Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Neurosci Lett ; 609: 152-8, 2015 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-26483321

RESUMO

Dephosphorylation of phosphorylated Tau (pTau) protein, which is essential for the preservation of neuronal microtubule assemblies and for protection against trauma-induced tauopathy and chronic traumatic encephalopathy (CTE), is primarily achieved in brain by tissue non-specific alkaline phosphatase (TNAP). Paired helical filaments (PHFs) and Tau isolated from Alzheimer's disease (AD) patients' brains have been shown to form microtubule assemblies with tubulin only after treatment with TNAP or protein phosphatase-2A, 2B and -1, suggesting that Tau protein in the PHFs of neurons in AD brain is hyperphosphorylated, which prevents microtubule assembly. Using blast or weight drop models of traumatic brain injury (TBI) in rats, we observed pTau accumulation in the brain as early as 6h post-injury and further accumulation which varied regionally by 24h post-injury. The pTau accumulation was accompanied by reduced TNAP expression and activity in these brain regions and a significantly decreased plasma total alkaline phosphatase activity after the weight drop. These results reveal that both blast- and impact acceleration-induced head injuries cause an acute decrease in the level/activity of TNAP in the brain, which potentially contributes to trauma-induced accumulation of pTau and the resultant tauopathy. The regional changes in the level/activity of TNAP or accumulation of pTau after these injuries did not correlate with the accumulation of amyloid precursor protein, suggesting that the basic mechanism underlying tauopathy in TBI might be distinct from that associated with AD.


Assuntos
Fosfatase Alcalina/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/enzimologia , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Masculino , Fosforilação , Ratos Sprague-Dawley , Fatores de Tempo
2.
Brain Res ; 1549: 32-41, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-24412202

RESUMO

Repeated blast exposures commonly induce traumatic brain injury (TBI) characterized by diffuse axonal injury (DAI). We hypothesized that degradation of cytoskeletal proteins in the brain can lead to DAI, and evaluated α-II spectrin degradation in the pathophysiology of blast-induced TBI using the tightly-coupled three repetitive blast exposure mice model with a 1-30 min window in between exposures. Degradation of α-II spectrin and the expression profiles of caspase-3 and calpain-2, the major enzymes involved in the degradation were analyzed in the frontal cortex and cerebellum using Western blotting with specific antibodies. DAI at different brain regions was evaluated by neuropathology with silver staining. Repeated blast exposures resulted in significant increases in the α-II spectrin degradation products in the frontal cortex and cerebellum compared to sham controls. Expression of active caspase-3, which degrades α-II spectrin, showed significant increase in the frontal cortex after blast exposure at all the time points studied, while cerebellum showed an acute increase which was normalized over time. The expression of another α-II spectrin degrading enzyme, calpain-2, showed a rapid increase in the frontal cortex after blast exposure and it was significantly higher in the cerebellum at later time points. Neuropathological analysis showed significant levels of DAI at the frontal cortex and cerebellum at multiple time points after repeated blast injury. In summary, repeated blast exposure results in specific degradation of α-II spectrin in the brain along with differential expression of caspase-3/calpain-2 suggesting cytoskeletal breakdown as a possible contributor of DAI after repeated blast exposure.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Espectrina/metabolismo , Animais , Axônios/patologia , Traumatismos por Explosões/patologia , Encéfalo/patologia , Lesões Encefálicas/patologia , Calpaína/metabolismo , Caspase 3/metabolismo , Proteínas do Citoesqueleto/metabolismo , Camundongos
3.
J Neurotrauma ; 31(5): 498-504, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24074345

RESUMO

The pathophysiology of blast-induced traumatic brain injury (TBI) and subsequent behavioral deficits are not well understood. Unraveling the mechanisms of injury is critical to derive effective countermeasures against this form of neurotrauma. Preservation of the integrity of cellular DNA is crucial for the function and survival of cells. We evaluated the effect of repeated blast exposures on the integrity of brain DNA and tested the utility of cell-free DNA (CFD) in plasma as a biomarker for the diagnosis and prognosis of blast-induced polytrauma. The results revealed time-dependent breakdown in cellular DNA in different brain regions, with the maximum damage at 24 h post-blast exposures. CFD levels in plasma showed a significant transient increase, which was largely independent of the timing and severity of brain DNA damage; maximum levels were recorded at 2 h after repeated blast exposure and returned to baseline at 24 h. A positive correlation was observed between the righting reflex time and CFD level in plasma at 2 h after blast exposure. Brain DNA damage subsequent to repeated blast was associated with decreased mitochondrial membrane potential, increased release of cytochrome C, and up-regulation of caspase-3, all of which are indicative of cellular apoptosis. Shock-wave-induced DNA damage and initiation of mitochondrial-driven cellular apoptosis in the brain after repeated blast exposures indicate that therapeutic strategies directed toward inhibition of DNA damage or instigation of DNA repair may be effective countermeasures.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Fragmentação do DNA , Mitocôndrias/metabolismo , Animais , Apoptose/fisiologia , Biomarcadores/metabolismo , Traumatismos por Explosões/fisiopatologia , Encéfalo/fisiopatologia , Lesões Encefálicas/fisiopatologia , Caspase 3/metabolismo , Explosões , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regulação para Cima
4.
Neurosci Lett ; 552: 87-91, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-23933206

RESUMO

Glial fibrillary acidic protein (GFAP), a protein enriched in astrocytes, and Tau, a protein abundant in neuronal microtubules, are being widely studied as biomarkers of brain injury, and persistent severity-dependent increases in brain and blood have been reported. Studies on the acute changes of these proteins after blast exposure are limited. Using a mouse model of closely-coupled repeated blast exposures, we have evaluated acute changes in the levels of GFAP and total Tau by Western blotting. Brain levels of GFAP and Tau proteins decreased significantly at 6 h and increased considerably at 24 h after repeated blast exposures. Plasma samples showed a similar initial decrease and later increase over this timeframe. This biphasic pattern points to possible absorption or sequestration of these proteins from plasma immediately after repeated blast exposures. Liver and spleen tissue showed significant increases in the levels of GFAP and Tau protein at 6 and 24 h post-blast exposures whereas semi-quantitative RT-PCR analysis of liver showed no significant changes in the levels of GFAP or Tau mRNAs. These results suggest that blast exposure causes transient changes in cell membrane integrity in multiple organs leading to abnormal migration of proteins from the tissues to the plasma and vice versa. This transient changes in cell membrane permeability and subsequent bidirectional movement of molecules may contribute to the pathophysiology of TBI and polytrauma after blast exposure.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas/metabolismo , Permeabilidade da Membrana Celular , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas tau/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Traumatismos por Explosões/sangue , Traumatismos por Explosões/complicações , Lesões Encefálicas/sangue , Lesões Encefálicas/complicações , Proteína Glial Fibrilar Ácida/sangue , Fígado/metabolismo , Masculino , Camundongos , Baço/metabolismo , Proteínas tau/sangue
5.
J Neurotrauma ; 30(19): 1645-51, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23600763

RESUMO

Use of improvised explosive devices has significantly increased the incidence of traumatic brain injury (TBI) and associated neuropsychiatric deficits in the recent wars in Iraq and Afghanistan. Acute deleterious effects of single and repeated blast exposure can lead to long-term neurobiological effects and neuropsychiatric deficits. Using in vitro and in vivo shock tube models of blast-induced TBI, we studied changes in mitochondrial energy metabolism after blast exposure. Single and repeated blast exposures in vitro resulted in significant decreases in neuronal adenosine triphosphate (ATP) levels at 6 h post-blast that returned towards normal levels by 24 h. Similar changes in ATP also were observed in the cerebral cortices of mice subjected to single and repeated blast exposures. In neurons, mitochondrial glutamate oxaloacetate transaminase (GOT2) plays a critical role in metabolism and energy production. Proteomic analysis of brain cortices showed a significant decrease in GOT2 levels 6 h after repeated blast exposures, which was further confirmed by Western blotting. Western blot analysis of GOT2 and pyruvate dehydrogenase in the cortex showed direct correlation only between GOT2 and ATP levels. Activity of GOT2 in the isolated cortical mitochondria also showed significant decrease at 6 h supporting the results of proteomic and Western blot analyses. Knowing the significant role of GOT2 in the neuronal mitochondrial energy metabolism, it is quite likely that the down regulation of GOT2 after blast exposure is playing a significant role in mitochondrial dysfunction after blast exposure.


Assuntos
Aspartato Aminotransferases/metabolismo , Traumatismos por Explosões/enzimologia , Traumatismos por Explosões/patologia , Mitocôndrias/enzimologia , Mitocôndrias/fisiologia , Doenças Mitocondriais/patologia , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Linhagem Celular , Córtex Cerebral/enzimologia , Córtex Cerebral/lesões , Córtex Cerebral/metabolismo , Ciclo do Ácido Cítrico , Eletroforese em Gel de Poliacrilamida , Metabolismo Energético , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Proteômica , Complexo Piruvato Desidrogenase/metabolismo
6.
Neurosci Res ; 76(1-2): 98-100, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23511555

RESUMO

Blast-induced traumatic brain injury (TBI) and subsequent neurobehavioral deficits are major disabilities suffered by the military and civilian population worldwide. Rigorous scientific research is underway to understand the mechanism of blast TBI and thereby develop effective therapies for protection and treatment. By using an in vitro shock tube model of blast TBI with SH-SY5Y human neuroblastoma cells, we have demonstrated that blast exposure leads to neurobiological changes in an overpressure and time dependent manner. Paradoxically, repeated blast exposures resulted in less neuronal injury compared to single blast exposure and suggested a potential neuroprotective mechanism involving released cyclophilin A (CPA). In the present study, we demonstrate accumulation of CPA in the culture medium after repeated blast exposures supporting the notion of extracellular CPA mediated neuroprotection. Post-exposure treatment of the cells with purified recombinant CPA caused significant protection against blast-induced neuronal injury. Furthermore, repeated blast exposure was associated with phosphorylation of the proteins ERK1/2 and Bad suggesting a potential mechanism of neuroprotection by extracellular CPA and may aid in the development of targeted therapies for protection against blast-induced TBI.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas/metabolismo , Ciclofilina A/metabolismo , Neurônios/metabolismo , Traumatismos por Explosões/patologia , Western Blotting , Lesões Encefálicas/patologia , Linhagem Celular , Ciclofilina A/farmacologia , Humanos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia
7.
Neurosci Lett ; 539: 1-6, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23370286

RESUMO

Blast-induced traumatic brain injury is complex and involves multiple factors including systemic pathophysiological factors in addition to direct brain injuries. We hypothesize that systemic activation of platelets/leukocytes plays a major role in the development and exacerbation of brain injury after blast exposure. A mouse model of repeated blast exposure that results in significant neuropathology, neurobehavioral changes and regional specific alterations in various biomolecules in the brain was used for the proposed study. Activation of platelets was evaluated by flow cytometry and serotonin content was analyzed by ELISA. Expression of myeloperoxidase was analyzed by Western blotting. Histopathology of the brain was used to assess blast-induced cerebral vasoconstriction. The data showed an increase in the activation of platelets at 4h after repeated blast exposures, indicating changes in platelet phenotype in blast neurotrauma. Platelet serotonin concentration showed a significant decrease at 4h after blast with a concurrent increase in the plasma serotonin levels, confirming the early onset of platelet activation after repeated blast exposures. Blood, plasma and brain myeloperoxidase enzyme activity and expression was increased in repeated blast exposed mice at multiple time points. Histopathological analysis of the brains of blast exposed mice showed constriction of blood vessels compared to the respective controls, a phenomenon similar to the reported cerebral vasoconstriction in blast affected victims. These results suggest that repeated blast exposure leads to acute activation of platelets/leukocytes which can augment the pathological effects of brain injury. Platelet/leukocyte targeted therapies can be evaluated as potential acute treatment strategies to mitigate blast-induced neurotrauma.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Animais , Traumatismos por Explosões/fisiopatologia , Plaquetas/metabolismo , Encéfalo/irrigação sanguínea , Lesões Encefálicas/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Peroxidase/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Serotonina/metabolismo , Vasoconstrição
8.
Bioorg Med Chem Lett ; 23(5): 1544-7, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23395652

RESUMO

Chemical warfare nerve agents such as soman exert their toxic effects through an irreversible inhibition of acetylcholinesterase (AChE) and subsequently glutamatergic function, leading to uncontrolled seizures. The natural alkaloid (-)-huperzine A is a potent inhibitor of AChE and has been demonstrated to exert neuroprotection at an appropriate dose. It is hypothesized that analogs of both (+)- and (-)-huperzine A with an improved ability to interact with NMDA receptors together with reduced AChE inhibition will exhibit more effective neuroprotection against nerve agents. In this manuscript, the tested huperzine A analogs 2 and 3 were demonstrated to improve survival of guinea pigs exposed to soman at either 1.2 or 2×LD(50).


Assuntos
Alcaloides/química , Alcaloides/farmacologia , Inibidores da Colinesterase/química , Fármacos Neuroprotetores/química , Sesquiterpenos/química , Sesquiterpenos/farmacologia , Animais , Inibidores da Colinesterase/farmacologia , Cobaias , Fármacos Neuroprotetores/farmacologia , Soman/toxicidade , Análise de Sobrevida
9.
Chem Biol Interact ; 203(1): 120-4, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23123250

RESUMO

The neuropathologic mechanisms after exposure to lethal doses of nerve agent are complex and involve multiple biochemical pathways. Effective treatment requires drugs that can simultaneously protect by reversible binding to the acetylcholinesterase (AChE) and blocking cascades of seizure related brain damage, inflammation, neuronal degeneration as well as promoting induction of neuroregeneration. [-]-Huperzine A ([-]-Hup A), is a naturally occurring potent reversible AChE inhibitor that penetrates the blood-brain barrier. It also has several neuroprotective effects including modification of beta-amyloid peptide, reduction of oxidative stress, anti-inflammatory, anti-apoptotic and induction and regulation of nerve growth factor. Toxicities at higher doses restrict the neuroporotective ability of [-]-Hup A for treatment. The synthetic stereoisomer, [+]-Hup A, is less toxic due to poor AChE inhibition and is suitable for both pre-/post-exposure treatments of nerve agent toxicity. [+]-Hup A block the N-methyl-D-aspartate (NMDA)-induced seizure in rats, reduce excitatory amino acid induced neurotoxicity and also prevent soman induced toxicity with minimum performance decrement. Unique combinations of two stereo-isomers of Hup A may provide an excellent pre/post-treatment drug for the nerve agent induced seizure/status epilepticus. We investigated a combination of [+]-Hup A with a small dose of [-]-Hup A ([+] and [-]-Hup A) against soman toxicity. Our data showed that pretreatment with a combination [+] and [-]-Hup A significantly increased the survival rate and reduced behavioral abnormalities after exposure to 1.2 × LD(50) soman compared to [+]-Hup A in guinea pigs. In addition, [+] and [-]-Hup A pretreatment inhibited the development of high power of EEG better than [+]-Hup A pretreatment alone. These data suggest that a combination of [+] and [-]-Hup A offers better protection than [+]-Hup A and serves as a potent medical countermeasure against lethal dose nerve agent toxicity in guinea pigs.


Assuntos
Alcaloides/administração & dosagem , Inibidores da Colinesterase/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Sesquiterpenos/administração & dosagem , Soman/antagonistas & inibidores , Soman/toxicidade , Acetilcolinesterase/metabolismo , Alcaloides/química , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Substâncias para a Guerra Química/toxicidade , Inibidores da Colinesterase/química , Cobaias , Masculino , Fármacos Neuroprotetores/química , Ratos , Convulsões/induzido quimicamente , Convulsões/prevenção & controle , Sesquiterpenos/química , Estereoisomerismo
10.
Chem Biol Interact ; 203(1): 371-5, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23159883

RESUMO

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


Assuntos
Acetilcolina/metabolismo , Traumatismos por Explosões/metabolismo , Lesões Encefálicas/metabolismo , Mediadores da Inflamação/metabolismo , Acetilcolinesterase/metabolismo , Animais , Traumatismos por Explosões/genética , Encéfalo/metabolismo , Lesões Encefálicas/genética , Cerebelo/lesões , Cerebelo/metabolismo , Progressão da Doença , Proteínas Ligadas por GPI/metabolismo , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Transdução de Sinais , Distribuição Tecidual
11.
Front Neurol ; 3: 177, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23267342

RESUMO

Blast-induced traumatic brain injury (TBI) has been a major cause of morbidity and mortality in the conflicts in Iraq and Afghanistan. How the primary blast wave affects the brain is not well understood. In particular, it is unclear whether blast injures the brain through mechanisms similar to those found in non-blast closed impact injuries (nbTBI). The ß-amyloid (Aß) peptide associated with the development of Alzheimer's disease is elevated acutely following TBI in humans as well as in experimental animal models of nbTBI. We examined levels of brain Aß following experimental blast injury using enzyme-linked immunosorbent assays for Aß 40 and 42. In both rat and mouse models of blast injury, rather than being increased, endogenous rodent brain Aß levels were decreased acutely following injury. Levels of the amyloid precursor protein (APP) were increased following blast exposure although there was no evidence of axonal pathology based on APP immunohistochemical staining. Unlike the findings in nbTBI animal models, levels of the ß-secretase, ß-site APP cleaving enzyme 1, and the γ-secretase component presenilin-1 were unchanged following blast exposure. These studies have implications for understanding the nature of blast injury to the brain. They also suggest that strategies aimed at lowering Aß production may not be effective for treating acute blast injury to the brain.

12.
Inhal Toxicol ; 24(9): 539-49, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22860999

RESUMO

We evaluated the efficacy of aerosolized acetylcholinesterase (AChE) reactivator oxime MMB-4 in combination with the anticholinergic atropine sulfate for protection against respiratory toxicity and lung injury following microinstillation inhalation exposure to nerve agent soman (GD) in guinea pigs. Anesthetized animals were exposed to GD (841 mg/m(3), 1.2 LCt(50)) and treated with endotracheally aerosolized MMB-4 (50 µmol/kg) plus atropine sulfate (0.25 mg/kg) at 30 sec post-exposure. Treatment with MMB-4 plus atropine increased survival to 100% compared to 38% in animals exposed to GD. Decreases in the pulse rate and blood O(2) saturation following exposure to GD returned to normal levels in the treatment group. The body-weight loss and lung edema was significantly reduced in the treatment group. Similarly, bronchoalveolar cell death was significantly reduced in the treatment group while GD-induced increase in total cell count was decreased consistently but was not significant. GD-induced increase in bronchoalveolar protein was diminished after treatment with MMB-4 plus atropine. Bronchoalveolar lavage AChE and BChE activity were significantly increased in animals treated with MMB-4 plus atropine at 24 h. Lung and diaphragm tissue also showed a significant increase in AChE activity in the treatment group. Treatment with MMB-4 plus atropine sulfate normalized various respiratory dynamics parameters including respiratory frequency, tidal volume, peak inspiratory and expiratory flow, time of inspiration and expiration, enhanced pause and pause post-exposure to GD. Collectively, these results suggest that aerosolization of MMB-4 plus atropine increased survival, decreased respiratory toxicity and lung injury following GD inhalation exposure.


Assuntos
Atropina/administração & dosagem , Inibidores da Colinesterase/toxicidade , Reativadores da Colinesterase/administração & dosagem , Oximas/administração & dosagem , Substâncias Protetoras/administração & dosagem , Soman/toxicidade , Acetilcolinesterase/metabolismo , Administração por Inalação , Aerossóis , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Butirilcolinesterase/metabolismo , Substâncias para a Guerra Química/toxicidade , Combinação de Medicamentos , Cobaias , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Pulmão/fisiopatologia , Masculino
13.
PLoS One ; 7(4): e33798, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22493674

RESUMO

Explosive blast results in multiple organ injury and polytrauma, the intensity of which varies with the nature of the exposure, orientation, environment and individual resilience. Blast overpressure alone may not precisely indicate the level of body or brain injury after blast exposure. Assessment of the extent of body injury after blast exposure is important, since polytrauma and systemic factors significantly contribute to blast-induced traumatic brain injury. We evaluated the activity of plasma enzymes including aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase (LDH) and creatine kinase (CK) at different time points after blast exposure using a mouse model of single and repeated blast exposures to assess the severity of injury. Our data show that activities of all the enzymes in the plasma were significantly increased as early as 1 h after blast exposure. The elevated enzyme activity remained up to 6 h in an overpressure dose-dependent manner and returned close to normal levels at 24 h. Head-only blast exposure with body protection showed no increase in the enzyme activities suggesting that brain injury alone does not contribute to the systemic increase. In contrast to plasma increase, AST, ALT and LDH activity in the liver and CK in the skeletal muscle showed drastic decrease at 6 h after blast exposures. Histopathology showed mild necrosis at 6 h and severe necrosis at 24 h after blast exposures in liver and no changes in the skeletal muscle suggesting that the enzyme release from the tissue to plasma is probably triggered by transient cell membrane disruption from shockwave and not due to necrosis. Overpressure dependent transient release of tissue enzymes and elevation in the plasma after blast exposure suggest that elevated enzyme activities in the blood can be potentially used as a biological dosimeter to assess the severity of blast injury.


Assuntos
Biomarcadores/sangue , Traumatismos por Explosões/sangue , Lesões Encefálicas/sangue , Fígado/enzimologia , Músculo Esquelético/enzimologia , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Traumatismos por Explosões/patologia , Creatina Quinase/metabolismo , Explosões , Histocitoquímica , L-Lactato Desidrogenase/sangue , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Fatores de Tempo
14.
Neuroreport ; 23(6): 342-6, 2012 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-22426026

RESUMO

The biochemical mechanisms of explosive blast-induced traumatic brain injury and the subsequent long-term neurobehavioral abnormalities are still not completely understood. We studied the biochemical mechanism of blast traumatic brain injury using our recently reported in-vitro model system with a shock tube. Primary blast exposure of in-vitro models leads to neurobiological changes in an overpressure dose-dependent and time-dependent manner. Lactate dehydrogenase was released significantly into the extracellular medium without cell death after blast exposure, indicating compromised cell membrane integrity. We further explored the integrity of cell membrane after blast exposure by fluorescent dye uptake/release techniques in SH-SY5Y human neuroblastoma cells. Our data indicate that blast exposure leads to an overpressure-dependent transient increase in the release of preloaded calcein AM into the culture medium with proportional intracellular decrease. Uptake of an extracellular nucleic acid-binding dye TO-PRO-3 iodide was also increased significantly after blast exposure, indicating that the increased molecular transport is bidirectional and nuclear membrane integrity is also affected by blast exposure. These results suggest that blast exposure perturbs the integrity of the neuronal cell membrane, leading to increased bidirectional transport of molecules--a potential mechanism that can lead to traumatic brain injury.


Assuntos
Traumatismos por Explosões/metabolismo , Lesões Encefálicas/metabolismo , Permeabilidade da Membrana Celular/fisiologia , Neurônios/metabolismo , Carbocianinas , Linhagem Celular , Sobrevivência Celular , Fluoresceínas , Humanos , Modelos Biológicos
15.
Toxicol Lett ; 210(1): 87-94, 2012 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-22301377

RESUMO

Human paraoxonase 1 (PON1), a 45kDa arylesterase associated with circulating high density lipoproteins (HDL), has been described as an anti-atherogenic element in cardiovascular disorders. The efficacy of PON1 as a catalytic bioscavenger against OP and CWNA toxicity has been on debate for the last few decades. Hydrolysis of various organophosphates (OPs) and chemical warfare nerve agents (CWNAs) by PON1 has been demonstrated in both in vitro and in vivo experiments. Recently, we established the protective efficacy of human and rabbit serum purified PON1 as well as human recombinant PON1 expressed in Trichoplusia ni larvae against nerve agent toxicity in guinea pigs. Exogenous administration of purified PON1 was effective in protecting against 1.2 X LCt(50) of sarin and soman administered endotracheally with microinstillation technology. However, the short half-life of exogenously administered PON1, probably due to poor association with circulating HDL, warrant alternative approaches for successful utility of PON1 in the treatment of OP/CWNA toxicity. In this mini review, we address the pros and cons of current PON1 prophylaxis and propose potential solutions for successful development of PON1 as an effective catalytic bioscavenger.


Assuntos
Arildialquilfosfatase/uso terapêutico , Substâncias para a Guerra Química/metabolismo , Organofosfatos/antagonistas & inibidores , Animais , Arildialquilfosfatase/farmacocinética , Cobaias , Meia-Vida , Humanos , Organofosfatos/metabolismo , Coelhos
16.
Neurotoxicology ; 33(2): 169-77, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22245390

RESUMO

The chemical warfare nerve agent, soman irreversibly inhibits acetylcholinesterase (AChE) leading to hypercholinergy and seizures which trigger glutamate toxicity and status epilepticus ultimately resulting in neuropathology and neurobehavioral deficits. The standard emergency treatment comprising of anticholinergic, AChE reactivator and anticonvulsant does not completely protect against soman toxicity. We have evaluated imidazenil, a new anticonvulsant imidazo benzodiazepine with high affinity and intrinsic efficacy at α5-, α2-, and α3- but low intrinsic efficacy at α1-containing GABA(A) receptors and is devoid of cardiorespiratory depression, sedative/hypnoitc and amnestic actions and does not elicit tolerance and dependence liabilities unlike diazepam, for protection against soman toxicity. Guinea pigs implanted with bipotential radiotelemetry probes for recording EEG and ECG were administered with 26 µg/kg pyridostigmine bromide 30 min prior to 2× LD(50) soman exposure and 1 min later treated with a combination of 2mg/kg atropine sulfate and 25mg/kg 2-pralidoxime and various doses of imidazenil. Intramuscular administration of imidazenil, dose-dependently protected against 2× LD(50) of soman toxicity up to 1mg/kg. Further increase in the dose of imidazenil to 2.5mg/kg was less effective than 1mg/kg probably due to non-specific actions at sites other than GABA(A) receptors. Compared to vehicle group, 1mg/kg imidazenil treatment showed optimal increase in survival rate, reduction in behavioral manifestations and high power of EEG spectrum as well as neuronal necrosis. These data suggest that imidazenil is an effective anticonvulsant for medical countermeasure against soman-induced toxicity.


Assuntos
Benzodiazepinas/uso terapêutico , Substâncias para a Guerra Química/toxicidade , Imidazóis/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Soman/toxicidade , Acetilcolinesterase/sangue , Acetilcolinesterase/metabolismo , Análise de Variância , Animais , Atropina/uso terapêutico , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Reativadores da Colinesterase/uso terapêutico , Modelos Animais de Doenças , Esquema de Medicação , Eletrocardiografia/métodos , Eletroencefalografia/métodos , Cobaias , Dose Letal Mediana , Masculino , Antagonistas Muscarínicos/uso terapêutico , Compostos de Pralidoxima/uso terapêutico , Convulsões/induzido quimicamente , Convulsões/prevenção & controle , Telemetria , Fatores de Tempo
17.
Toxicol In Vitro ; 26(1): 182-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22120822

RESUMO

Human prolidase (PROL), which has structural homology to bacterial organophosphate acid anhydrolase that hydrolyze organophosphates and nerve agents has been proposed recently as a potential catalytic bioscavenger. To develop PROL as a catalytic bioscavenger, we evaluated the in vitro hydrolysis efficiency of purified recombinant human PROL against organophosphates and nerve agents. Human liver PROL was purified by chromatographic procedures, whereas recombinant human skin and kidney PROL was expressed in Trichoplusia ni larvae, affinity purified and analyzed by gel electrophoresis. The catalytic efficiency of PROL against diisopropylfluorophosphate (DFP) and nerve agents was evaluated by acetylcholinesterase back-titration assay. Partially purified human liver PROL hydrolyzed DFP and various nerve agents, which was abolished by specific PROL inhibitor showing the specificity of hydrolysis. Both the recombinant human skin and kidney PROL expressed in T. ni larvae showed ∼99% purity and efficiently hydrolyzed DFP and sarin. In contrast to human liver PROL, both skin and kidney PROL showed significantly low hydrolyzing potential against nerve agents soman, tabun and VX. In conclusion, compared to human liver PROL, recombinant human skin and kidney PROL hydrolyze only DFP and sarin showing the substrate specificity of PROL from various tissue sources.


Assuntos
Substâncias para a Guerra Química/química , Inibidores da Colinesterase/química , Dipeptidases/química , Proteínas Recombinantes/química , Acetilcolinesterase/química , Humanos , Hidrólise , Isoflurofato/química , Rim/enzimologia , Fígado/enzimologia , Organofosfatos/química , Compostos Organotiofosforados/química , Sarina/química , Pele/enzimologia
18.
Toxicol Mech Methods ; 22(4): 250-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22145985

RESUMO

Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activities were measured in the blood and tissues of animals that are treated with a number of endotracheally aerosolized therapeutics for protection against inhalation toxicity to sarin. Therapeutics included, aerosolized atropine methyl bromide (AMB), scopolamine or combination of AMB with salbutamol, sphingosine 1-phosphate, keratinocyte growth factor, adenosine A1 receptor antisense oligonucleotide (EPI2010), 2,3-diacetyloxybenzoic acid (2,3 DABA), oxycyte, and survanta. Guinea pigs exposed to 677.4 mg/m(3) or 846.5 mg/m(3) (1.2 LCt(50)) sarin for 4 min using a microinstillation inhalation exposure technique and treated 1 min later with the aerosolized therapeutics. Treatment with all therapeutics significantly increased the survival rate with no convulsions throughout the 24 h study period. Blood AChE activity determined using acetylthiocholine as substrate showed 20% activity remaining in sarin-exposed animals compare to controls. In aerosolized AMB and scopolamine-treated animals the remaining AChE activity was significantly higher (45-60%) compared to sarin-exposed animals (p < 0.05). Similarly, treatment with all the combination therapeutics resulted in significant increase in blood AChE activity in comparison to sarin-exposed animals although the increases varied between treatments (p < 0.05). BChE activity was increased after treatment with aerosolized therapeutics but was lesser in magnitude compared to AChE activity changes. Various tissues showed elevated AChE activity after therapeutic treatment of sarin-exposed animals. Increased AChE and BChE activities in animals treated with nasal therapeutics suggest that enhanced breathing and reduced respiratory toxicity/lung injury possibly contribute to rapid normalization of chemical warfare nerve agent inhibited cholinesterases.


Assuntos
Acetilcolinesterase/metabolismo , Broncodilatadores/uso terapêutico , Inibidores da Colinesterase/toxicidade , Antagonistas Muscarínicos/uso terapêutico , Sarina/toxicidade , Acetilcolinesterase/sangue , Animais , Antídotos/uso terapêutico , Butirilcolinesterase/sangue , Butirilcolinesterase/metabolismo , Substâncias para a Guerra Química/toxicidade , Cobaias , Pulmão/efeitos dos fármacos , Pneumopatias/induzido quimicamente , Pneumopatias/enzimologia , Masculino , Terapia Respiratória
19.
Neurosci Lett ; 506(1): 141-5, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22079491

RESUMO

Acetylcholinesterase (AChE) which catalyzes the hydrolysis of the neurotransmitter acetylcholine has been recognized as one of the major regulators of stress responses after traumatic brain injury (TBI). Repeated blast exposure induces TBI (blast TBI) with a variable neuropathology at different brain regions. Since AChE inhibitors are being used as a line of treatment for TBI, we sought to determine the time course of AChE activity in the blood and different brain regions after repeated blast exposures using modified Ellman assay. Our data showed that repeated blast exposures significantly reduced AChE activity in the whole-blood and erythrocytes by 3-6h, while plasma AChE activity was significantly increased by 3h post-blast. In the brain, significant increase in AChE activity was observed at 6h in the frontal cortex, while hind cortex and hippocampus showed a significant decrease at 6h post-blast, which returned to normal levels by 7 days. AChE activity in the cerebellum and mid brain showed a decrease at 6h, followed by significant increase at 3 days and that was decreased significantly at 14 days post-blast. Medulla region showed decreased AChE activity at 24h post-blast, which was significantly increased at 14 days. These results suggest that there are brain regional and time-related changes in AChE activity after tightly coupled repeated blast exposures in mice. In summary, acute and chronic regional specific changes in the AChE activity after repeated blast exposures warrant systematic evaluation of the possibility of AChE inhibitor therapeutics against blast TBI.


Assuntos
Acetilcolinesterase/metabolismo , Lesões Encefálicas/patologia , Encéfalo/enzimologia , Acetilcolina/sangue , Acetilcolinesterase/sangue , Animais , Lesões Encefálicas/sangue , Lesões Encefálicas/enzimologia , Modelos Animais de Doenças , Eritrócitos/enzimologia , Eritrócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estatísticas não Paramétricas , Fatores de Tempo
20.
J Rehabil Res Dev ; 49(7): 1153-62, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23341286

RESUMO

The mechanisms of central auditory processing involved in auditory/vestibular injuries and subsequent tinnitus and hearing loss in Active Duty servicemembers exposed to blast are not currently known. We analyzed the expression of hearing-related genes in different regions of the brain 6 h after repeated blast exposures in mice. Preliminary data showed that the expression of the deafness-related genes otoferlin and otoancorin was significantly changed in the hippocampus after blast exposures. Differential expression of cadherin and protocadherin genes, which are involved in hearing impairment, was observed in the hippocampus, cerebellum, frontal cortex, and midbrain after repeated blasts. A series of calcium-signaling genes that are known to be involved in auditory signal processing were also found to be significantly altered after repeated blast exposures. The hippocampus and midbrain showed significant increase in the gene expression of hearing loss-related antioxidant enzymes. Histopathology of the auditory cortex showed more significant injury in the inner layer compared to the outer layer. In summary, mice exposed to repeated blasts showed injury to the auditory cortex and significant alterations in multiple genes in the brain known to be involved in age- or noise-induced hearing impairment.


Assuntos
Córtex Auditivo/fisiopatologia , Doenças Auditivas Centrais/genética , Traumatismos por Explosões/fisiopatologia , Proteínas Ligadas por GPI/metabolismo , Perda Auditiva Provocada por Ruído/genética , Hipocampo/metabolismo , Proteínas de Membrana/metabolismo , Animais , Doenças Auditivas Centrais/metabolismo , Lesões Encefálicas/fisiopatologia , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Explosões , Proteínas Ligadas por GPI/genética , Perda Auditiva Provocada por Ruído/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...