Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Thromb J ; 15: 22, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28860945

RESUMEN

BACKGROUND: It is well accepted that functional activity of platelet integrin αIIbß3 is crucial for hemostasis and thrombosis. The ß3 subunit of the complex undergoes tyrosine phosphorylation shown to be critical for outside-in integrin signaling and platelet clot retraction ex vivo. However, the role of this important signaling event in other aspects of prothrombotic platelet function is unknown. METHOD: Here, we assess the role of ß3 tyrosine phosphorylation in platelet function regulation with a knock-in mouse strain, where two ß3 cytoplasmic tyrosines are mutated to phenylalanine (DiYF). We employed platelet transfusion technique and intravital microscopy for observing the cellular events involved in specific steps of thrombus growth to investigate in detail the role of ß3 tyrosine phosphorylation in arterial thrombosis in vivo. RESULTS: Upon injury, DiYF mice exhibited delayed arterial occlusion and unstable thrombus formation. The mean thrombus volume in DiYF mice formed on collagen was only 50% of that in WT. This effect was attributed to DiYF platelets but not to other blood cells and endothelium, which also carry these mutations. Transfusion of isolated DiYF but not WT platelets into irradiated WT mice resulted in reversal of the thrombotic phenotype and significantly prolonged blood vessel occlusion times. DiYF platelets exhibited reduced adhesion to collagen under in vitro shear conditions compared to WT platelets. Decreased platelet microparticle release after activation, both in vitro and in vivo, were observed in DiYF mice compared to WT mice. CONCLUSION: ß3 tyrosine phosphorylation of platelet αIIbß3 regulates both platelet pro-thrombotic activity and the formation of a stable platelet thrombus, as well as arterial microparticle release.

2.
Nat Med ; 13(9): 1086-95, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17721545

RESUMEN

Dyslipidemia is associated with a prothrombotic phenotype; however, the mechanisms responsible for enhanced platelet reactivity remain unclear. Proatherosclerotic lipid abnormalities are associated with both enhanced oxidant stress and the generation of biologically active oxidized lipids, including potential ligands for the scavenger receptor CD36, a major platelet glycoprotein. Using multiple mouse in vivo thrombosis models, we now demonstrate that genetic deletion of Cd36 protects mice from hyperlipidemia-associated enhanced platelet reactivity and the accompanying prothrombotic phenotype. Structurally defined oxidized choline glycerophospholipids that serve as high-affinity ligands for CD36 were at markedly increased levels in the plasma of hyperlipidemic mice and in the plasma of humans with low HDL levels, were able to bind platelets via CD36 and, at pathophysiological levels, promoted platelet activation via CD36. Thus, interactions of platelet CD36 with specific endogenous oxidized lipids play a crucial role in the well-known clinical associations between dyslipidemia, oxidant stress and a prothrombotic phenotype.


Asunto(s)
Plaquetas/fisiología , Antígenos CD36/fisiología , Eliminación de Gen , Hiperlipidemias/sangre , Estrés Oxidativo , Protrombina/genética , Trombosis/inmunología , Animales , Antígenos CD/fisiología , Antígenos CD36/genética , Dislipidemias/sangre , Dislipidemias/genética , Humanos , Hiperlipidemias/inmunología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Fosfolípidos/sangre , Activación Plaquetaria , Trombosis/sangre
3.
Proc Natl Acad Sci U S A ; 107(38): 16643-8, 2010 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-20823248

RESUMEN

Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) family members mediate the adherence of parasite-infected red blood cells (IRBCs) to various host receptors. A previous study has shown that the parasite protein, cytoadherence-linked asexual gene 9 (CLAG9), is also essential for IRBC adherence. However, how CLAG9 influences this process remains unknown. In this study, we show that CLAG9 interacts with VAR2CSA, a PfEMP1 that mediates IRBC adherence to chondroitin 4-sulfate in the placenta. Importantly, our results show that the adherent parasites synthesize CLAG9 at two stages--the early ring and late trophozoite stages. Localization studies revealed that a substantial level of CLAG9 is located mainly at or in close proximity of the IRBC membrane in association with VAR2CSA. Upon treatment of IRBCs with trypsin, a significant amount of CLAG9 (≈150 kDa) was converted into ≈142-kDa polypeptide. Together these data demonstrate that a considerable amount of CLAG9 is embedded in the IRBC membrane such that at least a portion of the polypeptide at either N or C terminus is exposed on the cell surface. In parasites lacking CLAG9, VAR2CSA failed to express on the IRBC surface and was located within the parasite. Based on these findings, we propose that CLAG9 plays a critical role in the trafficking of PfEMP1s onto the IRBC surface. These results have important implications for the development of therapeutics for cerebral, placental, and other cytoadherence-associated malaria illnesses.


Asunto(s)
Antígenos de Protozoos/fisiología , Moléculas de Adhesión Celular/fisiología , Plasmodium falciparum/fisiología , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/fisiología , Animales , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Secuencia de Bases , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/genética , Sulfatos de Condroitina/fisiología , ADN Protozoario/genética , Membrana Eritrocítica/parasitología , Membrana Eritrocítica/fisiología , Membrana Eritrocítica/ultraestructura , Eritrocitos/parasitología , Femenino , Técnicas de Inactivación de Genes , Genes Protozoarios , Interacciones Huésped-Parásitos/fisiología , Humanos , Técnicas In Vitro , Microscopía Inmunoelectrónica , Complejos Multiproteicos , Placenta/parasitología , Placenta/fisiología , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Embarazo , Proteínas Protozoarias/biosíntesis , Proteínas Protozoarias/química , Proteínas Protozoarias/genética
4.
Biochem Biophys Res Commun ; 403(1): 97-102, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21040699

RESUMEN

Paraoxonase 1 (PON1) has been described as an efficient catalytic bioscavenger due to its ability to hydrolyze organophosphates (OPs) and chemical warfare nerve agents (CWNAs). It is the future most promising candidate as prophylactic medical countermeasure against highly toxic OPs and CWNAs. Most of the studies conducted so far have been focused on the hydrolyzing potential of PON1 against nerve agents, sarin, soman, and VX. Here, we investigated the hydrolysis of tabun by PON1 with the objective of comparing the hydrolysis potential of human and rabbit serum purified and recombinant human PON1. The hydrolysis potential of PON1 against tabun, sarin, and soman was evaluated by using an acetylcholinesterase (AChE) back-titration Ellman method. Efficient hydrolysis of tabun (100 nM) was observed with ∼25-40 mU of PON1, while higher concentration (80-250 mU) of the enzyme was required for the complete hydrolysis of sarin (11 nM) and soman (3 nM). Our data indicate that tabun hydrolysis with PON1 was ∼30-60 times and ∼200-260 times more efficient than that with sarin and soman, respectively. Moreover, the catalytic activity of PON1 varies from source to source, which also reflects their efficiency of hydrolyzing different types of nerve agents. Thus, efficient hydrolysis of tabun by PON1 suggests its promising potential as a prophylactic treatment against tabun exposure.


Asunto(s)
Arildialquilfosfatasa/metabolismo , Sustancias para la Guerra Química/metabolismo , Inhibidores de la Colinesterasa/metabolismo , Sistema Nervioso/efectos de los fármacos , Organofosfatos/metabolismo , Animales , Humanos , Hidrólisis , Conejos , Proteínas Recombinantes/metabolismo
5.
Mol Biochem Parasitol ; 134(1): 115-26, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14747149

RESUMEN

Infection with Plasmodium falciparum during pregnancy leads to chondroitin 4-sulfate-mediated adhesion of the infected red blood cells (IRBCs) in the placenta, causing severe health complications to fetus and the mother. The IRBCs are also frequently found in low density in the umbilical cord of infected placentas. In this study, the CSPGs of umbilical vein and arteries were purified, characterized, and their localization and IRBC-binding abilities were studied. While a versican type CSPG was found both in the vein and arteries, a serglycin type CSPG was present exclusively in the vein. The CSPGs were present at significant level on the endothelial surface of the umbilical vein but not on that of arteries. Although the purified versican and serglycin type CSPGs could bind IRBCs, their binding abilities were significantly less compared to the low sulfated CSPGs of the placenta because of the predominance of 6-sulfated disaccharide moieties in the CS chains. Therefore, IRBCs were unable to bind efficiently onto the umbilical cord endothelial surface. Unexpectedly, however, the IRBCs adhered densely in the blood vessels of fetal villi in the placental tissue sections and sparingly in the blood spaces of the umbilical cord vein, presumably because the CSPG that can efficiently bind IRBCs is present at high levels in the fetal blood vessels and at very low levels in the umbilical cord blood vessels. Since the C4S-adherent IRBCs that enter the fetal blood vessels cannot adhere to the cord endothelial surface and parasites cannot efficiently grow due to fetal hemoglobin toxicity and protection by maternal antibodies, transplacental infection may be quickly cleared without clinical episodes.


Asunto(s)
Adhesión Celular , Proteoglicanos Tipo Condroitín Sulfato/química , Proteoglicanos Tipo Condroitín Sulfato/aislamiento & purificación , Células Endoteliales/química , Eritrocitos/parasitología , Plasmodium falciparum/fisiología , Animales , Femenino , Glicosaminoglicanos/química , Humanos , Lectinas Tipo C , Placenta/parasitología , Embarazo , Proteoglicanos/química , Proteoglicanos/aislamiento & purificación , Arterias Umbilicales/química , Arterias Umbilicales/parasitología , Venas Umbilicales/química , Venas Umbilicales/parasitología , Versicanos , Proteínas de Transporte Vesicular
6.
J Neurotrauma ; 31(5): 498-504, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24074345

RESUMEN

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.


Asunto(s)
Traumatismos por Explosión/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Fragmentación del ADN , Mitocondrias/metabolismo , Animales , Apoptosis/fisiología , Biomarcadores/metabolismo , Traumatismos por Explosión/fisiopatología , Encéfalo/fisiopatología , Lesiones Encefálicas/fisiopatología , Caspasa 3/metabolismo , Explosiones , Masculino , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba
7.
Neurosci Res ; 76(1-2): 98-100, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23511555

RESUMEN

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.


Asunto(s)
Traumatismos por Explosión/metabolismo , Lesiones Encefálicas/metabolismo , Ciclofilina A/metabolismo , Neuronas/metabolismo , Traumatismos por Explosión/patología , Western Blotting , Lesiones Encefálicas/patología , Línea Celular , Ciclofilina A/farmacología , Humanos , Neuronas/patología , Fármacos Neuroprotectores/farmacología
8.
Neurosci Lett ; 539: 1-6, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23370286

RESUMEN

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.


Asunto(s)
Traumatismos por Explosión/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Animales , Traumatismos por Explosión/fisiopatología , Plaquetas/metabolismo , Encéfalo/irrigación sanguínea , Lesiones Encefálicas/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Peroxidasa/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Serotonina/metabolismo , Vasoconstricción
9.
J Neurotrauma ; 30(19): 1645-51, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23600763

RESUMEN

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.


Asunto(s)
Aspartato Aminotransferasas/metabolismo , Traumatismos por Explosión/enzimología , Traumatismos por Explosión/patología , Mitocondrias/enzimología , Mitocondrias/fisiología , Enfermedades Mitocondriales/patología , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Línea Celular , Corteza Cerebral/enzimología , Corteza Cerebral/lesiones , Corteza Cerebral/metabolismo , Ciclo del Ácido Cítrico , Electroforesis en Gel de Poliacrilamida , Metabolismo Energético , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proteómica , Complejo Piruvato Deshidrogenasa/metabolismo
10.
Neurosci Lett ; 552: 87-91, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-23933206

RESUMEN

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.


Asunto(s)
Traumatismos por Explosión/metabolismo , Lesiones Encefálicas/metabolismo , Permeabilidad de la Membrana Celular , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas tau/metabolismo , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Traumatismos por Explosión/sangre , Traumatismos por Explosión/complicaciones , Lesiones Encefálicas/sangre , Lesiones Encefálicas/complicaciones , Proteína Ácida Fibrilar de la Glía/sangre , Hígado/metabolismo , Masculino , Ratones , Bazo/metabolismo , Proteínas tau/sangre
11.
Chem Biol Interact ; 203(1): 371-5, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23159883

RESUMEN

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.


Asunto(s)
Acetilcolina/metabolismo , Traumatismos por Explosión/metabolismo , Lesiones Encefálicas/metabolismo , Mediadores de Inflamación/metabolismo , Acetilcolinesterasa/metabolismo , Animales , Traumatismos por Explosión/genética , Encéfalo/metabolismo , Lesiones Encefálicas/genética , Cerebelo/lesiones , Cerebelo/metabolismo , Progresión de la Enfermedad , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Transducción de Señal , Distribución Tisular
12.
Toxicol Lett ; 210(1): 87-94, 2012 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-22301377

RESUMEN

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.


Asunto(s)
Arildialquilfosfatasa/uso terapéutico , Sustancias para la Guerra Química/metabolismo , Organofosfatos/antagonistas & inhibidores , Animales , Arildialquilfosfatasa/farmacocinética , Cobayas , Semivida , Humanos , Organofosfatos/metabolismo , Conejos
13.
Neuroreport ; 23(6): 342-6, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22426026

RESUMEN

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.


Asunto(s)
Traumatismos por Explosión/metabolismo , Lesiones Encefálicas/metabolismo , Permeabilidad de la Membrana Celular/fisiología , Neuronas/metabolismo , Carbocianinas , Línea Celular , Supervivencia Celular , Fluoresceínas , Humanos , Modelos Biológicos
14.
J Rehabil Res Dev ; 49(7): 1153-62, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23341286

RESUMEN

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.


Asunto(s)
Corteza Auditiva/fisiopatología , Enfermedades Auditivas Centrales/genética , Traumatismos por Explosión/fisiopatología , Proteínas Ligadas a GPI/metabolismo , Pérdida Auditiva Provocada por Ruido/genética , Hipocampo/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Enfermedades Auditivas Centrales/metabolismo , Lesiones Encefálicas/fisiopatología , Cadherinas/genética , Cadherinas/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Explosiones , Proteínas Ligadas a GPI/genética , Pérdida Auditiva Provocada por Ruido/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
15.
Neurosci Lett ; 506(1): 141-5, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22079491

RESUMEN

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.


Asunto(s)
Acetilcolinesterasa/metabolismo , Lesiones Encefálicas/patología , Encéfalo/enzimología , Acetilcolina/sangre , Acetilcolinesterasa/sangre , Animales , Lesiones Encefálicas/sangre , Lesiones Encefálicas/enzimología , Modelos Animales de Enfermedad , Eritrocitos/enzimología , Eritrocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estadísticas no Paramétricas , Factores de Tiempo
16.
Toxicol In Vitro ; 26(1): 182-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22120822

RESUMEN

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.


Asunto(s)
Sustancias para la Guerra Química/química , Inhibidores de la Colinesterasa/química , Dipeptidasas/química , Proteínas Recombinantes/química , Acetilcolinesterasa/química , Humanos , Hidrólisis , Isoflurofato/química , Riñón/enzimología , Hígado/enzimología , Organofosfatos/química , Compuestos Organotiofosforados/química , Sarín/química , Piel/enzimología
17.
PLoS One ; 7(4): e33798, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22493674

RESUMEN

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.


Asunto(s)
Biomarcadores/sangre , Traumatismos por Explosión/sangre , Lesiones Encefálicas/sangre , Hígado/enzimología , Músculo Esquelético/enzimología , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Traumatismos por Explosión/patología , Creatina Quinasa/metabolismo , Explosiones , Histocitoquímica , L-Lactato Deshidrogenasa/sangre , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Factores de Tiempo
18.
Toxicol In Vitro ; 25(4): 905-13, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21382471

RESUMEN

Paraoxonase 1 (PON1) has been described as a potential catalytic bioscavenger due to its ability to hydrolyze organophosphate (OP) insecticides and nerve agents. In vitro catalytic efficiency of purified human and rabbit serum PON1 against different OP substrates was compared to human recombinant PON1, expressed in Trichoplusia ni larvae. Highly purified human and rabbit serum PON1s were prepared by multiple chromatography methods. Purified enzymes showed higher catalytic activity with the substrate p-nitrophenyl acetate compared to diethyl paraoxon. The hydrolyzing potential of PON1s against multiple OPs was evaluated by using an in vitro acetylcholinesterase back-titration assay. Significant differences in the catalytic efficiency of all the three PON1s with regard to various OP substrates were observed. Purified PON1s showed higher catalytic activity towards diisopropylfluorophosphate followed by diethylparaoxon compared to dimethyl paraoxon. Heat inactivation or incubation of PON1 with specific inhibitor resulted in complete loss of the enzyme catalytic activity indicating that OP hydrolysis was intrinsic to PON1. In conclusion, purified PON1s from multiple sources show significant differences in the catalytic activity against several OP substrates. These results underscore the importance of systematic analysis of candidate PON1 molecules for developing as an effective catalytic bioscavenger against toxic OPs and chemical warfare nerve agents.


Asunto(s)
Arildialquilfosfatasa/farmacología , Sustancias para la Guerra Química/metabolismo , Insecticidas/metabolismo , Compuestos Organofosforados/metabolismo , Acetilcolinesterasa/metabolismo , Animales , Catálisis , Calor , Humanos , Hidrólisis , Larva , Mariposas Nocturnas , Conejos
19.
Toxicol Lett ; 202(3): 203-8, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21329748

RESUMEN

To explore the efficacy of paraoxonase 1 (PON1) as a catalytic bioscavenger, we evaluated human recombinant PON1 (rePON1) expressed in Trichoplusia ni larvae against sarin and soman toxicity using microinstillation inhalation exposure in guinea pigs. Animals were pretreated intravenously with catalytically active rePON1, followed by exposure to 1.2 X LCt50 sarin or soman. Administration of 5 units of rePON1 showed mild increase in the blood activity of the enzyme after 30 min, but protected the animals with a significant increase in survival rate along with minimal signs of nerve agent toxicity. Recombinant PON1 pretreated animals exposed to sarin or soman prevented the reduction of blood O2 saturation and pulse rate observed after nerve agent exposure. In addition, rePON1 pretreated animals showed significantly higher blood PON1, acetylcholinesterase (AChE), and butyrylcholinesterase activity after nerve agent exposure compared to the respective controls without treatments. AChE activity in different brain regions of rePON1 pretreated animals exposed to sarin or soman were also significantly higher than respective controls. The remaining activity of blood PON1, cholinesterases and brain AChE in PON1 pretreated animals after nerve agent exposure correlated with the survival rate. In summary, these data suggest that human rePON1 protects against sarin and soman exposure in guinea pigs.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Arildialquilfosfatasa/farmacología , Sustancias para la Guerra Química/toxicidad , Fármacos Neuroprotectores/farmacología , Sarín/toxicidad , Soman/toxicidad , Lesión Pulmonar Aguda/sangre , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Colinesterasas/sangre , Cobayas , Humanos , Exposición por Inhalación , Inyecciones Intravenosas , Instilación de Medicamentos , Estimación de Kaplan-Meier , Longevidad/efectos de los fármacos , Masculino , Oxígeno/sangre , Conejos , Proteínas Recombinantes/farmacología
20.
J Neurotrauma ; 28(10): 2171-83, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21770761

RESUMEN

A mouse model of repeated blast exposure was developed using a compressed air-driven shock tube, to study the increase in severity of traumatic brain injury (bTBI) after multiple blast exposures. Isoflurane anesthetized C57BL/6J mice were exposed to 13.9, 20.6, and 25 psi single blast overpressure (BOP1) and allowed to recover for 5 days. BOP1 at 20.6 psi showed a mortality rate of 2% and this pressure was used for three repeated blast exposures (BOP3) with 1 and 30 min intervals. Overall mortality rate in BOP3 was increased to 20%. After blast exposure, righting reflex time and body-weight loss were significantly higher in BOP3 animals compared to BOP1 animals. At 4 h, brain edema was significantly increased in BOP3 animals compared to sham controls. Reactive oxygen species in the cortex were increased significantly in BOP1 and BOP3 animals. Neuropathological analysis of the cerebellum and cerebral cortex showed dense silver precipitates in BOP3 animals, indicating the presence of diffuse axonal injury. Fluoro-Jade B staining showed increased intensity in the cortex of BOP3 animals indicating neurodegeneration. Rota Rod behavioral test showed a significant decrease in performance at 10 rpm following BOP1 or BOP3 at 2 h post-blast, which gradually recovered during the 5 days. At 20 rpm, the latency to fall was significantly decreased in both BOP1 and BOP3 animals and it did not recover in the majority of the animals through 5 days of testing. These data suggest that repeated blast exposures lead to increased impairment severity in multiple neurological parameters of TBI in mice.


Asunto(s)
Traumatismos por Explosión/patología , Lesiones Encefálicas/patología , Animales , Traumatismos por Explosión/mortalidad , Traumatismos por Explosión/fisiopatología , Encéfalo/patología , Química Encefálica/fisiología , Edema Encefálico/etiología , Edema Encefálico/patología , Lesiones Encefálicas/mortalidad , Lesiones Encefálicas/fisiopatología , Corteza Cerebral/patología , Colorantes , Masculino , Ratones , Ratones Endogámicos C57BL , Equilibrio Postural/fisiología , Presión , Especies Reactivas de Oxígeno/metabolismo , Recurrencia , Sobrevida , Pérdida de Peso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA