Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Behav Brain Res ; 368: 111895, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-30978410

RESUMO

Mild traumatic brain injury is a common outcome of blast exposure, and current literature indicates high rates of comorbid posttraumatic stress disorder (PTSD) in military personnel. Blast-exposed rats display PTSD-like behavior, suggesting relationships may exist between PTSD and blast exposure. Other studies demonstrate the roles of stathmin and corticosterone associated with fear- and anxiety-like behaviors in rodent models. Furthermore, studies have observed ranges of responses to both physical and psychological trauma in animal populations (Elder 2012, Ritov 2016). This study exposed rodents to repeated blast overpressure (BOP) and analyzed behavioral responses and molecular variables at 3 weeks and 6 months after exposure. We applied a modified version of a previously reported behavioral profiling approach that separates "affected" and "unaffected" rats based on the presence of anxiety-like behaviors (Ritov, 2016). We report that "affected" 3 week animals showed higher plasma corticosterone and amygdalar stathmin levels, while "affected" 6 month animals had lower prefrontal cortex stathmin. Higher corticosterone also paralleled anxiety behavior in "affected" 3 week animals, which was not observed in 6 month animals, indicating possible negative feedback loop mechanisms. Elevated levels of amygdalar stathmin correlated with anxiety behaviors in "affected" 3 week and 6 month animals, indicating sustained molecular changes. We conclude that this unique analysis may provide more information about response to blast. This type of analysis should also be considered when treating clinical populations, since individual differences may affect behavioral and long-term outcomes. Future studies should elucidate relationships of stress and fear responses in the context of BOP.


Assuntos
Ansiedade/fisiopatologia , Concussão Encefálica/metabolismo , Concussão Encefálica/psicologia , Tonsila do Cerebelo/metabolismo , Animais , Ansiedade/psicologia , Traumatismos por Explosões/psicologia , Lesões Encefálicas/psicologia , Comorbidade , Corticosterona/análise , Corticosterona/sangue , Modelos Animais de Doenças , Medo/fisiologia , Masculino , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Long-Evans , Estatmina/análise , Transtornos de Estresse Pós-Traumáticos/metabolismo , Transtornos de Estresse Pós-Traumáticos/psicologia
2.
J Trauma Acute Care Surg ; 86(1): 116-122, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29985235

RESUMO

BACKGROUND: Aeromedical evacuation to definitive care is standard in current military conflicts. However, there is minimal knowledge on the effects of hypobaria (HYPO) on either the flight crew or patients. The effects of HYPO were investigated using healthy swine. METHODS: Anesthetized Yorkshire swine underwent a simulated 4 h "transport" to an altitude of 2,441 m (8,000 feet.; HYPO, N = 6) or at normobaric conditions (NORMO, N = 6). Physiologic and biochemical data were collected. Organ damage was assessed for hemorrhage, inflammation, edema, necrosis, and for lungs only, microatelectasis. RESULTS: All parameters were similar prior to and after "transport" with no significant effects of HYPO on hemodynamic, neurologic, or oxygen transport parameters, nor on blood gas, chemistry, or complete blood count data. However, the overall Lung Injury Score was significantly worse in the HYPO than the NORMO group (10.78 ± 1.22 vs. 2.31 ± 0.71, respectively) with more edema/fibrin/hemorrhage in the subpleural, interlobular and alveolar space, more congestion in alveolar septa, and evidence of microatelectasis (vs. no microatelectasis in the NORMO group). There was also increased severity of pulmonary neutrophilic (1.69 ± 0.20 vs. 0.19 ± 0.13) and histiocytic inflammation (1.83 ± 0.23 vs. 0.47 ± 0.17) for HYPO versus NORMO, respectively. On the other hand, there was increased renal inflammation in NORMO compared with HYPO (1.00 ± 0.13 vs. 0.33 ± 0.17, respectively). There were no histopathological differences in brain (whole or individual regions), liver, pancreas, or adrenals. CONCLUSION: Hypobaria, itself, may have an adverse effect on the respiratory system, even in healthy individuals, and this may be superimposed on combat casualties where there may be preexisting lung injury. The additional effects of anesthesia and controlled ventilation on these results are unknown, and further studies are indicated using awake models to better characterize the mechanisms for this pathology and the factors that influence its severity.


Assuntos
Resgate Aéreo/estatística & dados numéricos , Barotrauma/complicações , Encéfalo/patologia , Pulmão/patologia , Altitude , Animais , Pressão Atmosférica , Gasometria/métodos , Lesões Encefálicas/etiologia , Modelos Animais de Doenças , Edema/patologia , Feminino , Hemodinâmica/fisiologia , Hemorragia/patologia , Inflamação/imunologia , Inflamação/patologia , Lesão Pulmonar/etiologia , Masculino , Necrose/patologia , Atelectasia Pulmonar/patologia , Suínos
3.
Neurosci Lett ; 684: 91-97, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30018022

RESUMO

INTRODUCTION: A stressful environment may contribute to poor outcomes after TBI. The current study evaluates the impact of acute stress in a polytrauma rat model. METHODS: Rats were stressed by a 45-minute immobilization period before instrumentation under ketamine (t1). Polytrauma was produced by blast overpressure and controlled hemorrhage (t2). Rats were euthanized immediately after a 3 h simulated Medevac-transport time (t3) or after 72 h post-trauma (t4). Corticosterone, ACTH, and ACTH receptor gene expression were measured at these time points. Physiological parameters were monitored throughout the study. RESULTS: HR was higher in stressed compared to unstressed animals at t1. Corticosterone and ACTH levels were similar for all conditions at t1 and t2; ACTH and corticosterone became elevated in all groups at t3 and at t4, respectively. The ACTH receptor gene expression trended towards higher values at t4 for the stressed animals whether being injured or not. Survival after injury was 83% in both unstressed and stressed animals. CONCLUSION: Overall, corticosterone was not significantly affected following acute stress in ketamine-anesthetized rats. Early mortality was primarily due to polytrauma and change in the animal's biochemical parameters appeared at t4 post trauma. The findings indicate that ketamine-anesthesia and/or surgery may have overshadowed the effect of the initial stress.


Assuntos
Hormônio Adrenocorticotrópico/sangue , Corticosterona/sangue , Modelos Animais de Doenças , Traumatismo Múltiplo/sangue , Estresse Psicológico/sangue , Doença Aguda , Animais , Masculino , Traumatismo Múltiplo/mortalidade , Ratos , Ratos Sprague-Dawley , Restrição Física , Estresse Psicológico/mortalidade , Taxa de Sobrevida/tendências
4.
Microcirculation ; 25(3): e12441, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29356218

RESUMO

OBJECTIVE: The aim of this study was to assess, in two experiments, the safety and efficacy of the PFC emulsion Oxycyte as an oxygen therapeutic for TBI to test the hypothesis that early administration of this oxygen-carrying fluid post-TBI would improve brain tissue oxygenation (Pbt O2 ). METHODS: The first experiment assessed the effects of Oxycyte on cerebral vasoactivity in healthy, uninjured rats using intravital microscopy. The second experiment investigated the effect of Oxycyte on cerebral Pbt O2 using the PQM in TBI model. Animals in the Oxycyte group received a single injection of Oxycyte (6 mL/kg) shortly after TBI, while NON animals received no treatment. RESULTS: Oxycyte did not cause vasoconstriction in small- (<50 µm) or medium- (50-100 µm) sized pial arterioles nor did it cause a significant change in blood pressure. Treatment with Oxycyte while breathing 100% O2 did not improve Pbt O2 . However, in rats ventilated with ~40% O2 , Pbt O2 improved to near pre-TBI values within 105 minutes after Oxycyte injection. CONCLUSIONS: Although Oxycyte did not cause cerebral vasoconstriction, its use at the dose tested while breathing 100% O2 did not improve Pbt O2 following TBI. However, Oxycyte treatment while breathing a lower enriched oxygen concentration may improve Pbt O2 after TBI.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Fluorocarbonos/uso terapêutico , Oxigênio/sangue , Animais , Arteríolas/fisiologia , Encéfalo/metabolismo , Circulação Cerebrovascular , Microscopia Intravital , Oxigênio/administração & dosagem , Ratos , Vasoconstrição/efeitos dos fármacos
5.
Acta Neuropathol Commun ; 5(1): 80, 2017 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-29126430

RESUMO

Blast-related traumatic brain injury (TBI) has been a common cause of injury in the recent conflicts in Iraq and Afghanistan. Blast waves can damage blood vessels, neurons, and glial cells within the brain. Acutely, depending on the blast energy, blast wave duration, and number of exposures, blast waves disrupt the blood-brain barrier, triggering microglial activation and neuroinflammation. Recently, there has been much interest in the role that ongoing neuroinflammation may play in the chronic effects of TBI. Here, we investigated whether chronic neuroinflammation is present in a rat model of repetitive low-energy blast exposure. Six weeks after three 74.5-kPa blast exposures, and in the absence of hemorrhage, no significant alteration in the level of microglia activation was found. At 6 weeks after blast exposure, plasma levels of fractalkine, interleukin-1ß, lipopolysaccharide-inducible CXC chemokine, macrophage inflammatory protein 1α, and vascular endothelial growth factor were decreased. However, no differences in cytokine levels were detected between blast-exposed and control rats at 40 weeks. In brain, isolated changes were seen in levels of selected cytokines at 6 weeks following blast exposure, but none of these changes was found in both hemispheres or at 40 weeks after blast exposure. Notably, one animal with a focal hemorrhagic tear showed chronic microglial activation around the lesion 16 weeks post-blast exposure. These findings suggest that focal hemorrhage can trigger chronic focal neuroinflammation following blast-induced TBI, but that in the absence of hemorrhage, chronic neuroinflammation is not a general feature of low-level blast injury.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Citocinas/metabolismo , Encefalite/etiologia , Hemorragias Intracranianas/complicações , Hemorragias Intracranianas/etiologia , Animais , Lesões Encefálicas Traumáticas/etiologia , Lesões Encefálicas Traumáticas/patologia , Córtex Cerebral/patologia , Quimiocina CCL3/metabolismo , Quimiocina CCL5/metabolismo , Modelos Animais de Doenças , Feminino , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Mutação/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
6.
Lung ; 194(6): 945-957, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27704259

RESUMO

PURPOSE: Perfluorocarbons (PFCs) can transport 50 times more oxygen than human plasma. Their properties may be advantageous in preservation of tissue viability in oxygen-deprived states, such as in acute lung injury. We hypothesized that an intravenous dose of the PFC emulsion Oxycyte® would improve tissue oxygenation and thereby mitigate the effects of acute lung injury. METHODS: Intravenous oleic acid (OA) was used to induce lung injury in anesthetized and instrumented Yorkshire swine assigned to three experimental groups: (1) PFC post-OA received Oxycyte® (5 ml/kg) 45 min after oleic acid-induced lung injury (OALI); (2) PFC pre-OA received Oxycyte® 45 min before OALI; and (3) Controls which received equivalent dose of normal saline. Animals were observed for 3 h after OALI began, and then euthanized. RESULTS: The median survival times for PFC post-OA, PFC pre-OA, and control were 240, 87.5, and 240 min, respectively (p = 0.001). Mean arterial pressure and mean pulmonary arterial pressure were both higher in the PFC post-OA (p < 0.001 for both parameters). Oxygen content was significantly different between PFC post-OA and the control (p = 0.001). Histopathological grading of lung injury indicated that edema and congestion was significantly less severe in the PFC post-OA compared to control (p = 0.001). CONCLUSION: The intravenous PFC Oxycyte® improves blood oxygen content and lung histology when used as a treatment after OALI, while Oxycyte® used prior to OALI was associated with increased mortality. Further exploration in other injury models is indicated.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/patologia , Fluorocarbonos/administração & dosagem , Oxigênio/sangue , Equilíbrio Ácido-Base , Lesão Pulmonar Aguda/induzido quimicamente , Administração Intravenosa , Animais , Pressão Arterial/efeitos dos fármacos , Gasometria , Modelos Animais de Doenças , Feminino , Fluorocarbonos/efeitos adversos , Ácido Láctico/sangue , Masculino , Ácido Oleico , Pressão Propulsora Pulmonar/efeitos dos fármacos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Índice de Gravidade de Doença , Taxa de Sobrevida , Suínos
7.
J Trauma Acute Care Surg ; 81(1): 101-7, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26998778

RESUMO

BACKGROUND: There is inadequate information on the physiologic effects of aeromedical evacuation on wounded war fighters with traumatic brain injury (TBI). At altitudes of 8,000 ft, the inspired oxygen is lower than standard sea level values. In troops experiencing TBI, this reduced oxygen may worsen or cause secondary brain injury. We tested the hypothesis that the effects of prolonged aeromedical evacuation on critical neurophysiologic parameters (i.e., brain oxygenation [PbtO2]) of swine with a fluid percussion injury/TBI would be detrimental compared with ground (normobaric) transport. METHODS: Yorkshire swine underwent fluid percussion injury/TBI with pretransport stabilization before being randomized to a 4-hour aeromedical transport at simulated flight altitude of 8,000 ft (HYPO, n = 8) or normobaric ground transport (NORMO, n = 8). Physiologic measurements (i.e., PbtO2, cerebral perfusion pressure, intracranial pressure, regional cerebral blood flow, mean arterial blood pressure, and oxygen transport variables) were analyzed. RESULTS: Survival was equivalent between groups. Measurements were similar in both groups at all phases up to and including onset of flight. During the flight, PbtO2, cerebral perfusion pressure, and mean arterial blood pressure were significantly lower in the HYPO than in the NORMO group. At the end of flight, regional cerebral blood flow was lower in the HYPO than in the NORMO group. Other parameters such as intracranial pressure, cardiac output, and mean pulmonary artery pressure were not significantly different between the two groups. CONCLUSION: A 4-hour aeromedical evacuation at a simulated flight altitude of 8,000 ft caused a notable reduction in neurophysiologic parameters compared with normobaric conditions in this TBI swine model. Results suggest that hypobaric conditions exacerbate cerebral hypoxia and may worsen TBI in casualties already in critical condition.


Assuntos
Resgate Aéreo , Altitude , Lesões Encefálicas Traumáticas/fisiopatologia , Hipóxia Encefálica/fisiopatologia , Animais , Lesões Encefálicas Traumáticas/mortalidade , Débito Cardíaco , Circulação Cerebrovascular , Modelos Animais de Doenças , Hipóxia Encefálica/mortalidade , Pressão Intracraniana , Oxigênio/sangue , Distribuição Aleatória , Taxa de Sobrevida , Suínos
8.
Front Neurol ; 6: 49, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25852633

RESUMO

Repeated exposure to low-level blast is a characteristic of a few select occupations and there is concern that such occupational exposures present risk for traumatic brain injury. These occupations include specialized military and law enforcement units that employ controlled detonation of explosive charges for the purpose of tactical entry into secured structures. The concern for negative effects from blast exposure is based on rates of operator self-reported headache, sleep disturbance, working memory impairment, and other concussion-like symptoms. A challenge in research on this topic has been the need for improved assessment tools to empirically evaluate the risk associated with repeated exposure to blast overpressure levels commonly considered to be too low in magnitude to cause acute injury. Evaluation of serum-based neurotrauma biomarkers provides an objective measure that is logistically feasible for use in field training environments. Among candidate biomarkers, ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1) has some empirical support and was evaluated in this study. We used daily blood draws to examine acute change in UCH-L1 among 108 healthy military personnel who were exposed to repeated low-level blast across a 2-week period. These research volunteers also wore pressure sensors to record blast exposures, wrist actigraphs to monitor sleep patterns, and completed daily behavioral assessments of symptomology, postural stability, and neurocognitive function. UCH-L1 levels were elevated as a function of participating in the 2-week training with explosives, but the correlation of UCH-L1 elevation and blast magnitude was weak and inconsistent. Also, UCH-L1 elevations did not correlate with deficits in behavioral measures. These results provide some support for including UCH-L1 as a measure of central nervous system effects from exposure to low-level blast. However, the weak relation observed suggests that additional indicators of blast effect are needed.

9.
Vet Immunol Immunopathol ; 161(1-2): 1-11, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25113962

RESUMO

Plasma from hibernating (HIB) woodchucks (Marmota monax) or 13-lined ground squirrels (Ictidomys tridecemlineatus) suppressed (3)H-thymidine uptake in mouse spleen cell cultures stimulated with Concanavalin A (ConA); plasma from non-hibernating animals were only slightly inhibitory. Maximum inhibition occurred when HIB plasma was added to the cultures prior to ConA. After HPLC size exclusion chromatography of the HIB ground squirrel plasma, a single fraction (fraction-14) demonstrated inhibitory activity. Assay of fraction-14 from 8 HIB squirrels showed inhibition ranging from 13 to 95%; inhibition was correlated to the time the squirrels were exposed to cold prior to hibernation. Western blot analysis showed the factor to be a large molecular weight protein (>300 kDa), and mass spectrometry identified sequences that were 100% homologous with alpha-2-macroglobulin from humans and other species. These findings indicate a hibernation-related protein that may be responsible for immune system down regulation.


Assuntos
Hibernação/fisiologia , Linfócitos/efeitos dos fármacos , Sciuridae/sangue , Sciuridae/fisiologia , alfa-Macroglobulinas/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Concanavalina A , Feminino , Camundongos , Mitógenos , Baço/citologia , alfa-Macroglobulinas/fisiologia
10.
Acta Neuropathol Commun ; 2: 67, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24938728

RESUMO

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


Assuntos
Traumatismos por Explosões/complicações , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Córtex Cerebral/patologia , Hemorragia Cerebral/etiologia , Vasculite do Sistema Nervoso Central/etiologia , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/ultraestrutura , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/etiologia , Masculino , Microscopia Eletrônica , Microvasos/metabolismo , Microvasos/patologia , Microvasos/ultraestrutura , Ratos , Ratos Long-Evans , Fatores de Tempo , Vasculite do Sistema Nervoso Central/patologia
11.
Curr Drug Discov Technol ; 10(4): 315-24, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24074305

RESUMO

Hemoglobin-based oxygen carrier-201 (HBOC) was developed as a resuscitative fluid but concerns exist over potentially adverse vasoconstriction. This study evaluated whether concurrent IV (intra venous) N-acetyl-L-cysteine (NAC) or hyaluronic acid (HA) would attenuate HBOC-associated vasoconstriction, assessed by systemic blood pressures and cerebral pial microvasculature, when administered to healthy, anesthetized rats. Rats (8-9/group) received a 30 min infusion of 3 ml/kg HBOC, HBOC plus 600 mg/kg NAC (HBOC/NAC), HBOC plus 1.5 mg/kg HA (HBOC/HA) or 3 ml/kg Albumin. Mean (MAP) and systolic (SBP) blood pressures, blood chemistries and cerebral pial vessel diameters were measured at baseline, end of infusion, and intermittently for an additional 90 min. HBOC caused immediate and sustained increases in SBP and MAP (35.3 ± 3.6 and 29.1 ± 2.5 mm Hg peak increases above baseline, respectively; mean ± SEM) and immediate but progressive vasoconstriction (11 µm maximum reduction) in medium-sized (50-100 µm) pial arterioles. When NAC was co-administered, blood pressure changes were attenuated and vessel changes were abolished. Similar trends were noted with co-administration of HA but were not statistically different from HBOC-alone. Small-sized (< 50 µm) pial vessels and blood parameters showed no differences from baseline or among groups. No adverse clinical signs were observed. We demonstrated that it is possible for adjuvant drugs to reduce the vasoconstriction associated with HBOC-201. Coinfusion of the anti-oxidant NAC mitigated HBOC-201-associated increases in blood pressures and vasoconstriction in medium-sized cerebral pial vessels. The drag-reducing polymer HA may be more effective at a higher dose as a similar but non-significant trend was observed.


Assuntos
Acetilcisteína/administração & dosagem , Artérias Cerebrais/efeitos dos fármacos , Hemoglobinas/administração & dosagem , Ácido Hialurônico/administração & dosagem , Vasoconstrição/efeitos dos fármacos , Animais , Pressão Sanguínea , Artérias Cerebrais/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley
12.
Aviat Space Environ Med ; 84(9): 913-20, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24024302

RESUMO

INTRODUCTION: Heat shock proteins (HSPs) and nuclear factor-kappa B (NF-kappaB) have been established as important mediators in lung injury; however, their role in preventing pulmonary toxicity from hyperbaric oxygen (HBO) has not been evaluated. METHODS: We aimed to study the effects of heat shock (HS) injury on hyperbaric hyperoxic lung injury (HHLI) in a rat model and identify a mechanism of protection by evaluating HSP 27 and HSP 70 mRNA and protein levels, NF-kappaB p65, lung injury and oxidative parameters. By varying the times between HS and exposure to HBO, the pathways of interaction between HSPs and NF-kappaB will be further clarified. RESULTS: Our results showed that HS exposure increases the mRNA and protein levels of HSP 27 and HSP 70; HS induced 10-fold increases of HSP 27 (9.77 +/- 0.60) and HSP 70 (10.33 +/- 2.4) within the first 10 h compared to control animals. Lesion scores were higher for the first 16 h after HS, but decreased again after 31 h (N = 7 animals; 5 lesions scores). Protein nitration showed no significant differences between groups exposed to HS or HBO; similarly there was no difference with a combination of both treatments. DISCUSSION: HBO appears to attenuate the HS response by HSP 27 and HSP 70. Histopathology results suggest that HS might mitigate pathology in animals exposed to HS and HBO. No significant effect of HS on HBO-induced HHLI was observed in animals treated with both HS and HBO.


Assuntos
Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Transtornos de Estresse por Calor/patologia , Oxigenoterapia Hiperbárica , Pulmão/patologia , Animais , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP70/genética , Hemorragia/patologia , Peroxidação de Lipídeos , Modelos Animais , Edema Pulmonar/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Fator de Transcrição RelA/metabolismo
13.
J Trauma Acute Care Surg ; 73(5): 1180-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22914081

RESUMO

OBJECTIVES: An advanced hemostatic dressing, Rapid Trauma Hemostat (RTH), was developed using nano-engineered inorganic nanofibers with hemostatic surface properties. METHODS: Yorkshire swine were treated with RTH or Combat Gauze (CBG) to stop bleeding from either an arterial puncture (G-RTH and G-CBG) or a liver lobe laceration (L-RTH and L-CBG). All animals received 500 mL of Hextend at 10 minutes after injury and were monitored for a total time of 180 minutes. RESULTS: Uncontrolled hemorrhage was similar in all animals in both models and was immediately controlled with the application of either dressing. After blood pressure was restored with fluid resuscitation, the RTH hemostatic treatment was less effective than CBG in the groin (puncture) model (rebleeding incidence, four of seven for G-RTH vs. one of seven for G-CBG; p = 0.034) but showed similar efficacy in the liver injury model (lower pressure bleeding). Interestingly, RTH exhibited a trend for higher efficacy in terms of hemostatic plug formation at the end of the experiment (no bleeding occurred after dressing removal) in the liver injury model. CONCLUSION: Overall, RTH was not as effective at stopping high-shear rate (arterial) bleeding, but it presented some advantages for intracavitary treatment with potential for long-term evacuation.


Assuntos
Bandagens , Hemorragia/prevenção & controle , Hemostáticos/administração & dosagem , Nanofibras/administração & dosagem , Compostos de Silício/administração & dosagem , Ferimentos Penetrantes/terapia , Animais , Modelos Animais de Doenças , Artéria Femoral/lesões , Hemorragia/etiologia , Hemorragia/patologia , Fígado/lesões , Propriedades de Superfície , Sus scrofa , Ferimentos Penetrantes/complicações , Ferimentos Penetrantes/patologia
14.
J Neurol Sci ; 318(1-2): 146-54, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22537900

RESUMO

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.


Assuntos
Traumatismos por Explosões/fisiopatologia , Lesões Encefálicas/fisiopatologia , Ativação do Complemento/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Traumatismos por Explosões/imunologia , Traumatismos por Explosões/patologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
15.
J Appl Physiol (1985) ; 111(4): 1007-13, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21757575

RESUMO

Previous studies demonstrated that animals exposed to repeated compression-decompression stress acclimated (i.e., developed reduced susceptibility) to rapid decompression. This study endeavored to characterize inflammatory and stress-related gene expression and signal transduction associated with acclimation to rapid decompression. Rats were divided into four groups: 1) control-sham: pressure naïve rats; 2) acclimation-sham: nine acclimation dives [70 feet seawater (fsw), 30 min]; 3) control-dive: test dive only (175 fsw, 60 min); and 4) acclimation-dive: nine acclimation dives and a test dive. After the test dive, rats were observed for decompression sickness (DCS). Expression of 13 inflammatory and stress-related genes and Akt (or PKB, a serine/threonine protein kinase) and MAPK phosphorylation of lung tissue were examined. The expression of immediate early gene/transcription factor early growth response gene 1 (Egr-1) was observed in both control and acclimation animals with DCS but not in animals without DCS. Increased Egr-1 in control-dive animals with DCS was significantly greater than in acclimation-dive animals with DCS. TNF-α, IL-1ß, IL-6, and IL-10 were significantly elevated in control-DCS animals. Acclimation-DCS animals had increased TNF-α, but there was no change in IL-1ß, IL-6, and IL-10. High levels of Akt phosphorylation were observed in lungs of acclimation-sham, acclimation-dive, and control-dive animals; phosphorylated ERK1/2 was only observed in animals with DCS. This study suggests that activation of ERK1/2 and upregulation of Egr-1 and its target cytokine genes by rapid decompression may play a role in the initiation and progression of DCS. It may be that the downregulated expression of these genes in animals with DCS is associated with previous exposure to repeated compression-decompression cycles. This study represents an initial step toward understanding the molecular mechanisms associated with acclimation to decompression.


Assuntos
Aclimatação/fisiologia , Citocinas/biossíntese , Interleucinas/genética , Pulmão/fisiologia , Estresse Fisiológico/fisiologia , Fator de Necrose Tumoral alfa/genética , Aclimatação/genética , Animais , Citocinas/genética , Descompressão/métodos , Doença da Descompressão/genética , Doença da Descompressão/metabolismo , Regulação para Baixo , Proteína 1 de Resposta de Crescimento Precoce/genética , Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Interleucinas/metabolismo , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/metabolismo , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Fosforilação/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Estresse Fisiológico/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
16.
Cytokine ; 54(1): 20-3, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21251848

RESUMO

Traumatic brain injury (TBI) is associated with significant infectious and inflammatory complications. Though increasing evidence suggests that rFVIIa administration may be efficacious for the pre-hospital treatment of TBI, the FVIIa-tissue factor complex has been shown to be immunologically active. To date the cytokine response to rFVIIa administration for the treatment of TBI has not been evaluated. Twenty anesthetized immature Yorkshire swine underwent fluid percussion TBI. At 15 min following injury, animals were randomized to receive either 90 µg/kg rFVIIa (rFVIIa) or nothing. Animals were observed for 6 h and then euthanized. Plasma and cerebrospinal (CSF) samples were collected at 0 min and 360 min, and ELISA analysis of TNF-α, IL-1ß and IL-10 was performed. Survival in both groups was 100%. Baseline cytokine concentrations were not statistically different between rFVIIa and control animals in plasma or CSF. Animals in both groups did not have significant changes in plasma cytokine concentrations following TBI. Control animals did not demonstrate significant changes from baseline of CSF cytokine concentrations following TBI. The administration of rFVIIa however, resulted in significant increases in CSF TNF-α concentration (232.0 pg/ml ± 75.9 vs 36.4 pg/ml ± 10.4, p = 0.036) and IL-10 concentration (10.7 pg/ml ± 0.6 vs 8.8 pg/ml ± 0.1, p = 0.015). IL-1ß concentrations were not significantly changed over the experimental time course. These results suggest that rFVIIa administration for the treatment of TBI is not immunologically inert, and is associated with increased CSF concentrations of TNF-α and IL-10.


Assuntos
Anti-Inflamatórios/farmacologia , Lesões Encefálicas/sangue , Lesões Encefálicas/líquido cefalorraquidiano , Citocinas/sangue , Citocinas/líquido cefalorraquidiano , Fator VIIa/metabolismo , Animais , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Interleucina-10/sangue , Interleucina-10/líquido cefalorraquidiano , Interleucina-1beta/sangue , Interleucina-1beta/líquido cefalorraquidiano , Masculino , Proteínas Recombinantes/metabolismo , Suínos , Fatores de Tempo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/líquido cefalorraquidiano
17.
J Vasc Surg ; 50(3): 632-9, 639.e1, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19700097

RESUMO

BACKGROUND: The use of mineral (clay) or biologic (chitosan) materials has improved the efficacy of dressings used in the bleeding control of noncompressible areas. A series of novel manufactured products already evaluated in a vascular transection model was further compared in a severe vascular puncture injury model. METHODS: Ten hemostatic dressings were tested in anesthetized Yorkshire swine hemorrhaged for 45 seconds in a femoral arterial puncture model. Application of these dressings was followed by 5 minutes of compression (about 175 mm Hg), and at 15 minutes, 500 mL resuscitation fluid (Hextand) was infused during a 30-minute period. The animals were monitored for a 3-hour experimental observation period. Primary outcomes were incidence of bleeding after dressing application and animal survival. RESULTS: Blood loss was 18.8% +/- 5.2% estimated blood volume (EBV) after 45 seconds of free bleeding. Relative performance of dressings is characterized as groups of dressings that performed similarly. Recurrence of bleeding after application was observed with most dressings and was lower with Woundstat, Celox, X-Sponge, and ACS+ (35% +/- 49%) compared with FP-21, Hemcon, Chitoflex, and Bloodstop (79% +/- 43%; P < .01). Blood loss after treatment was 25.3% +/- 18.4% EBV for the top four dressings and 53.0% +/- 18.4% EBV for the bottom four (P < .05). Survival was higher for top four vs bottom four dressings (78% +/- 12% vs 25% +/- 0%, respectively; P < .01). Overall performance of these dressings according to survival, incidence of bleeding, and post-treatment blood loss, yielded similar ranking as with a previously tested transection injury model. CONCLUSIONS: The findings indicated that the efficacy of Woundstat, Celox, X-Sponge, and ACS+ were similar and superior in improving survival, hemostasis, and maintenance of mean arterial pressure in an actively bleeding wound caused in this severe vascular injury model.


Assuntos
Bandagens , Artéria Femoral/lesões , Virilha/irrigação sanguínea , Hemorragia/prevenção & controle , Técnicas Hemostáticas/instrumentação , Ferimentos e Lesões/terapia , Animais , Pressão Sanguínea , Volume Sanguíneo , Modelos Animais de Doenças , Desenho de Equipamento , Hemorragia/etiologia , Teste de Materiais , Punções , Recidiva , Índice de Gravidade de Doença , Suínos , Fatores de Tempo , Ferimentos e Lesões/etiologia
18.
J Trauma ; 66(2): 365-76, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19204509

RESUMO

BACKGROUND: Vasoactivity has hampered progress of hemoglobin-based oxygen carriers (HBOCs) due to concern for adverse blood pressure responses and secondary complications. A recent formulation, highly polymerized HBOC-201 (Biopure, Cambridge, MA), has been found to be less vasoactive than prior less polymerized formulations, and to improve outcome in animal models of hemorrhagic shock (HS) compared with standard resuscitation fluids. HBOCs are envisioned to have life- saving potential for severe trauma patients for whom death due to HS is common despite transport to level I trauma centers. As part of a benefit:risk analysis for a proposed clinical trial of HBOC-201 in patients with traumatic HS, we analyzed data from a previous phase III clinical trial of this HBOC that involved orthopedic surgery patients, for vasoactivity and related effects, with focus on patients more representative of the trauma population. STUDY DESIGN: In a previous phase III study involving orthopedic surgery patients, HEM-0115, consented/stabilized patients were randomized to receive HBOC-201 (N = 350) (up to ten 30 g Hb units) or red blood cells (RBC) (N = 338) (up to 9 units) at the first transfusion decision. Systolic blood pressure (SBP) responses, key system and individual adverse events (AEs) and serious adverse events, and cardiac biomarker elevation incidences, were compared in the overall population and subpopulations with stable trauma, hypotension, and with age stratification (Student's t and Fisher's exact tests, significance p < 0.05). RESULTS: Mild to moderate peak SBP responses were common in HBOC-201 subjects and more common than with RBC in the overall population (mean, 60.8 years old), but less frequent in HBOC-201 subjects with stable trauma, younger age (<50 years old), and hypotension, in whom group differences were narrowed. SBP Delta responses were more common with HBOC-201 than RBC in the overall population, but not in subjects with stable trauma and <50 year olds, in whom response rates were lower. In the overall population, AEs were more common than with RBC in most systems (also, hypertension and stroke); only cardiac system serious adverse events were more common with HBOC-201. In contrast, there were few significant group differences in stable trauma, hypotensive, and <70 and especially <50-year-old subjects, in whom AE incidences were generally lower. A disproportionate number of key AEs occurred in elderly subjects. Troponin (but not CK-MB) elevation was more frequent with HBOC-201 than RBC in the overall population but not in <50 year olds, and was not associated with acute coronary syndrome (ACS) or death. CONCLUSIONS: Our limited HEM-0115 safety analysis shows that key potentially vasoactivity-related adverse safety signals were more frequent with HBOC-201 than RBC in older patients undergoing orthopedic surgery with rapid access to safe blood transfusions. That incidences of these safety signals were generally lower and group differences narrowed in subpopulations with stable trauma, hypotension, and younger age, suggests an acceptable safety profile in younger acute trauma populations, especially in settings where rapid access to safe blood transfusions is unavailable; confirmation in controlled clinical trials is urgently warranted.


Assuntos
Substitutos Sanguíneos/farmacologia , Transfusão de Eritrócitos/estatística & dados numéricos , Hemoglobinas/farmacologia , Ortopedia , Adulto , Idoso , Substitutos Sanguíneos/efeitos adversos , Ensaios Clínicos como Assunto , Transfusão de Eritrócitos/efeitos adversos , Feminino , Hemoglobinas/efeitos adversos , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Risco , Método Simples-Cego
19.
Shock ; 32(3): 325-31, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19174737

RESUMO

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


Assuntos
Acetilcisteína/análogos & derivados , Antioxidantes/uso terapêutico , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/imunologia , Pneumonia/tratamento farmacológico , Pneumonia/imunologia , Acetilcisteína/uso terapêutico , Animais , Antígeno CD11b/genética , Quimiocina CCL2/genética , Quimiocina CXCL1/genética , Glutationa Redutase/genética , Heme Oxigenase-1/metabolismo , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Proteínas Inflamatórias de Macrófagos/genética , Masculino , Infiltração de Neutrófilos/imunologia , Infiltração de Neutrófilos/fisiologia , Peroxidase/metabolismo , Pneumonia/metabolismo , Pneumonia/patologia , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/genética
20.
J Trauma ; 65(5): 1140-5; discussion 1145, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19001988

RESUMO

BACKGROUND: A critical immediate determinant of survival after exposure to blast overpressure (BOP) is cardiovascular and respiratory impairment related to disruption of the alveolar septa and pulmonary capillaries and resulting in acute pulmonary hemorrhage. Hemoglobin (Hb) released from red cells can contribute to lethality by activation of oxidative stress reactions and severe vasoconstriction associated with hypoperfusion in the pulmonary microcirculation. Heme oxygenase-1 (HO-1) is activated by hemin, a product of Hb degradation and may confer protection against hemoglobin-mediated oxidative cell and tissue damage. METHODS: Rats were injected intraperitoneally with hemin (50 mg/kg) or phosphate buffered saline (PBS). Twenty hours later, animals were placed in a shock tube and exposed to blast overpressure with mean intensity of approximately 160 kPa. Nonblasted sham-injected animals served as controls. RESULTS: HO-1 mRNA and HO-1 protein in lungs was induced by injection of hemin. Exposure to blast resulted in confluent lung hemorrhage and mortality ( approximately 65%). Hemin injection significantly improved the survival rate of animals compared with PBS injected animals (p = 0.01). CONCLUSIONS: The protection by hemin against blast may involve antioxidative and vasodilatory effects of HO-1, although, the precise mechanisms of the protection are unknown.


Assuntos
Antioxidantes/farmacologia , Traumatismos por Explosões/mortalidade , Traumatismos por Explosões/prevenção & controle , Heme Oxigenase-1/farmacologia , Hemina/farmacologia , Vasodilatadores/farmacologia , Animais , Traumatismos por Explosões/complicações , Heme Oxigenase-1/biossíntese , Hemina/metabolismo , Hemorragia/etiologia , Hemorragia/prevenção & controle , Pulmão/efeitos dos fármacos , Lesão Pulmonar/etiologia , Lesão Pulmonar/prevenção & controle , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA