Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Comp Med ; 74(3): 179-185, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38649263

RESUMEN

Chinchillas are a relatively novel research model compared with other rodent species. They require special considerations when it comes to their husbandry and daily care. Chinchillas tend to be shy animals that are well adapted to masking clinical signs of illness. These characteristics can make them a difficult species to maintain in a research setting. The authors' institution has maintained chinchillas and established standardized daily animal care procedures for them. Chinchillas are most commonly used for auditory research. They are often used to study the mechanism of different induced auditory conditions or injuries as well as exploration for potential alleviating treatments. Often, tested therapeutics have demonstrated potentially beneficial effects but have not been applied in the specific condition or injury of interest. The development of new applications for therapeutics can lead to groundbreaking discoveries, but testing of new therapeutic applications is often initially performed in an animal model without knowing how the therapeutic will behave in the species. During testing, unexpected adverse events may manifest that require more focused monitoring and supportive care. This scenario occurred when adverse effects were observed in a chinchilla blast-injury model after receiving an acylated glucagon-like peptide-1 (GLP-1) receptor agonist. The study involved evaluation of this therapeutic over an extended amount of time after inducing a controlled pressurized blast-injury followed by multiple repeated hearing tests under anesthesia. Chinchillas enrolled in the study exhibited several clinical signs including weight loss, lethargy, labored breathing, neurologic abnormalities, decreased appetite or decreased fecal output, and otitis. Five primary abnormalities were reported on pathology: aspiration pneumonia, hepatic steatosis, right ventricular dilation, pancreatitis, and tubulointerstitial nephritis. Initially abnormal clinical signs, early mortality rates, and pathology were attributed to multiple anesthetic events. However, a retrospective analysis evaluating the association of different study variable exposures in a stratified comparison demonstrated that the early mortality rates were actually associated with the therapeutic drug given for the first time in this species. In this study, we describe the detailed findings of the retrospective analysis and explore different strategies that can be incorporated to maintain good animal welfare and decrease early animal loss.


Asunto(s)
Traumatismos por Explosión , Chinchilla , Modelos Animales de Enfermedad , Liraglutida , Animales , Traumatismos por Explosión/tratamiento farmacológico , Traumatismos por Explosión/patología , Liraglutida/farmacología , Liraglutida/uso terapéutico , Estudios Retrospectivos , Masculino
2.
Mil Med ; 188(Suppl 6): 271-279, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37948226

RESUMEN

INTRODUCTION: Combat injuries are complex and multimodal. Most injuries to the extremities occur because of explosive devices such as improvised explosive devices. Blast exposure dramatically increases the risk of infection in combat wounds, and there is limited available information on the best antibiotic treatments for these injuries. We previously demonstrated that mice exposed to blast displayed a delayed clearance of cefazolin from the plasma and liver; further semi-mechanistic modeling determined that cefazolin concentrations in the skin of these mice were reduced. Our objective was to investigate the effects of blast on the pharmacokinetics of antibiotics of different types used for the treatment of combat wounds in the rat model. MATERIALS AND METHODS: Male Sprague Dawley rats were exposed to blast overpressure followed by injection of a bolus of animal equivalent doses of an antibiotic (cefazolin, cefepime, ertapenem, or clindamycin) into the tail vein at 1-hour post-blast exposure. Blood was collected at predetermined time points via repeated sampling from the tail vein. Animals were also euthanized at predetermined time points, at which time liver, kidney, skin, and blood via cardiac puncture were collected. Antibiotic concentrations were determined by ultra-performance liquid chromatography-tandem mass spectrometry. RESULTS: Blast-exposed rats exhibited a similar rate of clearance compared to non-blasted rats in the blood, liver, kidney, and skin, which is inconsistent with the data regarding cefazolin in blast-exposed mice. CONCLUSIONS: Our results in rats do not recapitulate our previous observation of delayed cefazolin clearance in mice following the blast overpressure exposure. Although using rats permitted us to collect multiple blood samples from the same animals, rats may not be a suitable model for measuring the pharmacokinetics of antibiotics following blast. The interpretation of the results may be challenging because of variation in data among rat subjects in the same sample groups.


Asunto(s)
Antibacterianos , Traumatismos por Explosión , Humanos , Ratas , Masculino , Ratones , Animales , Ratas Sprague-Dawley , Antibacterianos/uso terapéutico , Traumatismos por Explosión/tratamiento farmacológico , Cefazolina/uso terapéutico , Explosiones , Modelos Animales de Enfermedad
3.
Neurochem Int ; 163: 105472, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36599378

RESUMEN

Blast-induced traumatic brain injury (bTBI) is a health concern in military service members who are exposed to multiple blasts throughout their training and deployment. Our group has previously reported decreased long term potentiation (LTP) following repeated bTBI in a rat organotypic hippocampal slice culture (OHSC) model. In this study, we investigated changes in inflammatory markers like cyclooxygenase (COX) and tested the efficacy of COX or prostaglandin EP3 receptor (EP3R) inhibitors in attenuating LTP deficits. Expression of COX-2 was increased 48 h following repeated injury, whereas COX-1 expression was unchanged. EP3R expression was upregulated, and cyclic adenosine monophosphate (cAMP) concentration was decreased after repeated blast exposure. Post-traumatic LTP deficits improved after treatment with a COX-1 specific inhibitor, SC-560, a COX-2 specific inhibitor, rofecoxib, a pan-COX inhibitor, ibuprofen, or an EP3R inhibitor, L-798,106. Delayed treatment with ibuprofen and L-798,106 also prevented LTP deficits. These findings suggest that bTBI induced neuroinflammation may be responsible for some functional deficits that we have observed in injured OHSCs. Additionally, COX and EP3R inhibition may be viable therapeutic strategies to reduce neurophysiological deficits after repeated bTBI.


Asunto(s)
Traumatismos por Explosión , Lesiones Traumáticas del Encéfalo , Ratas , Animales , Ciclooxigenasa 2 , Potenciación a Largo Plazo/fisiología , Ibuprofeno , Traumatismos por Explosión/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Hipocampo/fisiología
4.
Mil Med ; 186(11-12): 1241-1245, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34453163

RESUMEN

A military soldier sustained a blast injury in Afghanistan, resulting in amputations and hemipelvectomy. He developed New Delhi metallo-beta-lactamase-producing E. coli bacteremia, soft-tissue infection, and sacral osteomyelitis. These organisms are being increasingly discovered in different communities around the world. He was successfully treated with tigecycline and cefiderocol. Cefiderocol is a novel siderophore-based cephalosporine developed to treat serious infections, including those caused by carbapenem-resistant Enterobacterales.


Asunto(s)
Traumatismos por Explosión , Carbapenémicos , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Traumatismos por Explosión/tratamiento farmacológico , Carbapenémicos/farmacología , Carbapenémicos/uso terapéutico , Cefalosporinas , Escherichia coli , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Cefiderocol
5.
Life Sci ; 280: 119722, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34153300

RESUMEN

Although melatonin has been demonstrated to exert a potent antioxidant effect, the ability of melatonin to alleviate blast-induced oxidative stress in the hypothalamic-pituitary-gonadal (HPG) axis remains unclear. This study aimed to elucidate the effects and underlying mechanism of melatonin pretreatment on the HPG axis disrupted by blast injury. Sixty C57BL/6 mice were randomly divided into control, blast, and blast + melatonin groups for behavioral experiments. The elevated maze experiment, open field experiment, and Morris Water Maze experiment were carried out on the 7th, 14th and 28th day after the blast injury. Fifty Sprague Dawley rats were randomly divided into control, blast, blast + melatonin, and blast + melatonin + luzindole groups for hormone assays and molecular and pathological experiments. Blood samples were used for HPG axis hormone detection and ELISA assays, and tissue samples were used to detect oxidative stress, inflammation, apoptosis, and stress-related protein levels. The results showed that melatonin pretreatment alleviated blast-induced behavioral abnormalities in mice and maintained the HPG axis hormone homeostasis in rats. Additionally, melatonin significantly reduced MDA5 expression and increased the expression of Nrf2/HO-1. Moreover, melatonin significantly inhibited NF-κB expression and upregulated IL-10 expression, and it reversed the blast-induced high expression of caspase-3 and Bax and the low expression of Bcl-2. Furthermore, luzindole counteracted melatonin inhibition of NF-κB and upregulated Nrf2/HO-1. Melatonin significantly alleviated blast-induced HPG axis hormone dyshomeostasis, behavioral abnormalities, oxidative stress, inflammation, and apoptosis, which may be achieved by upregulating the Nrf2/HO-1 signaling pathway. Our study suggested that melatonin pretreatment is a potential treatment for blast-induced HPG axis hormonal and behavioral abnormalities.


Asunto(s)
Antioxidantes/uso terapéutico , Traumatismos por Explosión/tratamiento farmacológico , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Melatonina/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Antioxidantes/farmacología , Traumatismos por Explosión/metabolismo , Traumatismos por Explosión/patología , Hemo-Oxigenasa 1/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/patología , Masculino , Melatonina/farmacología , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Ratas Sprague-Dawley , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/patología
6.
Neurosci Lett ; 749: 135722, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33592306

RESUMEN

Much attention has been given to effects of repeated exposure to a shock wave as a possible factor causing severe higher brain dysfunction and post-traumatic stress disorder (PTSD)-like symptoms in patients with mild to moderate blast-induced traumatic brain injury (bTBI). However, it is unclear how the repeated exposure and the inter-exposure time affect the brain. In this study, we topically applied low-impulse (∼54 Pa·s) laser-induced shock waves (LISWs; peak pressure, ∼75.7 MPa) to the rat brain once or twice with the different inter-exposure times (15 min, 1 h, 3 h, 24 h and 7 days) and examined anxiety-related behavior and motor dysfunction in the rats as well as expression of ß-amyloid precursor protein (APP) as an axonal damage marker in the brains of the rats. The averaged APP expression scores for the rat brains doubly-exposed to LISWs with inter-exposure times from 15 min to 24 h were significantly higher than those for rats with a single exposure (P < 0.0001). The rats with double exposure to LISWs showed significantly more frequent anxiety-related behavior (P < 0.05) and poorer motor function (P < 0.01) than those of rats with a single exposure. When the inter-exposure time was extended to 7 days, however, the rats showed no significant differences either in axonal damage score or level of motor dysfunction. The results suggest that the cumulative effects of shock wave-related brain injury can be avoided with an appropriate inter-exposure time. However, clinical bTBI occurs in much more complex environments than those in our model. Further study considering other factors, such as the effects of acceleration, is needed to know the clinically-relevant, necessary inter-exposure time.


Asunto(s)
Axones/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Traumatismos por Explosión/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Rayos Láser , Animales , Ansiedad/fisiopatología , Axones/metabolismo , Traumatismos por Explosión/fisiopatología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Rayos Láser/efectos adversos , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/fisiopatología
7.
Laryngoscope ; 131(6): E2018-E2025, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33427310

RESUMEN

OBJECTIVE/HYPOTHESIS: We examined a neuroinflammatory response associated with glial activation in the cochlea exposed to blast overpressure and evaluated the potential therapeutic efficacy of specialized pro-resolving mediators such as neuroprotectin D1, NPD1; (10R, 17S-dihydroxy-4Z, 7Z, 11E, 13E, 15Z, 19Z-docosahexaenoic acid) in a rodent blast-induced auditory injury model. STUDY DESIGN: Animal Research. METHODS: A compressed-air driven shock tube was used to expose anesthetized adult male Long-Evan rats to shock waves simulating an open-field blast exposure. Approximately 30 minutes after blast exposure, rats were treated with NPD1 (100 ng/kg body wt.) or vehicle delivered intravenously via tail vein injection. Rats were then euthanized 48 hours after blast exposure. Unexposed rats were included as controls. Tissue sections containing both middle and inner ear were prepared with hematoxylin-eosin staining to elucidate histopathological changes associated with blast exposure. Cochlear tissues were evaluated for relative expression of ionized calcium-binding adaptor 1 (Iba1), as an indicator of microglial activation by immunohistochemistry and western blot analyses. RESULTS: Our animal model resulted in an acute injury mechanism manifested by damage to the tympanic membrane, hemorrhage, infiltration of inflammatory cells, and increased expression of Iba1 protein. Moreover, therapeutic intervention with NPD1 significantly reduced Iba1 expression in the cochlea, suggesting a reduction of a neuroinflammatory response caused by blast overpressure. CONCLUSIONS: Blast overpressure resulted in an increased expression of proteins involved in gliosis within the auditory system, which were reduced by NPD1. Treatment of NPD1 suggests an effective strategy to reduce or halt auditory microglial cell activation due to primary blast exposure. LEVEL OF EVIDENCE: NA Laryngoscope, 131:E2018-E2025, 2021.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Cóclea/lesiones , Cóclea/metabolismo , Ácidos Docosahexaenoicos/farmacología , Microglía/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Explosiones , Masculino , Proteínas de Microfilamentos/metabolismo , Ratas
8.
Sci Rep ; 10(1): 9420, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32523011

RESUMEN

We investigated the role of nitric oxide synthase (NOS) in mediating blood-brain barrier (BBB) disruption and peripheral immune cell infiltration in the cerebellum following blast exposure. Repetitive, but not single blast exposure, induced delayed-onset BBB disruption (72 hours post-blast) in cerebellum. The NOS inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) administered after blast blocked BBB disruption and prevented CD4+ T-cell infiltration into cerebellum. L-NAME also blocked blast-induced increases in intercellular adhesion molecule-1 (ICAM-1), a molecule that plays a critical role in regulating blood-to-brain immune cell trafficking. Blocking NOS-mediated BBB dysfunction during this acute/subacute post-blast interval (24-71 hours after the last blast) also prevented sensorimotor impairment on a rotarod task 30 days later, long after L-NAME cleared the body. In postmortem brains from Veterans/military Servicemembers with blast-related TBI, we found marked Purkinje cell dendritic arbor structural abnormalities, which were comparable to neuropathologic findings in the blast-exposed mice. Taken collectively, these results indicate that blast provokes delayed-onset of NOS-dependent pathogenic cascades that can later emerge as behavioral dysfunction. These results also further implicate the cerebellum as a brain region vulnerable to blast-induced mTBI.


Asunto(s)
Traumatismos por Explosión/metabolismo , Traumatismos por Explosión/fisiopatología , Conmoción Encefálica/fisiopatología , Enfermedades Cerebelosas/metabolismo , Enfermedades Cerebelosas/fisiopatología , Cerebelo/fisiopatología , Óxido Nítrico Sintasa/metabolismo , Animales , Traumatismos por Explosión/tratamiento farmacológico , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Conmoción Encefálica/tratamiento farmacológico , Conmoción Encefálica/metabolismo , Enfermedades Cerebelosas/tratamiento farmacológico , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster/farmacología , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Células de Purkinje/patología
9.
J Neurotrauma ; 37(12): 1463-1480, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32056479

RESUMEN

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.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Inmunidad/inmunología , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Receptores de Interleucina-1/antagonistas & inhibidores , Retina/inmunología , Percepción Visual/inmunología , Animales , Traumatismos por Explosión/diagnóstico por imagen , Traumatismos por Explosión/tratamiento farmacológico , Traumatismos por Explosión/inmunología , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Electrorretinografía/métodos , Inmunidad/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Retina/diagnóstico por imagen , Retina/efectos de los fármacos , Tomografía de Coherencia Óptica/métodos , Resultado del Tratamiento , Percepción Visual/efectos de los fármacos
10.
Mil Med ; 185(Suppl 1): 628-636, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-32074316

RESUMEN

INTRODUCTION: We examined antibiotic management of combat-related extremity wound infections (CEWI) among wounded U.S. military personnel (2009-2012). METHODS: Patients were included if they sustained blast injuries, resulting in ≥1 open extremity wound, were admitted to participating U.S. hospitals, developed a CEWI (osteomyelitis or deep soft-tissue infections) within 30 days post-injury, and received ≥3 days of relevant antibiotic (s) for treatment. RESULTS: Among 267 patients, 133 (50%) had only a CEWI, while 134 (50%) had a CEWI plus concomitant non-extremity infection. In the pre-diagnosis period (4-10 days prior to CEWI diagnosis), 95 (36%) patients started a new antibiotic with 28% of patients receiving ≥2 antibiotics. During CEWI diagnosis week (±3 days of diagnosis), 209 (78%) patients started a new antibiotic (71% with ≥2 antibiotics). In the week following diagnosis (4-10 days after CEWI diagnosis), 121 (45%) patients started a new antibiotic with 39% receiving ≥2 antibiotics. Restricting to ±7 days of CEWI diagnosis, patients commonly received two (35%) or three (27%) antibiotics with frequent combinations involving carbapenem, vancomycin, and fluoroquinolones. CONCLUSIONS: Substantial variation in antibiotic prescribing patterns related to CEWIs warrants development of combat-related clinical practice guidelines beyond infection prevention, to include strategies to reduce the use of unnecessary antibiotics and improve stewardship.


Asunto(s)
Extremidades/lesiones , Pautas de la Práctica en Medicina/normas , Infección de Heridas/tratamiento farmacológico , Adulto , Antibacterianos/uso terapéutico , Programas de Optimización del Uso de los Antimicrobianos/normas , Programas de Optimización del Uso de los Antimicrobianos/estadística & datos numéricos , Traumatismos por Explosión/complicaciones , Traumatismos por Explosión/tratamiento farmacológico , Traumatismos por Explosión/fisiopatología , Explosiones/estadística & datos numéricos , Extremidades/fisiopatología , Femenino , Humanos , Masculino , Pautas de la Práctica en Medicina/estadística & datos numéricos , Estados Unidos , Infección de Heridas/etiología
11.
Toxicol Lett ; 319: 49-57, 2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-31693926

RESUMEN

Blast lung injury is associated with high morbidity and mortality. Vaporized perfluorocarbon (PFC) inhalation has been reported to attenuate acute respiratory distress syndrome in humans and animal models. However, the effect of vaporized PFC on blast lung injury is still unknown. In this study, we investigated the protective effects and potential underlying mechanisms of action of vaporized PFC on blast lung injury in a canine model. This was a prospective, controlled, animal study in adult male hybrid dogs randomized to sham, blast (B), blast plus mechanical ventilation (B + M), and blast plus PFC (B + P) groups. All groups except for the sham were exposed to blast wave. The B + P group was treated with vaporized PFC for 1.5 h followed by 5.5 h mechanical ventilation. B + M group received 7.5 h mechanical ventilation and B group was observed for 7.5 h. Blast lung injury was induced using a shock tube. Blood gas, inflammatory cytokines, and oxidative stress were measured. Expression of nuclear factor (NF)-κB activation, mitogen-activated protein kinase (MAPK) and nuclear factor, erythroid 2 like 2 (Nrf2) were measured using western blot. Lung injury observed after blast exposure was marked by increased histopathological scores, ratio of lung wet to dry weight. PFC treatment attenuated blast lung injury as indicated by histopathological scores and ratio of lung wet to dry weight. PFC treatment downregulated interleukin (IL)-6, tumor necrosis factor (TNF)-α, and malondialdehyde (MDA), and upregulated superoxide dismutase (SOD) activity. PFC also suppressed expression of MAPK/NF-κB and Nrf2 protein levels. Our results suggest that PFC attenuated blast-induced acute lung injury by inhibiting MAPK/NF-κB activation and inducing Nrf2 expression in dogs.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Fluorocarburos/uso terapéutico , Lesión Pulmonar/tratamiento farmacológico , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Administración por Inhalación , Animales , Traumatismos por Explosión/patología , Líquido del Lavado Bronquioalveolar , Citocinas/metabolismo , Perros , Fluorocarburos/administración & dosificación , Pulmón/patología , Lesión Pulmonar/patología , Masculino , Estrés Oxidativo/efectos de los fármacos
12.
Mil Med ; 184(Suppl 1): 282-290, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30901474

RESUMEN

OBJECTIVE: Blast injury is associated with multi-organ failure (MOF), causing significant morbidity and mortality in trauma patients. However, the pathogenesis of blast-induced MOF still remains obscure. In this study, we evaluate the pathophysiological changes related to blast-induced MOF in a clinically relevant rat model of blast injury. METHODS: A moderate blast overpressure was applied to induce injury in anesthetized rats. Pathological changes were evaluated by H&E staining. Complement activation, plasminogen, and myeloperoxidase levels were analyzed by complement hemolytic assay (CH50) and/or ELISA in blood samples. RESULTS: Analysis of lung, brain, and liver tissue at 24 hour after blast overpressure revealed severe injuries. The level of complement components C3 and C1q decreased in parallel with the reduction of CH50 level in injured animals at 1, 3, and 6 hours after blast. Consumption of plasminogen was also detected as early as 1 hour post-injury. Myeloperoxidase levels were elevated within 1 hour of blast injury. CONCLUSION: Our data reveal that blast injury triggers the complement and fibrinolytic systems, which likely contribute to blast-induced MOF. Conceivably, therapies that target these systems early may improve clinical outcomes in blast patients.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Fibrinolíticos/farmacocinética , Insuficiencia Multiorgánica/etiología , Animales , Traumatismos por Explosión/complicaciones , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Fibrinolíticos/sangre , Fibrinolíticos/uso terapéutico , Puntaje de Gravedad del Traumatismo , Hígado/efectos de los fármacos , Hígado/patología , Hígado/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Masculino , Insuficiencia Multiorgánica/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley/sangre , Estadísticas no Paramétricas
13.
Toxicol Lett ; 301: 90-97, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30423366

RESUMEN

Caffeine is a substance that is consumed worldwide, and it may exert neuroprotective effects against various cerebral insults, including neurotrauma, which is the most prevalent injury among military personnel. To investigate the effects of caffeine on high-intensity blast wave-induced severe blast injury in mice, three different paradigms of caffeine were applied to male C57BL/6 mice with severe whole body blast injury (WBBI). The results demonstrated that chronic caffeine treatment alleviated blast-induced traumatic brain injury (bTBI); however, both chronic and acute caffeine treatments exacerbated blast-induced lung injuries and, more importantly, increased both the cumulative and time-segmented mortalities postinjury. Interestingly, withdrawing caffeine intake preinjury resulted in favorable outcomes in mortality and lung injury, similar to the findings in water-treated mice, and had the trend to attenuate brain injury. These findings demonstrated that although drinking coffee or caffeine preparations attenuated blast-induced brain trauma, these beverages may place personnel in the battlefield at high risk of casualties, which will help us re-evaluate the therapeutic strategy of caffeine application, particularly in multiple-organ-trauma settings. Furthermore, these findings provided possible strategies for reducing the risk of casualties with caffeine consumption, which may help to change the coffee-drinking habits of military personnel.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Cafeína/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Traumatismos por Explosión/mortalidad , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL
14.
Neuropharmacology ; 145(Pt B): 220-229, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30227150

RESUMEN

A striking observation among veterans returning from the recent conflicts in Iraq and Afghanistan has been the co-occurrence of blast-related mild traumatic brain injury (mTBI) and post-traumatic stress disorder (PTSD). PTSD and mTBI might coexist due to additive effects of independent psychological and physical traumas experienced in a war zone. Alternatively blast injury might induce PTSD-related traits or damage brain structures that mediate responses to psychological stressors, increasing the likelihood that PTSD will develop following a subsequent psychological stressor. Rats exposed to repetitive low-level blasts consisting of three 74.5 kPa exposures delivered once daily for three consecutive days develop a variety of anxiety and PTSD-related behavioral traits that are present for at least 9 months after blast exposure. A single predator scent challenge delivered 8 months after the last blast exposure induces additional anxiety-related changes that are still present 45 days later. Because the blast injuries occur under general anesthesia, it appears that blast exposure in the absence of a psychological stressor can induce chronic PTSD-related traits. The reaction to a predator scent challenge delivered many months after blast exposure suggests that blast exposure in addition sensitizes the brain to react abnormally to subsequent psychological stressors. The development of PTSD-related behavioral traits in the absence of a psychological stressor suggests the existence of blast-induced "PTSD". Findings that PTSD-related behavioral traits can be reversed by BCI-838, a group II metabotropic glutamate receptor antagonist offers insight into pathogenesis and possible treatment options for blast-related brain injury. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".


Asunto(s)
Traumatismos por Explosión/complicaciones , Lesiones Traumáticas del Encéfalo/etiología , Trastornos por Estrés Postraumático/etiología , Animales , Traumatismos por Explosión/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Humanos , Trastornos por Estrés Postraumático/tratamiento farmacológico
15.
PLoS One ; 13(8): e0202594, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30133517

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Traumatismos por Explosión/tratamiento farmacológico , Antígenos CD55/administración & dosificación , Proteína HMGB1/genética , Inflamación/tratamiento farmacológico , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/fisiopatología , Animales , Traumatismos por Explosión/genética , Traumatismos por Explosión/patología , Activación de Complemento/efectos de los fármacos , Complemento C3a/antagonistas & inhibidores , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , FN-kappa B/genética , Presión/efectos adversos , Ratas , Ratas Sprague-Dawley
16.
Sci Rep ; 8(1): 10622, 2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-30006635

RESUMEN

Explosions account for 79% of combat related injuries and often lead to polytrauma, a majority of which include blast-induced traumatic brain injuries (bTBI). These injuries lead to internal bleeding in multiple organs and, in the case of bTBI, long term neurological deficits. Currently, there are no treatments for internal bleeding beyond fluid resuscitation and surgery. There is also a dearth of treatments for TBI. We have developed a novel approach using hemostatic nanoparticles that encapsulate an anti-inflammatory, dexamethasone, to stop the bleeding and reduce inflammation after injury. We hypothesize that this will improve not only survival but long term functional outcomes after blast polytrauma. Poly(lactic-co-glycolic acid) hemostatic nanoparticles encapsulating dexamethasone (hDNPs) were fabricated and tested following injury along with appropriate controls. Rats were exposed to a single blast wave using an Advanced Blast Simulator, inducing primary blast lung and bTBI. Survival was elevated in the hDNPs group compared to controls. Elevated anxiety parameters were found in the controls, compared to hDNPs. Histological analysis indicated that apoptosis and blood-brain barrier disruption in the amygdala were significantly increased in the controls compared to the hDNPs and sham groups. Immediate intervention is crucial to mitigate injury mechanisms that contribute to emotional deficits.


Asunto(s)
Ansiedad/tratamiento farmacológico , Dexametasona/administración & dosificación , Portadores de Fármacos/química , Hemostáticos/administración & dosificación , Traumatismo Múltiple/tratamiento farmacológico , Animales , Ansiedad/etiología , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Traumatismos por Explosión/tratamiento farmacológico , Traumatismos por Explosión/etiología , Traumatismos por Explosión/mortalidad , Traumatismos por Explosión/psicología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Lesiones Encefálicas/mortalidad , Lesiones Encefálicas/psicología , Modelos Animales de Enfermedad , Explosiones , Humanos , Inyecciones Intravenosas , Masculino , Traumatismo Múltiple/etiología , Traumatismo Múltiple/mortalidad , Traumatismo Múltiple/psicología , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Resultado del Tratamiento , Guerra
17.
eNeuro ; 5(1)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29387781

RESUMEN

Battlefield blast exposure related to improvised explosive devices (IEDs) has become the most common cause of traumatic brain injury (TBI) in the recent conflicts in Iraq and Afghanistan. Mental health problems are common after TBI. A striking feature in the most recent veterans has been the frequency with which mild TBI (mTBI) and posttraumatic stress disorder (PTSD) have appeared together, in contrast to the classical situations in which the presence of mTBI has excluded the diagnosis of PTSD. However, treatment of PTSD-related symptoms that follow blast injury has become a significant problem. BCI-838 (MGS0210) is a Group II metabotropic glutamate receptor (mGluR2/3) antagonist prodrug, and its active metabolite BCI-632 (MGS0039) has proneurogenic, procognitive, and antidepressant activities in animal models. In humans, BCI-838 is currently in clinical trials for refractory depression and suicidality. The aim of the current study was to determine whether BCI-838 could modify the anxiety response and reverse PTSD-related behaviors in rats exposed to a series of low-level blast exposures designed to mimic a human mTBI or subclinical blast exposure. BCI-838 treatment reversed PTSD-related behavioral traits improving anxiety and fear-related behaviors as well as long-term recognition memory. Treatment with BCI-838 also increased neurogenesis in the dentate gyrus (DG) of blast-exposed rats. The safety profile of BCI-838 together with the therapeutic activities reported here, make BCI-838 a promising drug for the treatment of former battlefield Warfighters suffering from PTSD-related symptoms following blast-induced mTBI.


Asunto(s)
Traumatismos por Explosión/complicaciones , Conmoción Encefálica/complicaciones , Compuestos Bicíclicos con Puentes/farmacología , Psicotrópicos/farmacología , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/psicología , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Traumatismos por Explosión/tratamiento farmacológico , Traumatismos por Explosión/psicología , Conmoción Encefálica/tratamiento farmacológico , Conmoción Encefálica/psicología , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Giro Dentado/patología , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Miedo/efectos de los fármacos , Miedo/fisiología , Masculino , Memoria a Largo Plazo/efectos de los fármacos , Memoria a Largo Plazo/fisiología , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas Long-Evans , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/patología
18.
Behav Brain Res ; 340: 183-194, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27793733

RESUMEN

Mild traumatic brain injury (mTBI) diagnoses have increased due to aggressive sports and blast-related injuries, but the cellular mechanisms and pathology underlying mTBI are not completely understood. Previous reports indicate that Nociceptin Orphanin/FQ (N/OFQ), an endogenous neuropeptide, contributes to post-injury ischemia following mechanical brain injury, yet its specific role in cerebral hypoxia, vestibulomotor function and injury marker expression following blast-induced TBI is not known. This study is the first to identify a direct association of N/OFQ and its N/OFQ peptide (NOP) receptor with TBI-induced changes following a single 80psi head blast exposure in male rats. N/OFQ and NOP receptor expression increased in brain tissue and plasma following TBI, concurrent with vestibular dysfunction but preceding hypoxia and appearance of injury markers compared to sham rats. A single post-blast treatment with the NOP receptor antagonist, SB-612111, transiently improved acute vestibulomotor performance. It also prevented increases in markers of TBI-induced hypoxia, pro-apoptotic proteins and injury seen 8-10days post-blast. This study reveals an apparent role for the N/OFQ-NOP receptor system in blast TBI and suggests potential therapeutic utility of NOP receptor antagonists for mTBI.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Conmoción Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Cicloheptanos/farmacología , Hipoxia Encefálica/prevención & control , Antagonistas de Narcóticos/farmacología , Piperidinas/farmacología , Animales , Traumatismos por Explosión/patología , Traumatismos por Explosión/fisiopatología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Conmoción Encefálica/etiología , Conmoción Encefálica/patología , Conmoción Encefálica/fisiopatología , Hipoxia Encefálica/etiología , Hipoxia Encefálica/patología , Hipoxia Encefálica/fisiopatología , Masculino , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteoma/efectos de los fármacos , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Receptor de Nociceptina
19.
Chem Biol Interact ; 271: 15-23, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28455230

RESUMEN

There is growing evidence that Mangiferin possess therapeutic benefit during neuroinflammation on various brain injury models due to its anti-inflammatory properties. It is reported that inflammatory plays a crucial role in the pathogenesis of secondary injury induced by the blast-induced traumatic brain injury (bTBI). However, the role of mangiferin in bTBI is yet to be studied. In our study, the potential effect of mangiferin in the duration of bTBI was examined first. Fortunately, the amelioration of cerebral cortex damage was found in rats suffering bTBI after mangiferin administration. Furthermore, the detail mechanism of mangiferin's beneficial actions in bTBI was also studied. The results revealed that mangiferin might alleviate brain damage in rats with bTBI by inhibiting the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome activation, which was accompanied by mangiferin's inhibition of oxidative stress and pro-inflammatory cytokines production. Therefore, this research allows us to speculate that, for first time, NLRP3 is involved in the anti-inflammatory effect of mangiferin in the cerebral cortex, and mangiferin could be a potential therapy drug for bTBI.


Asunto(s)
Traumatismos por Explosión/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Inflamasomas/antagonistas & inhibidores , Proteínas NLR/antagonistas & inhibidores , Xantonas/farmacología , Xantonas/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Traumatismos por Explosión/fisiopatología , Corteza Cerebral/efectos de los fármacos , Citocinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/tratamiento farmacológico , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas
20.
Free Radic Biol Med ; 108: 627-643, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28438658

RESUMEN

Cochlear neurodegeneration commonly accompanies hair cell loss resulting from aging, ototoxicity, or exposures to intense noise or blast overpressures. However, the precise pathophysiological mechanisms that drive this degenerative response have not been fully elucidated. Our laboratory previously demonstrated that non-transgenic rats exposed to blast overpressures exhibited marked somatic accumulation of neurotoxic variants of the microtubule-associated protein, Tau, in the hippocampus. In the present study, we extended these analyses to examine neurodegeneration and pathologic Tau accumulation in the auditory system in response to blast exposure and evaluated the potential therapeutic efficacy of antioxidants on short-circuiting this pathological process. Blast injury induced ribbon synapse loss and retrograde neurodegeneration in the cochlea in untreated animals. An accompanying perikaryal accumulation of neurofilament light chain and pathologic Tau oligomers were observed in neurons from both the peripheral and central auditory system, spanning from the spiral ganglion to the auditory cortex. Due to its coincident accumulation pattern and well-documented neurotoxicity, our results suggest that the accumulation of pathologic Tau oligomers may actively contribute to blast-induced cochlear neurodegeneration. Therapeutic intervention with a combinatorial regimen of 2,4-disulfonyl α-phenyl tertiary butyl nitrone (HPN-07) and N-acetylcysteine (NAC) significantly reduced both pathologic Tau accumulation and indications of ongoing neurodegeneration in the cochlea and the auditory cortex. These results demonstrate that a combination of HPN-07 and NAC administrated shortly after a blast exposure can serve as a potential therapeutic strategy for preserving auditory function among military personnel or civilians with blast-induced traumatic brain injuries.


Asunto(s)
Acetilcisteína/uso terapéutico , Antioxidantes/uso terapéutico , Bencenosulfonatos/uso terapéutico , Traumatismos por Explosión/tratamiento farmacológico , Células Ciliadas Auditivas/fisiología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuronas/fisiología , Enfermedades del Nervio Vestibulococlear/tratamiento farmacológico , Animales , Corteza Auditiva/patología , Muerte Celular , Células Cultivadas , Masculino , Ratas , Ratas Endogámicas , Ganglio Espiral de la Cóclea/patología , Respuesta de Proteína Desplegada , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...