Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
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.
Brain Inj ; 36(5): 652-661, 2022 04 16.
Article in English | MEDLINE | ID: mdl-35322723

ABSTRACT

BACKGROUND: Blast traumatic brain injury (TBI) and subconcussive blast exposure have been associated, pathologically, with chronic traumatic encephalopathy (CTE) and, clinically, with cognitive and affective symptoms, but the underlying pathomechanisms of these associations are not well understood. We hypothesized that exosomal microRNA (miRNA) expression, and their relation to neurobehavioral outcomes among Veterans with blunt or blast mild TBI (mTBI) may provide insight into possible mechanisms for these associations and therapeutic targets. METHODS: This is a subanalysis of a larger Chronic Effects of Neurotrauma Consortium Biomarker Discovery Project. Participants (n = 152) were divided into three groups: Controls (n = 35); Blunt mTBI only (n = 54); and Blast/blast+blunt mTBI (n = 63). Postconcussive and post-traumatic stress symptoms were evaluated using the NSI and PCL-5, respectively. Exosomal levels of 798 miRNA expression were measured. RESULTS: In the blast mTBI group, 23 differentially regulated miRNAs were observed compared to the blunt mTBI group and 23 compared to controls. From the pathway analysis, significantly dysregulated miRNAs in the blast exposure group correlated with inflammatory, neurodegenerative, and androgen receptor pathways. DISCUSSION: Our findings suggest that chronic neurobehavioral symptoms after blast TBI may pathomechanistically relate to dysregulated cellular pathways involved with neurodegeneration, inflammation, and central hormonal regulation.


Subject(s)
Blast Injuries , Brain Concussion , Brain Injuries, Traumatic , MicroRNAs , Stress Disorders, Post-Traumatic , Veterans , Blast Injuries/complications , Blast Injuries/genetics , Blast Injuries/psychology , Brain Concussion/complications , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/genetics , Explosions , Humans , MicroRNAs/genetics , Stress Disorders, Post-Traumatic/complications , Veterans/psychology
3.
Brain Res ; 1770: 147642, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34474000

ABSTRACT

Disruption of the blood-brain barrier (BBB) is a critical component of traumatic brain injury (TBI) progression. However, further research into the mechanism of BBB disruption and its specific role in TBI pathophysiology is necessary. To help make progress in elucidating TBI affected BBB pathophysiology, we report herein relative gene expression of eleven TBI biomarkers and other factors of neuronal function in human brain microvascular cells (HBMVEC), one of the main cell types in the BBB. Our in-vitro blast TBI model employs a custom acoustic shock tube to deliver injuries of varying intensities to HBMVECs in culture. Each of the investigated genes exhibit a significant change in expression as a response to TBI, which is dependent on both the injury intensity and time following the injury. This data suggests that cell signaling of HBMVECs could be essential to understanding the interaction of the BBB and TBI pathophysiology, warranting future investigation.


Subject(s)
Blast Injuries/metabolism , Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Gene Expression , Biomarkers/metabolism , Blast Injuries/genetics , Blast Injuries/pathology , Blood-Brain Barrier/pathology , Brain/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Endothelial Cells/pathology , Humans
4.
Med Sci Monit ; 27: e928676, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33621218

ABSTRACT

BACKGROUND Different responses to identical trauma may be related to the genetic background of individuals, but the molecular mechanism is unclear. In this study we investigated the heterogeneity of trauma in mice and the potential biological explanations for the differences. MATERIAL AND METHODS Compared with other organs, the pathological response of the lung after injury is the earliest and most serious. We used C57BL/6 and BALB/C mice to explore the genetic background of different responses to trauma in the lung. We measured mortality rate, pulmonary microvascular permeability, and Cxcl15 gene expression in BALB/C and C57BL/6 mice before and after blast-wave injury. Microvascular permeability was measured using a fluorescent tracer, and Cxcl15 gene expression level and expression distribution were measured using fluorogenic probe quantitative polymerase chain reaction and northern blot. RESULTS C57BL/6 mice showed lower mortality rates and pulmonary microvascular permeability than BALB/C mice after blast-wave injury; there was no significant difference in the permeability before blast-wave injury. The Cxcl15 gene was expressed specifically in the lung tissue of mice. The level of Cxcl15 expression in BALB/C mice was higher than in C57BL/6 mice before and after injury, and the variation trend of Cxcl15 expression level after injury was significantly different between BALB/C and C57BL/6 mice. CONCLUSIONS Our results indicated that BALB/C and C57BL/6 mice had significant heterogeneity in posttraumatic response in terms of mortality and degree of lung damage. The differences in genetic factors such as Cxcl15 may have played a role in this heterogeneity.


Subject(s)
Lung Injury/physiopathology , Lung/pathology , Wounds and Injuries/genetics , Animals , Blast Injuries/genetics , Blast Injuries/physiopathology , Capillary Permeability/genetics , Capillary Permeability/physiology , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Gene Expression/genetics , Lung/metabolism , Lung Injury/genetics , Lung Injury/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
5.
Chin J Traumatol ; 23(5): 249-257, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32917472

ABSTRACT

PURPOSE: Blast lung injury (BLI) is the most common damage resulted from explosion-derived shock wave in military, terrorism and industrial accidents. However, the molecular mechanisms underlying BLI induced by shock wave are still unclear. METHODS: In this study, a goat BLI model was established by a fuel air explosive power. The key genes involved in were identified. The goats of the experimental group were fixed on the edge of the explosion cloud, while the goats of the control group were 3 km far away from the explosive environment. After successful modeling for 24 h, all the goats were sacrificed and the lung tissue was harvested for histopathological observation and RNA sequencing. Gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) analysis were performed to identify the main enriched biological functions of differentially expressed genes (DEGs). Quantitative real-time polymerase chain reaction (qRT-PCR) was used to verify the consistency of gene expression. RESULTS: Of the sampled goat lungs, 895 genes were identified to be significantly differentially expressed, and they were involved in 52 significantly enriched GO categories. KEGG analysis revealed that DEGs were highly enriched in 26 pathways, such as cytokine-cytokine receptor interaction, antifolate resistance, arachidonic acid metabolism, amoebiasis and bile secretion, JAK-STAT, and IL-17 signaling pathway. Furthermore, 15 key DEGs involved in the biological processes of BLI were confirmed by qRT-PCR, and the results were consistent with RNA sequencing. CONCLUSION: Gene expression profiling provide a better understanding of the molecular mechanisms of BLI, which will help to set strategy for treating lung injury and preventing secondary lung injury induced by shock wave.


Subject(s)
Blast Injuries/genetics , Gene Expression Profiling/methods , High-Energy Shock Waves/adverse effects , Lung Injury/genetics , Transcriptome , Animals , Blast Injuries/etiology , Disease Models, Animal , Goats , Lung Injury/etiology , Male , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA
6.
J Neurotrauma ; 37(10): 1221-1232, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31621494

ABSTRACT

Injuries from exposure to explosions rose dramatically during the Iraq and Afghanistan wars, which motivated investigations of blast-related neurotrauma and operational breaching. In this study, military "breachers" were exposed to controlled, low-level blast during a 10-day explosive breaching course. Using an omics approach, we assessed epigenetic, transcriptional, and inflammatory profile changes in blood from operational breaching trainees, with varying levels of lifetime blast exposure, along with daily self-reported symptoms (with tinnitus, headaches, and sleep disturbances as the most frequently reported). Although acute exposure to blast did not confer epigenetic changes, specifically in DNA methylation, differentially methylated regions (DMRs) with coordinated gene expression changes associated with lifetime cumulative blast exposures were identified. The accumulative effect of blast showed increased methylation of PAX8 antisense transcript with coordinated repression of gene expression, which has been associated with sleep disturbance. DNA methylation analyses conducted in conjunction with reported symptoms of tinnitus in the low versus high blast incidents groups identified DMRS in KCNE1 and CYP2E1 genes. KCNE1 and CYP2E1 showed the expected inverse correlation between DNA methylation and gene expression, which have been previously implicated in noise-related hearing loss. Although no significant transcriptional changes were observed in samples obtained at the onset of the training course relative to chronic cumulative blast, we identified a large number of transcriptional perturbations acutely pre- versus post-blast exposure. Acutely, 67 robustly differentially expressed genes (fold change ≥1.5), including UFC1 and YOD1 ubiquitin-related proteins, were identified. Inflammatory analyses of cytokines and chemokines revealed dysregulation of MCP-1, GCSF, HGF, MCSF, and RANTES acutely after blast exposure. These data show the importance of an omics approach, revealing that transcriptional and inflammatory biomarkers capture acute low-level blast overpressure exposure, whereas DNA methylation marks encapsulate chronic long-term symptoms.


Subject(s)
Blast Injuries/blood , Blast Injuries/genetics , Cytokines/blood , Inflammation Mediators/blood , Military Personnel , Adult , Biomarkers/blood , Blast Injuries/psychology , Cytokines/genetics , DNA Methylation/physiology , Explosions , Humans , Male , Military Personnel/psychology , Sequence Analysis, RNA/methods , Time Factors , Transcription, Genetic/physiology
7.
Oxid Med Cell Longev ; 2019: 8460290, 2019.
Article in English | MEDLINE | ID: mdl-31885821

ABSTRACT

Blast exposure is a worldwide public health concern, but most related research has been focused on direct injury. Thoracic blast exposure-induced neurotrauma is a type of indirect injuries where research is lacking. As CD28 stimulates T cell activation and survival and contributes to inflammation initiation, it may play a role in thoracic blast exposure-induced neurotrauma. However, it has not been investigated. To explore the effects of CD28 on thoracic blast exposure-induced brain injury and its potential molecular mechanisms, a mouse model of thoracic blast exposure-induced brain injury was established. Fifty C57BL/6 wild-type (WT) and fifty CD28 knockout (CD28-/-) mice were randomly divided into five groups (one control group and four model groups), with ten mice (from each of the two models) for each group. Lung and brain tissue and serum samples were collected at 12 h, 24 h, 48 h, and 1 week after thoracic blast exposure. Histopathological changes were detected by hematoxylin-eosin staining. The expressions of inflammatory-related factors were detected by ELISA. Oxidative stress in the brain tissue was evaluated by determining the generation of reactive oxygen species (ROS) and the expressions of thioredoxin (TRX), malondialdehyde (MDA), SOD-1, and SOD-2. Apoptosis in the brain tissue was evaluated by TUNEL staining and the levels of Bax, Bcl-xL, Bad, Cytochrome C, and caspase-3. In addition, proteins of related pathways were also studied by western blotting and immunofluorescence. We found that CD28 deficiency significantly reduced thoracic blast exposure-induced histopathological changes and decreased the levels of inflammatory-related factors, including IL-1ß, TNF-α, and S100ß. In the brain tissue, CD28 deficiency also significantly attenuated thoracic blast exposure-induced generation of ROS and expressions of MDA, TRX, SOD-1, and SOD-2; lowered the number of apoptotic cells and the expression of Bax, cleaved caspase-3, Cytochrome C, and Bad; and maintained Bcl-xL expression. Additionally, CD28 deficiency significantly ameliorated thoracic blast exposure-induced increases of p-PI3K and Keap1 and the decrease of Nrf2 expression in the brain. Our results indicate that CD28 deficiency has a protective effect on thoracic blast exposure-induced brain injury that might be associated with the PI3K/Nrf2/Keap1 signaling pathway.


Subject(s)
Blast Injuries/metabolism , Brain/physiology , CD28 Antigens/metabolism , Thoracic Injuries/metabolism , Animals , Apoptosis/genetics , Blast Injuries/genetics , CD28 Antigens/genetics , Disease Models, Animal , Humans , Inflammation Mediators/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidative Stress/genetics , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Thoracic Injuries/genetics
8.
Psychiatry Res ; 280: 112519, 2019 10.
Article in English | MEDLINE | ID: mdl-31442670

ABSTRACT

The first of its kind, this study determined whether blast exposure interacts with genetic variant 5HTTLPR to predict posttraumatic stress (PTS) symptoms in 78 military explosives operators. In all models, blast-exposed 5HTTLPR S carriers registered definitively higher PTS symptoms in comparison to non-exposed S carriers, as well as exposed and non-exposed LL carriers (all p < 0.01). All findings were robust to confounding influences of age and traumatic brain injury diagnosis. Not only is blast exposure prevalent in EOD personnel, but it also interacts with genetic predisposition to predict trauma symptoms in this unique, at-risk military population.


Subject(s)
Blast Injuries/genetics , Blast Injuries/psychology , Military Personnel/psychology , Serotonin Plasma Membrane Transport Proteins/genetics , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/psychology , Adult , Blast Injuries/epidemiology , Brain Injuries, Traumatic/epidemiology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/psychology , Explosive Agents/adverse effects , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Genetic Predisposition to Disease/psychology , Genetic Variation/genetics , Humans , Male , Predictive Value of Tests , Stress Disorders, Post-Traumatic/epidemiology
9.
Mol Vis ; 25: 283-294, 2019.
Article in English | MEDLINE | ID: mdl-31263351

ABSTRACT

Purpose: Our study aimed to determine whether the altered expression of biomarkers linked to corneal injuries, such as the edema-regulating proteins aquaporin-1 and aquaporin-5 (AQP1 and AQP5), occurred following primary blast exposure. Methods: Adult male Dutch Belted rabbits were anesthetized and exposed to blast waves with peak overpressures of 142.5-164.1 kPa (20.4-23.4 psi). These exposure groups experienced peak blast overpressure-specific impulses (impulse per unit surface area) of 199.6-228.5 kPa-ms. Unexposed rabbits were included as controls. The animals were euthanized at 48 h post-exposure. Corneas obtained from the euthanized blast-exposed and control rabbits were processed for quantitative PCR and western blot to quantify mRNA and the protein expression of AQP1 and AQP5. Immunohistochemical analysis was conducted to determine the cellular localization of AQP1 and AQP5. Results: Corneal thickness increased up to 18% with the peak blast overpressure-specific impulses of 199.6-228.5 kPa-ms at 48 h after blast exposure. mRNA levels of AQP1 and AQP5 increased in the whole cornea lysates of blast-exposed rabbits relative to those of the controls. Western blot analyses of whole cornea lysates revealed that the expression levels of AQP1 and AQP5 were approximately 2- and 1.5-fold higher, respectively, in blast-exposed rabbits compared to controls. The extent of AQP1 immunostaining (AQP1-IS) increased in the epithelial cell layer after blast exposure. The AQP5-IS pattern changed from a mixed membrane and cytoplasmic expression in the controls to predominantly cytoplasmic expression in the basally located cornea epithelial cells of blast-exposed rabbits. Conclusions: Primary blast exposure resulted in edema-related changes in the cornea manifested by the altered expression of the edema-regulating proteins AQP1 and AQP5 with blast overpressure-specific impulses. These findings support potential acute corneal injury mechanisms in which the altered regulation of water permeability is caused by primary blast exposure.


Subject(s)
Aquaporin 1/genetics , Aquaporin 5/genetics , Blast Injuries/genetics , Cornea/metabolism , Corneal Injuries/genetics , Gene Expression Regulation , Animals , Aquaporin 1/metabolism , Aquaporin 5/metabolism , Blast Injuries/pathology , Cornea/pathology , Corneal Injuries/pathology , Corneal Pachymetry , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Slit Lamp
10.
Biofactors ; 45(4): 517-535, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31206893

ABSTRACT

Traumatic brain injury (TBI) is the most prevalent health problem affecting all age groups, and leads to many secondary problems in other organs especially kidneys, gastrointestinal tract, and heart function. In this review, the search terms were TBI, fluid percussion injury, cold injury, weight drop impact acceleration injury, lateral fluid percussion, cortical impact injury, and blast injury. Studies with Actaea racemosa, Artemisia annua, Aframomum melegueta, Carthamus tinctorius, Cinnamomum zeylanicum, Crocus sativus, Cnidium monnieri, Curcuma longa, Gastrodia elata, Malva sylvestris, Da Chuanxiong Formula, Erigeron breviscapus, Panax ginseng, Salvia tomentosa, Satureja khuzistanica, Nigella sativa, Drynaria fortune, Dracaena cochinchinensis, Polygonum cuspidatum, Rosmarinus officinalis, Rheum tanguticum, Centella asiatica, and Curcuma zedoaria show a significant decrease in neuronal injury by different mechanisms such as increasing superoxide dismutase and catalase activities, suppressing nuclear factor kappa B (NF-κB), interleukin 1 (IL-1), glial fibrillary acidic protein, and IL-6 expression. The aim of this study was to evaluate the neuroprotective effects of medicinal plants in central nervous system pathologies by reviewing the available literature.


Subject(s)
Blast Injuries/prevention & control , Brain Injuries, Traumatic/prevention & control , Cold Injury/prevention & control , Gene Expression Regulation/drug effects , Neuroprotective Agents/pharmacology , Plants, Medicinal/chemistry , Animals , Blast Injuries/genetics , Blast Injuries/metabolism , Blast Injuries/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Catalase/genetics , Catalase/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Cold Injury/genetics , Cold Injury/metabolism , Cold Injury/pathology , Disease Models, Animal , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Neuroprotective Agents/isolation & purification , Rats , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
11.
J Neurotrauma ; 36(23): 3264-3273, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31232163

ABSTRACT

Evidence suggests that blast exposure has profound negative consequences for the health of the human brain, and that it may confer risk for the development of neurodegenerative diseases such as chronic traumatic encephalopathy and Alzheimer's disease (AD). Although the molecular mechanisms linking blast exposure to subsequent neurodegeneration is an active focus of research, recent studies suggest that genetic risk for AD may elevate the risk of neurodegeneration following traumatic brain injury (TBI). However, it is currently unknown if blast exposure also interacts with AD risk to promote neurodegeneration. In this study we examined whether apolipoprotein (APOE) ɛ4, a well-known genetic risk factor for AD, influenced the relationship between blast exposure and white matter integrity in a cohort of 200 Iraq and Afghanistan war veterans. Analyses revealed a significant interaction between close-range blast exposure (CBE) (close range being within 10 m) and APOE ɛ4 carrier status in predicting white matter abnormalities, measured by a voxelwise cluster-based method that captures spatial heterogeneity in white matter disruptions. This interaction remained significant after controlling for TBI, pointing to the specificity of CBE and APOE in white matter disruptions. Further, among veteran ɛ4 carriers exposed to close-range blast, we observed a positive association between the number of CBEs and the number of white matter abnormalities. These results raise the possibility that CBE interacts with AD genetic influences on neuropathological processes such as the degradation of white matter integrity.


Subject(s)
Apolipoprotein E4/genetics , Blast Injuries/diagnostic imaging , Blast Injuries/genetics , Heterozygote , Veterans , White Matter/diagnostic imaging , Adult , Afghan Campaign 2001- , Cross-Sectional Studies , Female , Humans , Iraq War, 2003-2011 , Male , Young Adult
12.
Biomarkers ; 24(2): 166-179, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30280938

ABSTRACT

CONTEXT: Due to the wide use of improvised explosive devices during modern warfare, primary blast-derived mild traumatic brain injury (mTBI) has become a major medical condition in the military. With minimal visually identifiable symptoms, an effective molecular biomarker system is desirable. OBJECTIVE: We assessed the potential of mammalian hair follicle miRNAs as an mTBI biomarker. MATERIALS AND METHODS: Due to their well-established roles in mTBI molecular pathology, the expression level of miR-183, miR-26a, miR-181c, miR-29a, miR-34a and miR-27b was determined using qRT-PCR in whisker hair follicles from rats subject to head-only exposure to a single-pulse shock wave. Based on established transcriptomics profiles, sub-network enrichment analysis (SNEA) was also conducted. RESULTS: The results revealed that molecular networks involving miR-183, miR-26a and miR-181c were enriched in multiple treatments, whereas sub-networks of miR-29a, miR-34a and miR-27b were unique to individual exposure groups. DISCUSSION: Our study showed that all six miRNAs were reflective of the mTBI signature involved in cellular responses. Noteworthy was that the decrease in the transcript levels of miR-181c was correlated with shockwave exposure severity. CONCLUSION: This study demonstrates for the first time that mammalian hair follicles are capable of housing miRNA biomarkers for TBI.


Subject(s)
Biomarkers/metabolism , Blast Injuries/genetics , Brain Concussion/genetics , Hair Follicle/metabolism , MicroRNAs/genetics , Animals , Blast Injuries/pathology , Brain Concussion/pathology , Disease Models, Animal , Humans , MicroRNAs/metabolism , Rats
13.
Bone ; 117: 149-160, 2018 12.
Article in English | MEDLINE | ID: mdl-30219480

ABSTRACT

Trauma-induced heterotopic ossification is an intriguing phenomenon involving the inappropriate ossification of soft tissues within the body such as the muscle and ligaments. This inappropriate formation of bone is highly prevalent in those affected by blast injuries. Here, we developed a simplified cell culture model to evaluate the molecular events involved in heterotopic ossification onset that arise from the shock wave component of the disease. We exposed three subtypes of human mesenchymal cells in vitro to a single, high-energy shock wave and observed increased transcription in the osteogenic master regulators, Runx2 and Dlx5, and significantly accelerated cell mineralisation. Reduced representation bisulfite sequencing revealed that the shock wave altered methylation of gene promoters, leading to opposing changes in gene expression. Using a drug to target ITGAV, whose expression was perturbed by the shock wave, we found that we could abrogate the deposition of mineral in our model. These findings show how new therapeutics for the treatment of heterotopic ossification can be identified using cell culture models.


Subject(s)
Blast Injuries/pathology , Cell Differentiation , Demethylation , Integrin alphaV/metabolism , Models, Biological , Ossification, Heterotopic/pathology , Osteogenesis , Blast Injuries/genetics , Calcification, Physiologic/genetics , Cell Culture Techniques , Cell Nucleus/pathology , DNA Methylation , High-Energy Shock Waves , Humans , Promoter Regions, Genetic/genetics
14.
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
15.
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
16.
Brain Inj ; 32(1): 123-134, 2018.
Article in English | MEDLINE | ID: mdl-29157017

ABSTRACT

OBJECTIVE: Primary blast-induced mild traumatic brain injury (mTBI) is an injury experienced during modern warfare due to exposure to the pressure waves produced by the detonation of explosives. With virtually no apparent physical damage or symptoms presented, there is a need for more objective and accessible mTBI biomarkers posing minimal invasiveness risk. METHODS: We measured the transcriptomic sensitivity of the hair follicles in relation to the severity of primary blast-derived TBI. An Advanced Blast Simulator system was used to expose male rats to single pulse shock waves (intensities ranging from 15 to 30 psi) in a head-only fashion. Gene differential expression (DE) and gene set (GS) analyses were conducted in the rat whisker hair follicles and the whole blood samples. RESULTS: While shared cellular function, themes were found across the exposure groups, some gene sets under such themes were unique to the exposure conditions. Intensity-specific pathway enrichment patterns within shared GS themes were also identified. Furthermore, while exhibited shared pathways, the blood transcriptome showed substantially fewer enriched gene sets compared with the hair follicles across all exposure conditions. CONCLUSIONS: Accordingly, we demonstrate the potential of mammalian hair follicles serving as an additional source for biomarker discovery and for diagnosing mTBI with high accessibility.


Subject(s)
Blast Injuries/genetics , Brain Concussion/genetics , Hair Follicle/metabolism , Transcriptome , Animals , Biomarkers/metabolism , Blast Injuries/metabolism , Brain Concussion/metabolism , Disease Models, Animal , Explosions , High-Energy Shock Waves , Male , Rats
17.
Amino Acids ; 49(5): 871-886, 2017 05.
Article in English | MEDLINE | ID: mdl-28161798

ABSTRACT

This study investigated the benefit of ß-alanine (BA) supplementation on behavioral and cognitive responses relating to mild traumatic brain injury (mTBI) and post-traumatic stress disorder (PTSD) in rats exposed to a low-pressure blast wave. Animals were fed a normal diet with or without (PL) BA supplementation (100 mg kg-1) for 30-day, prior to being exposed to a low-pressure blast wave. A third group of animals served as a control (CTL). These animals were fed a normal diet, but were not exposed to the blast. Validated cognitive-behavioral paradigms were used to assess both mTBI and PTSD-like behavior on days 7-14 following the blast. Brain-derived neurotrophic factor (BDNF), neuropeptide Y, glial fibrillary acidic protein (GFAP) and tau protein expressions were analyzed a day later. In addition, brain carnosine and histidine content was assessed as well. The prevalence of animals exhibiting mTBI-like behavior was significantly lower (p = 0.044) in BA than PL (26.5 and 46%, respectively), but no difference (p = 0.930) was noted in PTSD-like behavior between the groups (10.2 and 12.0%, respectively). Carnosine content in the cerebral cortex was higher (p = 0.048) for BA compared to PL, while a trend towards a difference was seen in the hippocampus (p = 0.058) and amygdala (p = 0.061). BDNF expression in the CA1 subregion of PL was lower than BA (p = 0.009) and CTL (p < 0.001), while GFAP expression in CA1 (p = 0.003) and CA3 (p = 0.040) subregions were higher in PL than other groups. Results indicated that BA supplementation for 30-day increased resiliency to mTBI in animals exposed to a low-pressure blast wave.


Subject(s)
Blast Injuries/metabolism , Brain Injuries/metabolism , Dietary Supplements , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/prevention & control , beta-Alanine/administration & dosage , Animals , Blast Injuries/genetics , Blast Injuries/physiopathology , Brain Chemistry , Brain Injuries/genetics , Brain Injuries/physiopathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Carnosine/metabolism , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Histidine/metabolism , Male , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Rats , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/physiopathology , tau Proteins/genetics , tau Proteins/metabolism
18.
PLoS One ; 9(8): e104518, 2014.
Article in English | MEDLINE | ID: mdl-25136963

ABSTRACT

With wide adoption of explosive-dependent weaponry during military activities, Blast-induced neurotrauma (BINT)-induced traumatic brain injury (TBI) has become a significant medical issue. Therefore, a robust and accessible biomarker system is in demand for effective and efficient TBI diagnosis. Such systems will also be beneficial to studies of TBI pathology. Here we propose the mammalian hair follicles as a potential candidate. An Advanced Blast Simulator (ABS) was developed to generate shock waves simulating traumatic conditions on brains of rat model. Microarray analysis was performed in hair follicles to identify the gene expression profiles that are associated with shock waves. Gene set enrichment analysis (GSEA) and sub-network enrichment analysis (SNEA) were used to identify cell processes and molecular signaling cascades affected by simulated bomb blasts. Enrichment analyses indicated that genes with altered expression levels were involved in central nervous system (CNS)/peripheral nervous system (PNS) responses as well as signal transduction including Ca2+, K+-transportation-dependent signaling, Toll-Like Receptor (TLR) signaling and Mitogen Activated Protein Kinase (MAPK) signaling cascades. Many of the pathways identified as affected by shock waves in the hair follicles have been previously reported to be TBI responsive in other organs such as brain and blood. The results suggest that the hair follicle has some common TBI responsive molecular signatures to other tissues. Moreover, various TBI-associated diseases were identified as preferentially affected using a gene network approach, indicating that the hair follicle may be capable of reflecting comprehensive responses to TBI conditions. Accordingly, the present study demonstrates that the hair follicle is a potentially viable system for rapid and non-invasive TBI diagnosis.


Subject(s)
Blast Injuries/genetics , Brain Injuries/genetics , Brain/metabolism , Hair Follicle/metabolism , Transcriptome , Animals , Biomarkers/metabolism , Blast Injuries/diagnosis , Blast Injuries/pathology , Brain/pathology , Brain Injuries/diagnosis , Brain Injuries/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Disease Models, Animal , Explosions , Gene Expression , Gene Expression Profiling , High-Energy Shock Waves , Male , Metabolic Networks and Pathways/genetics , Microarray Analysis , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
19.
J Neurotrauma ; 30(11): 920-37, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23496248

ABSTRACT

Abstract Explosive blast-induced traumatic brain injury (TBI) is the signature insult in modern combat casualty care and has been linked to post-traumatic stress disorder, memory loss, and chronic traumatic encephalopathy. In this article we report on blast-induced mild TBI (mTBI) characterized by fiber-tract degeneration and axonal injury revealed by cupric silver staining in adult male rats after head-only exposure to 35 psi in a helium-driven shock tube with head restraint. We now explore pathways of secondary injury and repair using biochemical/molecular strategies. Injury produced ∼25% mortality from apnea. Shams received identical anesthesia exposure. Rats were sacrificed at 2 or 24 h, and brain was sampled in the hippocampus and prefrontal cortex. Hippocampal samples were used to assess gene array (RatRef-12 Expression BeadChip; Illumina, Inc., San Diego, CA) and oxidative stress (OS; ascorbate, glutathione, low-molecular-weight thiols [LMWT], protein thiols, and 4-hydroxynonenal [HNE]). Cortical samples were used to assess neuroinflammation (cytokines, chemokines, and growth factors; Luminex Corporation, Austin, TX) and purines (adenosine triphosphate [ATP], adenosine diphosphate, adenosine, inosine, 2'-AMP [adenosine monophosphate], and 5'-AMP). Gene array revealed marked increases in astrocyte and neuroinflammatory markers at 24 h (glial fibrillary acidic protein, vimentin, and complement component 1) with expression patterns bioinformatically consistent with those noted in Alzheimer's disease and long-term potentiation. Ascorbate, LMWT, and protein thiols were reduced at 2 and 24 h; by 24 h, HNE was increased. At 2 h, multiple cytokines and chemokines (interleukin [IL]-1α, IL-6, IL-10, and macrophage inflammatory protein 1 alpha [MIP-1α]) were increased; by 24 h, only MIP-1α remained elevated. ATP was not depleted, and adenosine correlated with 2'-cyclic AMP (cAMP), and not 5'-cAMP. Our data reveal (1) gene-array alterations similar to disorders of memory processing and a marked astrocyte response, (2) OS, (3) neuroinflammation with a sustained chemokine response, and (4) adenosine production despite lack of energy failure-possibly resulting from metabolism of 2'-3'-cAMP. A robust biochemical/molecular response occurs after blast-induced mTBI, with the body protected from blast and the head constrained to limit motion.


Subject(s)
Blast Injuries/metabolism , Brain Injuries/metabolism , Transcriptome , Animals , Blast Injuries/genetics , Blast Injuries/physiopathology , Brain Injuries/genetics , Brain Injuries/physiopathology , Disease Models, Animal , Gene Expression Profiling , Male , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Regeneration/physiology , Oligonucleotide Array Sequence Analysis , Rats , Rats, Sprague-Dawley
20.
Chem Biol Interact ; 203(1): 371-5, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23159883

ABSTRACT

Cholinergic activity has been recognized as a major regulatory component of stress responses after traumatic brain injury (TBI). Centrally acting acetylcholinesterase (AChE) inhibitors are also being considered as potential therapeutic candidates against TBI mediated cognitive impairments. We have evaluated the expression of molecules involved in cholinergic and inflammatory pathways in various regions of brain after repeated blast exposures in mice. Isoflurane anesthetized C57BL/6J mice were restrained and placed in a prone position transverse to the direction of the shockwaves and exposed to three 20.6 psi blast overpressures with 1-30 min intervals. Brains were collected at the 6h time point after the last blast exposure and subjected to cDNA microarray and microRNA analysis. cDNA microarray analysis showed significant changes in the expression of cholinergic (muscarinic and nicotinic) and gammaaminobutyric acid and glutamate receptors in the midbrain region along with significant changes in multiple genes involved in inflammatory pathways in various regions of the brain. MicroRNA analysis of cerebellum revealed differential expression of miR-132 and 183, which are linked to cholinergic anti-inflammatory signaling, after blast exposure. Changes in the expression of myeloperoxidase in the cerebellum were confirmed by Western blotting. These results indicate that early pathologic progression of blast TBI involves dysregulation of cholinergic and inflammatory pathways related genes. Acute changes in molecules involved in the modulation of cholinergic and inflammatory pathways after blast TBI can cause long-term central and peripheral pathophysiological changes.


Subject(s)
Acetylcholine/metabolism , Blast Injuries/metabolism , Brain Injuries/metabolism , Inflammation Mediators/metabolism , Acetylcholinesterase/metabolism , Animals , Blast Injuries/genetics , Brain/metabolism , Brain Injuries/genetics , Cerebellum/injuries , Cerebellum/metabolism , Disease Progression , GPI-Linked Proteins/metabolism , Gene Expression , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...