Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 167, 2024 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365798

RESUMEN

Targeting drugs to the mitochondrial level shows great promise for acute and chronic treatment of traumatic brain injury (TBI) in both military and civilian sectors. Perhaps the greatest obstacle to the successful delivery of drug therapies is the blood brain barrier (BBB). Intracerebroventricular and intraparenchymal routes may provide effective delivery of small and large molecule therapies for preclinical neuroprotection studies. However, clinically these delivery methods are invasive, and risk inadequate exposure to injured brain regions due to the rapid turnover of cerebral spinal fluid. The direct intranasal drug delivery approach to therapeutics holds great promise for the treatment of central nervous system (CNS) disorders, as this route is non-invasive, bypasses the BBB, enhances the bioavailability, facilitates drug dose reduction, and reduces adverse systemic effects. Using the intranasal method in animal models, researchers have successfully reduced stroke damage, reversed Alzheimer's neurodegeneration, reduced anxiety, improved memory, and delivered neurotrophic factors and neural stem cells to the brain. Based on literature spanning the past several decades, this review aims to highlight the advantages of intranasal administration over conventional routes for TBI, and other CNS disorders. More specifically, we have identified and compiled a list of most relevant mitochondria-targeted neuroprotective compounds for intranasal administration based on their mechanisms of action and pharmacological properties. Further, this review also discusses key considerations when selecting and testing future mitochondria-targeted drugs given intranasally for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Neuroprotección , Animales , Administración Intranasal , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Encéfalo , Barrera Hematoencefálica
2.
Anal Biochem ; 680: 115301, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37673410

RESUMEN

Owing to evidence that mitochondrial dysfunction plays a dominant role in the traumatic brain injury (TBI) pathophysiology, the Western blot (WB) based immunoblotting method is widely employed to identify changes in the mitochondrial protein expressions after neurotrauma. In WB method, the housekeeping proteins (HKPs) expression is routinely used as an internal control for sample normalization. However, the traditionally employed HKPs can be susceptible to complex cascades of TBI pathogenesis, leading to their inconsistent expression. Remarkably, our data illustrated here that mitochondrial HKPs, including Voltage-dependent anion channels (VDAC), Complex-IV, Cytochrome C and Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) yielded altered expressions following penetrating TBI (PTBI) as compared to Sham. Therefore, our goal was to identify more precise normalization procedure in WB. Adult male Sprague Dawley rats (N = 6 rats/group) were used to perform PTBI, and the novel REVERT Total Protein (RTP) method was used to quantify mitochondrial protein load consistency between samples at 6 h and 24 h post-injury. Notably, the RTP method displayed superior protein normalization compared to HKPs method with higher sensitivity at both time-points between experimental groups. Our data favors application of RTP based normalization to accurately quantify protein expression where inconsistent HKPs may be evident in neuroscience research.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Masculino , Animales , Ratas , Ratas Sprague-Dawley , Western Blotting , Proteínas Mitocondriales , Mitocondrias
3.
Microcirculation ; 25(3): e12441, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29356218

RESUMEN

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.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Fluorocarburos/uso terapéutico , Oxígeno/sangre , Animales , Arteriolas/fisiología , Encéfalo/metabolismo , Circulación Cerebrovascular , Microscopía Intravital , Oxígeno/administración & dosificación , Ratas , Vasoconstricción/efectos de los fármacos
4.
Microvasc Res ; 107: 83-90, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27287870

RESUMEN

Sanguinate, a polyethylene glycol-conjugated carboxyhemoglobin, was investigated for cerebral vasoactivity in healthy male Sprague-Dawley rats (Study 1) and for its ability to increase brain tissue oxygen pressure (PbtO2) after controlled cortical impact (CCI) - traumatic brain injury (TBI) (Study 2). In both studies ketamine-acepromazine anesthetized rats were ventilated with 40% O2. In Study 1, a cranial window was used to measure the diameters of medium - (50-100µm) and small-sized (<50µm) pial arterioles before and after four serial infusions of Sanguinate (8mL/kg/h, cumulative 16mL/kg IV), volume-matched Hextend, or normal saline. In Study 2, PbtO2 was measured using a phosphorescence quenching method before TBI, 15min after TBI (T15) and then every 10min thereafter for 155min. At T15, rats received either 8mL/kg IV Sanguinate (40mL/kg/h) or no treatment (saline, 4mL/kg/h). Results showed: 1) in healthy rats, percentage changes in pial arteriole diameter were the same among the groups, 2) in TBI rats, PbtO2 decreased from 36.5±3.9mmHg to 19.8±3.0mmHg at T15 in both groups after TBI and did not recover in either group for the rest of the study, and 3) MAP increased 16±4mmHg and 36±5mmHg after Sanguinate in healthy and TBI rats, respectively, while MAP was unchanged in control groups. In conclusion, Sanguinate did not cause vasoconstriction in the cerebral pial arterioles of healthy rats but it also did not acutely increase PbtO2 when administered after TBI. Sanguinate was associated with an increase in MAP in both studies.


Asunto(s)
Arteriolas/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Carboxihemoglobina/farmacología , Circulación Cerebrovascular/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Oxígeno/metabolismo , Piamadre/irrigación sanguínea , Sustitutos del Plasma/farmacología , Polietilenglicoles/farmacología , Animales , Presión Arterial/efectos de los fármacos , Arteriolas/metabolismo , Arteriolas/fisiopatología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Carboxihemoglobina/análogos & derivados , Carboxihemoglobina/toxicidad , Modelos Animales de Enfermedad , Derivados de Hidroxietil Almidón/farmacología , Masculino , Microcirculación/efectos de los fármacos , Sustitutos del Plasma/toxicidad , Polietilenglicoles/toxicidad , Ratas Sprague-Dawley , Factores de Tiempo , Vasoconstricción/efectos de los fármacos
5.
Lung ; 194(6): 945-957, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27704259

RESUMEN

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.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Fluorocarburos/administración & dosificación , Oxígeno/sangre , Equilibrio Ácido-Base , Lesión Pulmonar Aguda/inducido químicamente , Administración Intravenosa , Animales , Presión Arterial/efectos de los fármacos , Análisis de los Gases de la Sangre , Modelos Animales de Enfermedad , Femenino , Fluorocarburos/efectos adversos , Ácido Láctico/sangre , Masculino , Ácido Oléico , Presión Esfenoidal Pulmonar/efectos de los fármacos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Índice de Severidad de la Enfermedad , Tasa de Supervivencia , Porcinos
6.
Antioxidants (Basel) ; 13(3)2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38539837

RESUMEN

Traumatic brain injury (TBI) is a major global health problem that affects both civilian and military populations worldwide. Post-injury acute, sub-acute, and chronic progression of secondary injury processes may contribute further to other neurodegenerative diseases. However, there are no approved therapeutic options available that can attenuate TBI-related progressive pathophysiology. Recent advances in preclinical research have identified that mitochondria-centric redox imbalance, bioenergetics failure and calcium dysregulation play a crucial role in secondary injury progression after TBI. Mitochondrial antioxidants play an important role in regulating redox homeostasis. Based on the proven efficacy of preclinical and clinical compounds and targeting numerous pathways to trigger innate antioxidant defense, we may be able to alleviate TBI pathology progression by primarily focusing on preserving post-injury mitochondrial and cerebral function. In this review, we will discuss novel mitochondria-targeted antioxidant compounds, which offer a high capability of successful clinical translation for TBI management in the near future.

7.
J Neurotrauma ; 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38874230

RESUMEN

Traumatic brain injury (TBI)-induced intracerebral hematoma is a major driver of secondary injury pathology such as neuroinflammation, cerebral edema, neurotoxicity, and blood-brain barrier dysfunction, which contribute to neuronal loss, motor deficits, and cognitive impairment. Cluster of differentiation 47 (CD47) is an antiphagocytic cell surface protein inhibiting hematoma clearance. This study was designed to evaluate the safety and efficacy of blockade of CD47 via intravenous (i.v.) administration of anti-CD47 antibodies following penetrating ballistic-like brain injury (PBBI) with significant traumatic intracerebral hemorrhage (tICH). The pharmacokinetic (PK) profile of the anti-CD47 antibody elicited that antibody concentration decayed over 7 days post-administration. Blood tests and necropsy analysis indicated no severe adverse events following treatment. Cerebral hemoglobin levels were significantly increased after injury, however, anti-CD47 antibody administration at 0.1 mg/kg resulted in a significant reduction in cerebral hemoglobin levels at 72 h post-administration, indicating augmentation of hematoma clearance. Immunohistochemistry assessment of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule 1 (IBA1) demonstrated a significant reduction of GFAP levels in the lesion core and peri-lesional area. Based on these analyses, the optimal dose was identified as 0.1 mg/kg. Lesion volume showed a reduction following treatment. Rotarod testing revealed significant motor deficits in all injured groups but no significant therapeutic benefits. Spatial learning performance revealed significant deficits in all injured groups, which were significantly improved by the last testing day. Anti-CD47 antibody treated rats showed significantly improved attention deficits, but not retention scores. These results provide preliminary evidence that blockade of CD47 using i.v. administration of anti-CD47 antibodies may serve as a potential therapeutic for TBI with ICH.

8.
Exp Neurol ; 369: 114533, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37666386

RESUMEN

Traumatic brain injury (TBI) leads to long-term impairments in motor and cognitive function. TBI initiates a secondary injury cascade including a neuro-inflammatory response that is detrimental to tissue repair and limits recovery. Anti-inflammatory corticosteroids such as dexamethasone can reduce the deleterious effects of secondary injury; but challenges associated with dosing, administration route, and side effects have hindered their clinical application. Previously, we developed a hydrolytically degradable hydrogel (PEG-bis-AA/HA-DXM) composed of poly (ethylene) glycol-bis-(acryloyloxy acetate) (PEG-bis-AA) and dexamethasone-conjugated hyaluronic acid (HA-DXM) for local and sustained dexamethasone delivery. In this study, we evaluated the effect of locally applied PEG-bis-AA/HA-DXM hydrogel on secondary injury and motor function recovery after moderate controlled cortical impact (CCI) TBI. Hydrogel treatment significantly improved motor function evaluated by beam walk and rotarod tests compared to untreated rats over 7 days post-injury (DPI). We also observed that the hydrogel treatment reduced lesion volume, inflammatory response, astrogliosis, apoptosis, and increased neuronal survival compared to untreated rats at 7 DPI. These results suggest that PEG-bis-AA/HA-DXM hydrogels can mitigate secondary injury and promote motor functional recovery following moderate TBI.

9.
Shock ; 60(2): 248-254, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37267223

RESUMEN

ABSTRACT: Several studies have demonstrated the clinical utility of tranexamic acid (TXA) for use in trauma patients presenting with significant hemorrhage. Tranexamic acid is an antifibrinolytic that inhibits plasminogen activation, and plasmin activity has been shown to mitigate blood loss and reduce all-cause mortality in the absence of adverse vascular occlusive events. Recent clinical developments indicate TXA is safe to use in patients with concomitant traumatic brain injury (TBI); however, the prehospital effects are not well understood. Importantly, TXA has been associated with seizure activity. Therefore, this study sought to evaluate the effects of early administration of TXA on neurological recovery and electroencephalogram (EEG) abnormalities following penetrating TBI with concomitant hypoxemia and hemorrhagic shock. We hypothesized that early administration of TXA will provide hemodynamic stabilization and reduce intracerebral hemorrhage, which will result in improved neurological function. To test this hypothesis, Sprague-Dawley rats received a unilateral, frontal penetrating ballistic-like brain injury by inserting a probe into the frontal cortex of the anesthetized rat. Five minutes following brain injury, animals underwent 30 min of respiratory distress and 30 min of hemorrhage. Upon completion of the hemorrhage phase, animals received the initial dose of drug intravenously over 10 min after which the prehospital phase was initiated. During the prehospital phase, animals received autologous shed whole blood as needed to maintain a MAP of 65 mm Hg. After 90 min, "in-hospital" resuscitation was performed by administering the remaining shed whole blood providing 100% oxygen for 15 min. Upon recovery from surgery, animals were administered their second dose of vehicle or TXA intravenously over 8 h. Tranexamic acid induced an early improvement in neurologic deficit, which was statistically significant compared with vehicle at 24, 48, and 72 h at three doses tested. Analysis of cerebral hemoglobin content and intracerebral lesion progression revealed 100 mg/kg provided the optimal effects for improvement of neuropathology and was continued for determination of adverse treatment effects. We observed no exacerbation of cerebral thrombosis, but TXA treatment caused an increased risk of EEG abnormalities. These results suggest that TXA following polytrauma with concomitant brain injury may provide mild neuroprotective effects by preventing lesion progression, but this may be associated with an increased risk of abnormal EEG patterns. This risk may be associated with TXA inhibition of glycine receptors and may warrant additional considerations during the use of TXA in patients with severe TBI.


Asunto(s)
Antifibrinolíticos , Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Traumatismos Penetrantes de la Cabeza , Traumatismo Múltiple , Ácido Tranexámico , Animales , Ratas , Ácido Tranexámico/uso terapéutico , Ratas Sprague-Dawley , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Antifibrinolíticos/uso terapéutico , Traumatismo Múltiple/complicaciones , Traumatismo Múltiple/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Electroencefalografía/efectos adversos , Fibrina
10.
Free Radic Biol Med ; 198: 44-58, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36758906

RESUMEN

Traumatic Brain Injury (TBI) is caused by the external physical assaults damages the brain. It is a heterogeneous disorder that remains a leading cause of death and disability in the military and civilian population of the United States. Preclinical investigations of mitochondrial responses in TBI have ascertained that mitochondrial dysfunction is an acute indicator of cellular damage and plays a pivotal role in long-term injury progression through cellular excitotoxicity. The current study was designed to provide an in-depth evaluation of mitochondrial endpoints with respect to redox and calcium homeostasis, and cell death responses following penetrating TBI (PTBI). To evaluate these pathological cascades, anesthetized adult male rats (N = 6/group) were subjected to either 10% unilateral PTBI or Sham craniectomy. Animals were euthanized at 24 h post-PTBI, and purified mitochondrial fractions were isolated from the brain injury core and perilesional areas. Overall, increased reactive oxygen and nitrogen species (ROS/RNS) production, and elevated oxidative stress markers such as 4-hydroxynonenal (4-HNE), 3-nitrotyrosine (3-NT), and protein carbonyls (PC) were observed in the PTBI group compared to Sham. Mitochondrial antioxidants such as glutathione, peroxiredoxin (PRX-3), thioredoxin (TRX), nicotinamide adenine dinucleotide phosphate (NADPH), superoxide dismutase (SOD), and catalase (CAT) levels were significantly decreased after PTBI. Likewise, PTBI mitochondria displayed significant loss of Ca2+ homeostasis, early opening of mitochondrial permeability transition pore (mPTP), and increased mitochondrial swelling. Both, outer and inner mitochondrial membrane integrity markers, such as voltage-dependent anion channels (VDAC) and cytochrome c (Cyt C) expression were significantly decreased following PTBI. The apoptotic cell death was evidenced by significantly decreased B-cell lymphoma-2 (Bcl-2) and increased glyceraldehyde 3-phosphate dehydrogenase (GAPDH) expression after PTBI. Collectively, current results highlight the comprehensive picture of mitochondria-centric acute pathophysiological responses following PTBI, which may be utilized as novel prognostic indicators of disease progression and theragnostic indicators for evaluating neuroprotection therapeutics following TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Calcio , Ratas , Masculino , Animales , Calcio/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Oxidación-Reducción , Apoptosis/fisiología , Mitocondrias/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-21806503

RESUMEN

In a previous dose escalation study our group found that combining 90µg/kg rFVIIa with HBOC-201 reduced blood loss and improved physiologic parameters compared to HBOC alone. In this follow-up study in a swine liver injury model, we found that while there were no adverse hematology effects and trends observed in the previous study were confirmed, statistical significance could not be reached. Additional pre-clinical studies are indicated to identify optimal components of a multifunctional blood substitute for clinical use in trauma.


Asunto(s)
Factor VIIa/farmacología , Fluidoterapia/métodos , Hemoglobinas/farmacología , Hospitales , Proteínas Recombinantes/farmacología , Choque Hemorrágico/tratamiento farmacológico , Porcinos , Animales , Sustitutos Sanguíneos/farmacología , Volumen Sanguíneo/efectos de los fármacos , Interacciones Farmacológicas , Factor VIIa/uso terapéutico , Femenino , Hemoglobinas/uso terapéutico , Masculino , Oxígeno/metabolismo , Proteínas Recombinantes/uso terapéutico , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patología , Choque Hemorrágico/fisiopatología , Análisis de Supervivencia
12.
Injury ; 53(11): 3596-3604, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36163203

RESUMEN

INTRODUCTION: Traumatic brain injuries (TBI) represent a significant percentage of critical injuries in military conflicts. Following injury, wounded warfighters are often subjected to multiple aeromedical evacuations (AE) and associated hypobaria, yet the impact in TBI patients remains to be characterized. This study evaluated the impact of two consecutive simulated AEs in a fluid-percussion TBI model in swine to characterize these effects. METHODS: Following instrumentation, anesthetized Yorkshire swine underwent a frontal TBI via fluid-percussion. A hypobaric chamber was then used to simulate AE at simulated cabin pressure equivalent to 8000ft (hypobaria) in a 6 h initial flight on day 3, followed by a 9 h flight on day 6, and were monitored for 14 days. Animals in the normobaria group were subjected to the same steps at sea level while Sham animals in both groups were instrumented but not injured. Parameters measured included physiologic response, intracranial pressure (ICP), hematology, chemistry, and serum cytokines. Histopathology of brain, lung, intestine, and kidney was performed, as well as fluorojade staining to evaluate neurodegeneration. All animals were divided into sub-groups by block randomization utilizing a 2-way ANOVA to analyze independent variables. RESULTS: Survival was 100% in all groups. Physiologic parameters were largely similar across groups as well during both 6 and 9 h AE. Animals exposed to hypobaria in both the TBI and Sham groups had elevated heart rate (HR) during the 6 h flight (p<0.05). Three animals in the TBI hypo group demonstrated leukocytosis with histologic evidence of meningeal inflammatory response. Expression of serum cytokines was low across all groups. No significant neuronal degeneration was identified in areas away from the site of injury. CONCLUSION: Aeromedical evacuation in swine was not associated with significant differences in physiologic measures, cytokine expression or levels of neuronal degeneration. Histological examination revealed higher risk of meningeal inflammatory response and leucocytosis in swine exposed to hypobaria.


Asunto(s)
Ambulancias Aéreas , Lesiones Traumáticas del Encéfalo , Animales , Citocinas , Modelos Animales de Enfermedad , Presión Intracraneal , Porcinos
13.
Cytokine ; 54(1): 20-3, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21251848

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Lesiones Encefálicas/sangre , Lesiones Encefálicas/líquido cefalorraquídeo , Citocinas/sangre , Citocinas/líquido cefalorraquídeo , Factor VIIa/metabolismo , Animales , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Interleucina-10/sangre , Interleucina-10/líquido cefalorraquídeo , Interleucina-1beta/sangre , Interleucina-1beta/líquido cefalorraquídeo , Masculino , Proteínas Recombinantes/metabolismo , Porcinos , Factores de Tiempo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/líquido cefalorraquídeo
14.
Artículo en Inglés | MEDLINE | ID: mdl-21133651

RESUMEN

Sodium nitrite (NaNO(2)) was evaluated in a 55% EBV hemorrhage swine model to mitigate the increased blood pressure due to HBOC-201. Animals were resuscitated by three 10 ml/kg infusions of either HBOC-201 or Hextend with and without NaNO(2). All vital signs, coagulation and blood chemistry were measured for 2 hr. HBOC-201-vasoconstriction was attenuated only after the first 10.8 µmol/kg NaNO(2) infusion. Complete abolition was obtained with the highest 3 NaNO(2) dose, but side effects were observed. There was no reduction in platelet function due to NaNO(2). NaNO(2) ability to reduce HBOC-201 vasoactivity was transient and 10.8 µmol/kg NaNO(2) seems an acceptable dose for further investigation.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/fisiopatología , Hemoglobinas/farmacología , Hemorragia/fisiopatología , Nitrito de Sodio/farmacología , Porcinos , Animales , Volumen Sanguíneo/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hemoglobinas/administración & dosificación , Hemostasis/efectos de los fármacos , Resucitación , Nitrito de Sodio/administración & dosificación , Signos Vitales/efectos de los fármacos
15.
Artículo en Inglés | MEDLINE | ID: mdl-20645681

RESUMEN

Exsanguinating hemorrhage and unavailability of blood are major problems in pre-hospital trauma care. We investigated if combining rFVIIa with HBOC-201 reduces blood loss and improves physiologic parameters compared to HBOC alone. Swine underwent liver injury and were resuscitated with HBOC-201 alone or HBOC+90, 180 or 360 µg/kg rFVIIa before hospital arrival at 240 min; animals survived to 72 hours. Blood loss was reduced; MAP, CI, transcutaneous oxygen saturation, and 72-hour survival improved in the 90 and 180 µg/kg rFVIIa groups. Lactate was cleared faster in the HBOC+rFVIIa 90 µg/kg group. Verification in a large, well-powered study is indicated.


Asunto(s)
Sustitutos Sanguíneos/administración & dosificación , Factor VIIa/administración & dosificación , Fluidoterapia/métodos , Hemoglobinas/administración & dosificación , Hemorragia/terapia , Animales , Análisis de los Gases de la Sangre/métodos , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Frecuencia Cardíaca/fisiología , Hemorragia/mortalidad , Ácido Láctico/análisis , Ácido Láctico/sangre , Hígado/lesiones , Masculino , Proteínas Recombinantes/administración & dosificación , Resucitación , Tasa de Supervivencia , Porcinos
16.
Mil Med ; 185(Suppl 1): 57-66, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-32074309

RESUMEN

INTRODUCTION: Rapid aeromedical evacuation (AE) is standard of care in current conflicts. However, not much is known about possible effects of hypobaric conditions. We investigated possible effects of hypobaria on organ damage in a swine model of acute lung injury. METHODS: Lung injury was induced in anesthetized swine via intravenous oleic acid infusion. After a stabilization phase, animals were subjected to a 4 hour simulated AE at 8000 feet (HYPO). Control animals were kept at normobaria. After euthanasia and necropsy, organ damage was assessed by combined scores for hemorrhage, inflammation, edema, necrosis, and microatelectasis. RESULTS: Hemodynamic, neurological, or hematologic measurements were similar prior to transport. Hemodynamic instability became apparent during the last 2 hours of transport in the HYPO group. Histological injury scores in the HYPO group were higher for all organs (lung, kidney, liver, pancreas, and adrenal glands) except the brain, with the largest difference in the lungs (P < 0.001). CONCLUSIONS: Swine with mild acute lung injury subjected to a 4 hour simulated AE showed more injury to most organs and, in particular, to the lungs compared with ground transport. This may exacerbate otherwise subclinical pathology and, eventually, manifest as abnormalities in gas exchange or possibly end-organ function.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Insuficiencia Multiorgánica/patología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/fisiopatología , Medicina Aeroespacial/métodos , Animales , Modelos Animales de Enfermedad , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/fisiopatología , Ácido Oléico/efectos adversos , Ácido Oléico/farmacología , Porcinos/lesiones , Porcinos/fisiología
17.
Med Sci (Basel) ; 8(4)2020 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-32992571

RESUMEN

Pre-hospital treatment of traumatic brain injury (TBI) with co-existing polytrauma is complicated by requirements for intravenous fluid volume vs. hypotensive resuscitation. A low volume, small particle-size-oxygen-carrier perfluorocarbon emulsion NVX-428 (dodecafluoropentane emulsion; 2% w/v) could improve brain tissue with minimal additional fluid volume. This study examined whether the oxygen-carrier NVX-428 shows safety and efficacy for pre-hospital treatment of TBI. Anesthetized swine underwent fluid percussion injury TBI and received 1 mL/kg IV NVX-428 (TBI-NVX) at 15 min (T15) or normal saline (no-treatment) (TBI-NON). Similarly, uninjured swine received NVX-428 (SHAM-NVX) or normal saline (SHAM-NON). Animals were monitored and measurements were taken for physiological and neurological parameters before euthanasia at the six-hour mark (T360). Histopathological analysis was performed on paraffin embedded tissues. Physiological, biochemical and blood gas parameters were not different, with the exception of a significant but transient increase in mean pulmonary artery pressure observed in the TBI-experimental group immediately after drug administration. There were no initial differences in brain oxygenation at baseline, but over time oxygen decreased ~50% in both TBI groups. Histological brain injury scores were similar between TBI-NVX and TBI-NON, although a number of subcategories (spongiosis-ischemic/dead neurons-hemorrhage-edema) in TBI-NVX had a tendency for lower scores. The cerebellum showed significantly lower spongiosis and ischemic/dead neuron injury scores and a lower number of Fluoro-Jade-B-positive cerebellar-Purkinje-cells after NVX-428 treatment compared to controls. NVX-428 may assist in mitigating secondary cellular brain damage.

18.
J Trauma Acute Care Surg ; 87(1): 205-213, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31033888

RESUMEN

BACKGROUND: Aeromedical evacuation (AE) is often used as a rapid and effective way to evacuate patients. However, little is known about the possible effects of AE on patients with blast and traumatic brain injury. In the current study, we used blast overpressure (BOP) as a method to introduce traumatic brain injury in rats and investigated the effects of hypobaria during AE on histology and inflammatory response. METHODS: Animals were exposed to a 12-hour flight 2 days after BOP and euthanized 48 hours after flight. Control animals were kept at normobaria. RESULTS: Overall, BOP animals exposed to flight demonstrated higher histopathologic injury scores as compared to control animals in lungs, brain, kidney, heart, and intestine. The BOP animals exposed to normobaria exhibited a proinflammatory response compared to those that were not blasted, an observation that was not seen in BOP animals exposed to hypobaria. CONCLUSION: These data suggest that AE 48 hours post blast may lead to impairment in the inflammatory process and worsening of long-term outcomes. LEVEL OF EVIDENCE: Animal research, level II.


Asunto(s)
Ambulancias Aéreas , Presión Atmosférica , Traumatismos por Explosión/patología , Inflamación/etiología , Heridas y Lesiones/patología , Animales , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/etiología , Lesiones Traumáticas del Encéfalo/patología , Intestinos/patología , Riñón/patología , Pulmón/patología , Masculino , Miocardio/patología , Ratas , Ratas Sprague-Dawley
19.
J Trauma Acute Care Surg ; 86(1): 116-122, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29985235

RESUMEN

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.


Asunto(s)
Ambulancias Aéreas/estadística & datos numéricos , Barotrauma/complicaciones , Encéfalo/patología , Pulmón/patología , Altitud , Animales , Presión Atmosférica , Análisis de los Gases de la Sangre/métodos , Lesiones Encefálicas/etiología , Modelos Animales de Enfermedad , Edema/patología , Femenino , Hemodinámica/fisiología , Hemorragia/patología , Inflamación/inmunología , Inflamación/patología , Lesión Pulmonar/etiología , Masculino , Necrosis/patología , Atelectasia Pulmonar/patología , Porcinos
20.
Blood Coagul Fibrinolysis ; 19(7): 669-77, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18832908

RESUMEN

The combination of traumatic injury, hemorrhage, and fluid resuscitation results in consumption and dilution of coagulation factors, adversely impacting hematology outcome in trauma patients. The hemostatic effects of escalating doses of recombinant factor VIIa added to hemoglobin-based oxygen carrier-201 were assessed as prehospital fluid resuscitation in swine with severe uncontrolled hemorrhage. Swine underwent liver injury causing severe uncontrolled hemorrhage and shock. During a 4-h prehospital phase, either hypotensive or tachycardic, or both, animals were resuscitated with hemoglobin-based oxygen carrier-201 without (0x) or with escalating doses of recombinant factor VIIa [90 microg/kg (1x), 180 microg/kg (2x), or 360 microg/kg (4x)]. The animals received one initial full dose of 10 ml/kg at 15 min and up to four doses of 5 ml/kg thereafter. From 4 to 72 h (hospital phase), animals received either transfusions or isotonic saline or both as needed. Hematology profile (complete blood count), thromboelastography, in-vitro bleeding (platelet function analyzer), and coagulation (prothrombin time) were measured and the results were compared using mixed statistical models. In all groups, dilutional coagulopathy was evidenced by reduced hematocrit, platelets, and thromboelastography-maximum amplitude, and increased platelet function analyzer closure time and thromboelastography-reaction time. During the prehospital phase, hemoglobin-based oxygen carrier-201 restored hemoglobin in all groups. Recombinant factor VIIa decreased prothrombin time in recombinant factor VIIa groups compared with the hemoglobin-based oxygen carrier-201 group (P < 0.01). Unexpectedly, increasing recombinant factor VIIa dosage tended to increase fluid requirement (P > 0.05). Compared with hemoglobin-based oxygen carrier, 1x recombinant factor VIIa tended to decrease blood loss, lactate and thromboelastography-reaction time at 24 h but the 4x group increased these parameters. Platelets and thromboelastography-maximum amplitude decreased (P < 0.01) with the 4x group. In swine with severe uncontrolled hemorrhage, prehospital resuscitation with escalating doses of recombinant factor VIIa in combination with hemoglobin-based oxygen carrier-201 did not change survival or hemostasis. However, there were trends toward possible benefits of low recombinant factor VIIa doses, whereas high recombinant factor VIIa doses adversely affected hemostasis.


Asunto(s)
Sustitutos Sanguíneos/farmacología , Factor VIIa/farmacología , Hemoglobinas/farmacología , Hígado/lesiones , Choque Hemorrágico/tratamiento farmacológico , Animales , Tiempo de Sangría , Coagulación Sanguínea/efectos de los fármacos , Transfusión Sanguínea , Modelos Animales de Enfermedad , Fluidoterapia , Pruebas Hematológicas , Infusiones Intravenosas , Recuento de Plaquetas , Tiempo de Protrombina , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacología , Resucitación/métodos , Choque Hemorrágico/sangre , Porcinos , Porcinos Enanos , Tromboelastografía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA