMP3), with levels decreasing for moderate/severe cases (µ = - 3369.24 pg/ml; s.e. = 1701.68) and increasing for controls (µ = 1859.60 pg/ml; s.e. = 1737.51) from pre- to post-deployment/bTBI. Evidence was also suggestive of case-control differences, from pre- to post-deployment/bTBI for interleukin 1 alpha (IL-1α), interleukin 4 (IL-4), and interleukin 6 (IL-6) among moderate/severe cases. CONCLUSION: The findings of this longitudinal study indicate that in the chronic phase of bTBI, levels of IL-8 and MMP3 may be substantially lower than pre-injury. These results need confirmation in other studies, potentially those that account for treatment differences, which was not possible in our study.


Subject(s)
Blast Injuries/blood , Blast Injuries/complications , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/etiology , Cytokines/blood , Adult , Blast Injuries/immunology , Brain Injuries, Traumatic/immunology , Case-Control Studies , Female , Humans , Inflammation/blood , Longitudinal Studies , Male , Middle Aged , Military Personnel , Young Adult
4.
Curr Osteoporos Rep ; 17(6): 387-394, 2019 12.
Article in English | MEDLINE | ID: mdl-31721068

ABSTRACT

PURPOSE OF REVIEW: Heterotopic ossification (HO) is associated with inflammation. The goal of this review is to examine recent findings on the roles of inflammation and the immune system in HO. We examine how inflammation changes in fibrodysplasia ossificans progressiva, in traumatic HO, and in other clinical conditions of HO. We also discuss how inflammation may be a target for treating HO. RECENT FINDINGS: Both genetic and acquired forms of HO show similarities in their inflammatory cell types and signaling pathways. These include macrophages, mast cells, and adaptive immune cells, along with hypoxia signaling pathways, mesenchymal stem cell differentiation signaling pathways, vascular signaling pathways, and inflammatory cytokines. Because there are common inflammatory mediators across various types of HO, these mediators may serve as common targets for blocking HO. Future research may focus on identifying new inflammatory targets and testing combinatorial therapies based on these results.


Subject(s)
Inflammation/immunology , Myositis Ossificans/immunology , Ossification, Heterotopic/immunology , Wounds and Injuries/immunology , Adaptive Immunity/immunology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthroplasty, Replacement, Hip , Blast Injuries/immunology , Brain Injuries, Traumatic/immunology , Burns/immunology , Cell Differentiation/immunology , Cytokines/immunology , Humans , Hypoxia/immunology , Immunosuppressive Agents/therapeutic use , Inflammation/drug therapy , Janus Kinase Inhibitors/therapeutic use , Macrophages/immunology , Mast Cells/immunology , Mesenchymal Stem Cells , Myositis Ossificans/drug therapy , Ossification, Heterotopic/drug therapy , Postoperative Complications/immunology , Pyrazoles/therapeutic use , Receptors, Retinoic Acid/agonists , Signal Transduction , Sirolimus/therapeutic use , Spinal Cord Injuries/immunology , Stilbenes/therapeutic use , Retinoic Acid Receptor gamma
5.
Oxid Med Cell Longev ; 2019: 4848560, 2019.
Article in English | MEDLINE | ID: mdl-31565151

ABSTRACT

Although CD28 is associated with the expression of inflammatory mediators, apoptosis-related protein, immunosuppression, and tumorigenesis, the effects of CD28 deficiency on blast exposure-induced lung injury have not been investigated. In this study, we have explored the effects of CD28 on blast exposure-induced lung injury and studied its potential molecular mechanisms. A mouse model of blast exposure-induced acute lung injury was established. Sixty C57BL/6 wild-type (WT) and CD28 knockout (CD28-/-) mice were randomly divided into control or model groups. Lung tissue samples were collected 24 h and 48 h after blast injury. Histopathological changes and the expressions of inflammatory-related proteins were detected by hematoxylin-eosin, immunohistochemistry, and immunofluorescence staining. Apoptosis and oxidative stress were evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and reactive oxygen species (ROS). Inflammation, apoptosis, oxidative stress, and related pathway protein expression were studied by western blotting. In addition, the levels of CD3 and CD28 proteins were measured by flow cytometry. In the current study, we found that CD28 deficiency significantly inhibited blast exposure-induced increases in the lung weight/body weight ratio and wet weight/dry weight ratio; decreased the infiltration of CD44+ leukocytes, CD163+ macrophages, and CD3+ T cells into the lungs; reduced the expressions of proinflammatory cytokines including IL-1ß, TNF-α, and IL-6; and markedly increased IL-10 expression. CD28 deficiency also significantly attenuated blast exposure-induced ROS, MDA5, and IREα expressions; increased SOD-1 expression; lowered the number of apoptotic cells and Bax, Caspase-3, and active Caspase-8 expressions; and increased Bcl-2 expression. Additionally, CD28 deficiency significantly ameliorated blast exposure-induced increases of p-PI3K and p-Akt and ameliorated the decrease in the p-FoxO1 expression. Our results suggest that CD28 deficiency has a protective effect on blast exposure-induced lung injury, which might be associated with the PI3K/Akt/FoxO1 signaling pathway.


Subject(s)
Blast Injuries/immunology , CD28 Antigens/deficiency , Forkhead Box Protein O1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pneumonia/immunology , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocytes/immunology , Animals , Apoptosis/physiology , Blast Injuries/metabolism , Blast Injuries/pathology , CD28 Antigens/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/physiology , Pneumonia/metabolism , Random Allocation , Signal Transduction , T-Lymphocytes/pathology
6.
J Neurotrauma ; 35(19): 2306-2316, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29649942

ABSTRACT

Traumatic brain injury (TBI) is a major cause of neurological disorder and death in civilian and military populations. It comprises two components-direct injury from the traumatic impact and secondary injury from ensuing neural inflammatory responses. Blocking tumor necrosis factor-alpha (TNF-α), a central regulator of neural inflammation, has been shown to improve functional recovery after TBI. However, the mechanisms underlying those therapeutic effects are still poorly understood. Here, we examined effects of 3,6'-dithiothalidomide (dTT), a potentially therapeutic TNF-α inhibitor, in mice with blast-induced TBI. We found that blast exposure resulted in elevated expression of TNF-α, activation of microglial cells, enhanced excitatory synaptic transmission, reduced inhibitory synaptic transmission, and a loss of parvalbumin-positive (PV+) inhibitory interneurons. Administration of dTT for 5 days after the blast exposure completely suppressed blast-induced increases in TNF-α transcription, largely reversed blasted-induced synaptic changes, and prevented PV+ neuron loss. However, blocking TNF-α expression by dTT failed to mitigate blast-induced microglial activation in the hippocampus, as evidenced by their non-ramified morphology. These results indicate that TNF-α plays a major role in modulating neuronal functions in blast-induced TBI and that it is a potential target for treatment of TBI-related brain disorders.


Subject(s)
Blast Injuries/pathology , Brain Injuries, Traumatic/pathology , Hippocampus/pathology , Interneurons/pathology , Synaptic Transmission/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Blast Injuries/immunology , Brain Injuries, Traumatic/immunology , Hippocampus/immunology , Interneurons/immunology , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/immunology
7.
J Neurotrauma ; 35(1): 118-129, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28599600

ABSTRACT

Ocular blast injury is a major medical concern for soldiers and explosion victims due to poor visual outcomes. To define the changes in gene expression following a blast injury to the eye, we examined retinal ribonucleic acid (RNA) expression in 54 mouse strains 5 days after a single 50-psi overpressure air wave blast injury. We observe that almost 40% of genes are differentially expressed with a false discovery rate (FDR) of <0.001, even though the nominal changes in RNA expression are rather small. Moreover, we find through machine learning approaches that genetic networks related to the innate and acquired immune system are activated. Accompanied by lymphocyte invasion into the inner retina, blast injury also results in progressive loss of visual function and retinal ganglion cells (RGCs). Collectively, these data demonstrate how systems genetics can be used to put meaning to the transcriptome changes following ocular blast injury that eventually lead to blindness.


Subject(s)
Blast Injuries/genetics , Blast Injuries/immunology , Eye Injuries/pathology , Retina/pathology , Transcription, Genetic , Animals , Blast Injuries/pathology , Eye Injuries/immunology , Gene Expression/immunology , Gene Regulatory Networks/immunology , Mice , Retina/immunology , Transcription, Genetic/immunology
8.
J Trauma Acute Care Surg ; 81(3): 500-11, 2016 09.
Article in English | MEDLINE | ID: mdl-27306447

ABSTRACT

BACKGROUND: Understanding of the cellular immune response to primary blast lung injury (PBLI) is limited, with only the neutrophil response well documented. Moreover, its impact on the immune response in distal organs remains poorly understood. In this study, a rodent model of isolated primary blast injury was used to investigate the acute cellular immune response to isolated PBLI in the circulation and lung, including the monocyte response, and investigate distal subacute immune effects in the spleen and liver 6 hours after injury. METHODS: Rats were subjected to a shock wave (~135 kPa overpressure, 2 ms duration) inducing PBLI or sham procedure. Rat physiology was monitored, and at 1, 3, and 6 hours thereafter, blood, lung, and bronchoalveolar lavage fluid (BALF) were collected and analyzed by flow cytometry, enzyme-linked immunosorbent assay, and histologic examination. In addition, at 6 hours, spleen and liver were collected and analyzed by flow cytometry. RESULTS: Lung histology confirmed pulmonary barotrauma and inflammation. This was associated with rises in CXCL-1, interleukin 6 (IL-6), tumor necrosis factor α and albumin protein in the BALF. Significant acute increases in blood and lung neutrophils and CD43Lo/His48Hi (classical) monocytes/macrophages were detected. No significant changes were seen in blood or lung "nonclassical" monocyte and in natural killler, B, or T cells. In the BALF, significant increases were seen in neutrophils, CD43Lo monocyte-macrophages and monocyte chemoattractant protein-1. Significant increases in CD43Lo and Hi monocyte-macrophages were detected in the spleen at 6 hours. CONCLUSION: This study reveals a robust and selective response of CD43Lo/His48Hi (classical) monocytes, in addition to neutrophils, in blood and lung tissue following PBLI. An increase in monocyte-macrophages was also observed in the spleen at 6 hours. This profile of immune cells in the blood and BALF could present a new research tool for translational studies seeking to monitor, assess, or attenuate the immune response in blast-injured patients.


Subject(s)
Blast Injuries/immunology , Immunity, Cellular , Leukosialin/metabolism , Lung Injury/immunology , Monocytes/immunology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Chemokine CCL2/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Interleukin-6/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
9.
Shock ; 44(5): 470-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26418548

ABSTRACT

Extremity injury is a significant burden to those injured in explosive incidents and local ischaemia can result in poor functionality in salvaged limbs. This study examined whether blast injury to a limb resulted in a change in endothelial phenotype leading to changes to the surrounding tissue.The hind limbs of terminally anaesthetized rabbits were subjected to one of four blast exposures (high, medium, low, or no blast). Blood samples were analyzed for circulating endothelial cells pre-injury and at 1, 6, and 11 h postinjury as well as analysis for endothelial activation pre-injury and at 1, 6, and 12  h postinjury. Post-mortem tissue (12  h post-injury) was analysed for both protein and mRNA expression and also for histopathology. The high blast group had significantly elevated levels of circulating endothelial cells 6  h postinjury. This group also had significantly elevated tissue mRNA expression of IL-6, E-selection, TNF-α, HIF-1, thrombomodulin, and PDGF. There was a significant correlation between blast dose and the degree of tissue pathology (hemorrhage, neutrophil infiltrate, and oedema) with the worst scores in the high blast group. This study has demonstrated that blast injury can activate the endothelium and in some cases cause damage that in turn leads to pathological changes in the surrounding tissue. For the casualty injured by an explosion the damaging effects of hemorrhage and shock could be exacerbated by blast injury and vice versa so that even low levels of blast become damaging, all of which could affect tissue functionality and long-term outcomes.


Subject(s)
Blast Injuries/pathology , Endothelium, Vascular/injuries , Hindlimb/injuries , Animals , Blast Injuries/blood , Blast Injuries/complications , Blast Injuries/immunology , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Muscle, Skeletal/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Oxygen Consumption/physiology , Rabbits
10.
J Neuroinflammation ; 11: 192, 2014 Nov 29.
Article in English | MEDLINE | ID: mdl-25472427

ABSTRACT

BACKGROUND: Visual prognosis after an open globe injury is typically worse than after a closed globe injury due, in part, to the immune response that ensues following open globe trauma. There is a need for an animal model of open globe injury in order to investigate mechanisms of vision loss and test potential therapeutics. METHODS: The left eyes of DBA/2 J mice were exposed to an overpressure airwave blast. This strain lacks a fully functional ocular immune privilege, so even though the blast wave does not rupture the globe, immune infiltrate and neuroinflammation occurs as it would in an open globe injury. For the first month after blast wave exposure, the gross pathology, intraocular pressure, visual function, and retinal integrity of the blast-exposed eyes were monitored. Eyes were collected at three, seven, and 28 days to study the histology of the cornea, retina, and optic nerve, and perform immunohistochemical labeling with markers of cell death, oxidative stress, and inflammation. RESULTS: The overpressure airwave caused anterior injuries including corneal edema, neovascularization, and hyphema. Immune infiltrate was detected throughout the eyes after blast wave exposure. Posterior injuries included occasional retinal detachments and epiretinal membranes, large retinal pigment epithelium vacuoles, regional photoreceptor cell death, and glial reactivity. Optic nerve degeneration was evident at 28 days post-blast wave exposure. The electroretinogram (ERG) showed an early deficit in the a wave that recovered over time. Both visual acuity and the ERG b wave showed an early decrease, then a transient improvement that was followed by further decline at 28 days post-blast wave exposure. CONCLUSIONS: Ocular blast injury in the DBA/2 J mouse recapitulates damage that is characteristic of open globe injuries with the advantage of a physically intact globe that prevents complications from infection. The injury was more severe in DBA/2 J mice than in C57Bl/6 J mice, which have an intact ocular immune privilege. Early injury to the outer retina mostly recovers over time. In contrast, inner retinal dysfunction seems to drive later vision loss.


Subject(s)
Blast Injuries/immunology , Blast Injuries/pathology , Eye Injuries/immunology , Eye Injuries/pathology , Immunity, Cellular/immunology , Animals , Electroretinography/methods , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Visual Acuity/immunology
11.
J Neurotrauma ; 30(22): 1888-97, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23777197

ABSTRACT

Blast wave-induced traumatic injury from terrorist explosive devices can occur at any time in either military or civilian environments. To date, little work has focused on the central nervous system response to a non-penetrating blast injury. We have evaluated the effect of a single 80-psi blast-overpressure wave in a rat model. Histological and immunochemical studies showed an early inflammatory response, tissue damage and the initiation of apoptosis. With regard to inflammation, polymorphonuclear leukocytes and lymphocytes infiltrated brain parenchyma within 1 h post-blast. Glial-fibrillary protein, cyclo-oxygenase-2ir, interleukin-1ß and tumor necrosis factor were present by 1 h and remained detectable at three weeks post-injury. High mobility group box-1 protein was detectable at three weeks. With regard to tissue damage, S100ß and 4-hydroxynonenal were present at 1 h and remained detectable at three weeks. Amyloid precursor protein was detectable at three weeks. As for apoptosis, Cleaved Caspase-3 was detectable at three weeks. Morris water maze assessment of cognitive function showed that blast injured animals required significantly more time to reach the platform on day 1 of training and traveled a greater distance to get to the platform on days 1 and 2. Blast-injured animals showed a significant increase in swimming speed (p<0.001), increased total distance traveled (p<0.001) and increased number of entries into the previous quadrant that had contained the escape platform (p<0.05). Magnetic resonance imaging showed hyperintense regions in the somatosensory area within 1 h. T2 relaxation times and apparent diffusion coefficients show increasing trends in both somatosensory and cortical regions. These data indicate an early and lasting response of brain tissue to non-penetrating blast over-pressure injury. This early inflammatory response is indicative of a mild traumatic brain injury. There is evidence of early hippocampal dysfunction.


Subject(s)
Blast Injuries/physiopathology , Brain Injuries/physiopathology , Brain/physiopathology , Cognition Disorders/physiopathology , Inflammation/physiopathology , Animals , Biomarkers/blood , Biomarkers/metabolism , Blast Injuries/etiology , Blast Injuries/immunology , Brain/immunology , Brain Injuries/etiology , Brain Injuries/immunology , Cognition Disorders/etiology , Cognition Disorders/immunology , Disease Models, Animal , Hippocampus/immunology , Hippocampus/injuries , Hippocampus/physiopathology , Inflammation/etiology , Inflammation/immunology , Magnetic Resonance Imaging , Male , Maze Learning/physiology , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Time Factors
12.
J Surg Res ; 183(1): 377-84, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23374757

ABSTRACT

BACKGROUND: Posttraumatic lung contusion is common after blunt chest trauma, and patients often need ventilatory support. Lung contusion induces an inflammatory response signified by primed polymorph neutrophil granulocytes (PMNs) in blood and tissue. Mechanical ventilation (MV) can also cause an inflammatory response. The aim of this study was to develop an animal model to investigate the effect of high-volume ventilation on the inflammatory response in blunt chest trauma. MATERIALS AND METHODS: We assigned 23 male Sprague-Dawley rats to either MV or bilateral lung contusion followed by MV. We used three extra rats as controls. Lung contusion was induced by a blast generator, a device releasing a single pressure blast wave centered on the chest. We determined tissue and systemic inflammation by absolute PMN numbers in blood and bronchoalveolar lavage fluid (BALF), myeloperoxidase, interleukin (IL)-6, IL 1ß, growth-related oncogene-KC, and IL-10 in both plasma and BALF. RESULTS: Survival after blunt chest trauma was correlated to the distance to the blast generator. Compared with controls, both MV and blast plus MV rats showed increased systemic and pulmonary inflammation, expressed by higher PMNs, myeloperoxidase levels, and cytokine levels in both blood and BALF. Blast plus MV rats showed a higher systemic and pulmonary inflammatory response than MV rats. CONCLUSIONS: The blast generator generated reproducible blunt chest trauma in rats. Mechanical ventilation after lung contusion induced a larger overall inflammatory response than MV alone, which indicates that local damage contributes not only to local inflammation, but also to systemic inflammation. This emphasizes the importance of lung protective ventilation strategies after pulmonary contusion.


Subject(s)
Blast Injuries/complications , Contusions/complications , Disease Models, Animal , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/immunology , Animals , Blast Injuries/immunology , Contusions/immunology , Inflammation/etiology , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results
13.
J Orthop Trauma ; 26(11): e204-13, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22588530

ABSTRACT

OBJECTIVE: Heterotopic ossification (HO) develops frequently after modern high-energy penetrating war injuries. The purpose of this prospective study was to identify and characterize the unique cytokine and chemokine profile associated with the development of HO as it pertained to the systemic inflammatory response after penetrating combat-related trauma. METHODS: Patients with high-energy penetrating extremity wounds were prospectively enrolled. Surgical debridement along with the use of a pulse lavage and vacuum-assisted-closure device was performed every 48-72 hours until definitive wound closure. Wound bed tissue biopsy, wound effluent, and serum were collected before each debridement. Effluent and serum were analyzed for 22 relevant cytokines and chemokines. Tissue was analyzed quantitatively for bacterial colonization. Correlations between specific wound and patient characteristics were also analyzed. The primary clinical outcome measure was the formation of HO as confirmed by radiographs at a minimum of 2 months of follow-up. RESULTS: Thirty-six penetrating extremity war wounds in 24 patients were investigated. The observed rate of HO in the study population was 38%. Of the 36 wounds, 13 (36%) demonstrated HO at a minimum follow-up of 2 months. An elevated injury severity score was associated with the development of HO (P = 0.006). Wound characteristics that correlated with the development of HO included impaired healing (P = 0.005) and bacterial colonization (P < 0.001). Both serum (interleukin-6, interleukin-10, and MCP-1) and wound effluent (IP-10 and MIP-1α) cytokine and chemokine bioprofiles were individually associated and suggestive of the development of HO (P < 0.05). CONCLUSIONS: A severe systemic and wound-specific inflammatory state as evident by elevated levels of inflammatory cytokines, elevated injury severity score, and bacterial wound colonization is associated with the development of HO. These findings suggest that the development of HO in traumatic combat-related wounds is associated with a hyper-inflammatory systemic response to injury. LEVEL OF EVIDENCE: Prognostic Level II. See Instructions for Authors for a complete description of levels of evidence.


Subject(s)
Blast Injuries/immunology , Cytokines/immunology , Inflammation/immunology , Ossification, Heterotopic/immunology , Warfare , Wounds, Penetrating/immunology , Blast Injuries/blood , Cytokines/blood , Female , Humans , Inflammation/blood , Male , Ossification, Heterotopic/blood , Wounds, Penetrating/blood , Young Adult
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.
Injury ; 42(9): 905-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22081818

ABSTRACT

BACKGROUND: The regulation of neuroendocrine hormones on the innate immune responses in trauma has not been fully understood. Previous studies have shown that the neuroendocrine hormones are important factors in their effects on immune parameters, depending on their concentration and timing instead of the simple suppressive effects. MATERIALS AND METHODS: A total of 144 Sprague­Dawley rats were divided randomly into sham, pulmonary blast injury (BI) and adrenalectomy plus pulmonary BI groups. Bilateral adrenalectomy was performed on rats, which were then subjected to blast injury. Following this, peripheral leucocyte responsiveness to lipopolysaccharide (LPS) stimulation, phagocytosis activities of macrophages and bacteria translocation (BT) were examined. Tumour necrosis factor-a (TNF-a) levels and the expression levels of scavenger receptor (SR) A, CD14, Toll-like receptor (TLR) 4 and MD2 were assayed with enzyme-linked immunosorbent assay and reverse transcription-polymerase chain reaction, respectively. RESULTS: In adrenalectomised rats after pulmonary BI, the number of peripheral leucocytes was increased and the phagocytosis of peritoneal and splenic macrophages was decreased as compared to the BI group. Simultaneously, the gut-derived BT and TNF-a secretion in lung tissues were elevated, whilst the LPS-stimulated TNF-a synthesis by peripheral leucocyte responsiveness was reduced. Furthermore, the mRNA levels of SR-A, CD14, TLR4 and MD2 in lung tissues of adrenalectomised rats decreased. Adrenalectomised rats showed enhancement of inflammatory responses and severe tissue injuries in trauma. CONCLUSIONS: Release of adrenal hormones might enhance, rather than inhibit, the innate immune functions, particularly in the early stages of trauma.


Subject(s)
Adrenalectomy , Blast Injuries/immunology , Immunity, Innate/physiology , Lung Injury/immunology , Animals , Bacterial Translocation , Blast Injuries/surgery , Cell Count , Enzyme-Linked Immunosorbent Assay , Leukocytes , Lipopolysaccharides/metabolism , Lung/metabolism , Lung Injury/surgery , Lymph Nodes/metabolism , Macrophages/physiology , Male , Phagocytosis , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
16.
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
17.
Mil Med ; 172(2): 190-5, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17357775

ABSTRACT

The aim of this study was to assess the prognostic value of tumor necrosis factor (TNF) alpha, interleukin (IL)-8, IL-4, and IL-10 in combat casualties. Fifty-six casualties with severe trauma (blast and explosive) who developed sepsis and 20 casualties with the same severity of trauma without sepsis were enrolled in this study. Fifty-five casualties developed multiple organ dysfunction syndrome; 36 died. Blood was drawn on the first day of trauma. Concentrations of IL-8, TNF-alpha, IL-4, and IL-10 were determined in plasma using enzyme-linked immunosorbent assays. Mean values of IL-8 were 230-fold, IL-10 were 42-fold, and TNF-alpha were 17-fold higher in trauma and sepsis group (p < 0.01). Mean values of IL-8 were 60-fold, TNF-alpha were 43.5-fold, and IL-10 were 70-fold higher in the multiple organ dysfunction syndrome group (p < 0.01). Mean values of IL-8 were 2.3-fold and IL-10 were 1.4-fold higher in nonsurvivors and TNF-alpha were 2.2-fold higher in survivors (p < 0.01). IL-4 had no significance as a predictor of severity and outcome.


Subject(s)
Blast Injuries/immunology , Cytokines/blood , Interleukin-10/blood , Sepsis/immunology , Warfare , Adolescent , Adult , Aged , Biomarkers/blood , Blast Injuries/blood , Blast Injuries/complications , Child , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Follow-Up Studies , Humans , Interleukin-10/immunology , Interleukin-8/blood , Interleukin-8/immunology , Male , Middle Aged , Prognosis , Retrospective Studies , Sepsis/blood , Sepsis/etiology , Trauma Severity Indices , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
18.
Am J Disaster Med ; 2(1): 21-5, 2007.
Article in English | MEDLINE | ID: mdl-18268871

ABSTRACT

OBJECTIVE: Bombing is the primary weapon of global terrorism, and it results in a complicated, multidimensional injury pattern. It induces bodily injuries through the well-documented primary, secondary, tertiary, and quaternary mechanisms of blast. Their effects dictate special medical concern and timely implementation of diagnostic and management strategies. Our objective is to report on clinical observations of patients admitted to the Tel Aviv Medical Center following a terrorist bombing. RESULTS: The explosion injured 27 patients, and three died. Four survivors who had been in close proximity to the explosion, as indicated by their eardrum perforation and additional blast injuries, were exposed to the blast wave. They exhibited a unique and immediate hyperinflammatory state, two upon admission to the intensive care unit and two during surgery. This hyperinflammatory state manifested as hyperpyrexia, sweating, low central venous pressure, and positive fluid balance. This state did not correlate with the complexity of injuries sustained by any of the 67 patients admitted to the intensive care unit after previous bombings. CONCLUSION: The patients' hyperinflammatory behavior, unrelated to their injury complexity and severity of trauma, indicates a new injury pattern in explosions, termed the "quinary blast injury pattern." Unconventional materials used in the manufacture of the explosive can partly explain the observed early hyperinflammatory state. Medical personnel caring for blast victims should be aware of this new type of bombing injury.


Subject(s)
Blast Injuries/physiopathology , Bombs , Inflammation/physiopathology , Pentaerythritol Tetranitrate/adverse effects , Adult , Blast Injuries/immunology , Disaster Planning , Fever , Humans , Male , Trauma Severity Indices
19.
Int J Immunopathol Pharmacol ; 18(3): 547-56, 2005.
Article in English | MEDLINE | ID: mdl-16164836

ABSTRACT

Impact of blast shock waves (SW) with the body wall produces blast lung injuries characterized by bilateral traumatic hemorrhages. Such injuries often have no external signs, are difficult to diagnose, and therefore, are frequently underestimated. Predictive assessment of acute respiratory distress syndrome outcome in SW-related accidents should be based on experimental data from appropriate animal models. Blood plasma transferrin is a major carrier of blood iron essential for proliferative "emergency" response of hematopoietic and immune systems as well as injured tissue in major trauma. Iron-transferrin complexes (Fe3+ TRF) can be quantitatively analyzed in blood and tissue samples with low-temperature EPR techniques. We hypothesized that use of EPR techniques in combination with assays for pro-inflammatory cytokines and granulocytes in the peripheral blood and BAL would reveal a pattern of systemic sequestration of (Fe3+)TRF that could be useful for development of biomarkers of the systemic inflammatory response to lung injury. With this goal we (i) analyzed time-dependent dynamics of (Fe3+)TRF in the peripheral blood of rats after impacts of SW generated in a laboratory shock-tube and (ii) assayed the fluctuation of granulocyte (PMN) counts and expression of CD11b adhesion molecules on the surface of PMNs during the first 24 h after SW induced injury. Sham-treated animals were used as control. Exposure to SW led to a significant decrease in the amount of blood (Fe3+)TRF that correlated with the extent of lung injury and developed gradually during the first 24 h. Thus, sequestration of (Fe3+)TRF occurred as early as 3 h post-exposure. At that time, the steady state concentration of (Fe3+)TRF in blood samples decreased from 19.7+/-0.6 microM in controls to 7.5+/-1.3 microM in exposed animals. The levels of (Fe3+)TRF remained decreased throughout the entire study period. PMN counts increased 5-fold and 3.5-fold over controls respectively, at 3 and 6 h postexposure. These effects were accompanied by an increase in expression of CD11b on the surface membrane of PMNs. Extensive release of cytokines IL-1, IL-6, MCP-1, and MIP-2 was observed in BAL fluid and blood plasma during 24 h postexposure. We conclude that EPR monitoring of blood (Fe3+)TRF can be a useful approach for assessment of systemic pro-inflammatory alterations due to SW-induced lung injury.


Subject(s)
Blast Injuries/immunology , Disease Models, Animal , High-Energy Shock Waves , Iron/blood , Lung Injury , Air Pressure , Animals , Biomarkers/blood , Blast Injuries/pathology , Bronchoalveolar Lavage Fluid/cytology , CD11b Antigen/metabolism , Cold Temperature , Cytokines/blood , Cytokines/metabolism , Electron Spin Resonance Spectroscopy , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Granulocytes/immunology , Granulocytes/pathology , Leukocytes, Mononuclear/metabolism , Lung/immunology , Lung/pathology , Lymphocyte Count , Rats , Rats, Sprague-Dawley , Time Factors , Transferrin/metabolism
20.
J Hand Surg Am ; 29(6): 1020-7, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15576210

ABSTRACT

PURPOSE: Little is known about bone healing after composite tissue transplantation that requires pharmacologic immunosuppression. Bone integration and callus development were assessed in bilateral hand transplantation. METHODS: In this study the course of callus development and callus maturation were assessed by color Doppler sonography and radiography in a double hand transplant and compared with forearm replantation. RESULTS: After hand transplantation, ingrowth of small vessels at the bone junction was observed at week 3, calcified callus became visible at month 4, and bone union was completed at month 11. A similar time course of bone integration was observed after replantation. Plating offered sufficient stability. A recipient periostal flap is thought to have improved blood supply and favored development and induction of callus. CONCLUSIONS: Bone healing after hand transplantation under immunosuppression with tacrolimus, mycophenolate mofetil, and prednisolone is identical to that after forearm replantation.


Subject(s)
Blast Injuries/surgery , Forearm Injuries/surgery , Fracture Healing/drug effects , Hand Injuries/surgery , Hand Transplantation , Immunosuppressive Agents/therapeutic use , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Prednisolone/therapeutic use , Replantation , Tacrolimus/therapeutic use , Amputation, Traumatic/diagnostic imaging , Amputation, Traumatic/immunology , Amputation, Traumatic/surgery , Blast Injuries/diagnostic imaging , Blast Injuries/immunology , Bone Regeneration/drug effects , Bony Callus/blood supply , Bony Callus/diagnostic imaging , Bony Callus/drug effects , Bony Callus/immunology , Drug Therapy, Combination , Follow-Up Studies , Forearm Injuries/diagnostic imaging , Forearm Injuries/immunology , Hand/blood supply , Hand Injuries/diagnostic imaging , Hand Injuries/immunology , Humans , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Mycophenolic Acid/adverse effects , Neovascularization, Physiologic/drug effects , Prednisolone/adverse effects , Surgical Flaps/blood supply , Tacrolimus/adverse effects , Ultrasonography, Doppler
SELECTION OF CITATIONS
SEARCH DETAIL
...