Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Opt Lett ; 32(11): 1426-8, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17546143

RESUMEN

A four-stage laser system was developed, emitting at a wavelength of 6450 nm with a 3-5 ns pulse duration, < or = 2 mJ pulse energy, and 1/2 Hz pulse repetition rate. The laser system successfully ablated rat brain tissue, where both the collateral damage and the ablation rate compare favorably with that previously observed with a Mark-III Free-Electron Laser.


Asunto(s)
Rayos Láser , Óptica y Fotónica , Espectrometría Raman/métodos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Electrones , Diseño de Equipo , Oscilometría , Ratas , Ratas Sprague-Dawley , Dispersión de Radiación , Factores de Tiempo
2.
Neuroscience ; 116(2): 437-45, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12559098

RESUMEN

Apolipoprotein E (apoE) is a 34-kD protein with multiple biological properties. Recent clinical and preclinical observations implicate a role for apoE in modifying the response of the brain to focal and global ischemia. One mechanism by which apoE might exert these effects is by reducing glutamate-induced excitotoxic neuronal injury associated with ischemic insults. We demonstrate that human recombinant apoE confers a mild neuroprotective effect in primary neuronal-glial cultures exposed to 100 microM N-methyl-D-aspartate. Furthermore, a peptide derived from the receptor-binding region of apoE (residues 133-149) maintained a significant helical population as assessed by circular dichroism, and completely suppressed the neuronal cell death and calcium influx associated with N-methyl-D-aspartate exposure. Neuroprotection was greatest when the peptide was added concurrently with N-methyl-D-aspartate; however, a significant protection was observed when peptide was preincubated and washed off prior to N-methyl-D-aspartate exposure. These results suggest that one mechanism by which apoE may modify the CNS response to ischemia is by partially blocking glutamate excitotoxicity. Moreover, small peptide fragments derived from the receptor-binding region of apoE have enhanced bioactivity compared with the intact holoprotein, and may represent a novel therapeutic strategy for the treatment of brain ischemia.


Asunto(s)
Apolipoproteínas E/farmacología , Agonistas de Aminoácidos Excitadores/toxicidad , N-Metilaspartato/toxicidad , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Secuencia de Aminoácidos , Animales , Apolipoproteínas E/química , Apolipoproteínas E/genética , Isquemia Encefálica/tratamiento farmacológico , Células Cultivadas , Técnicas de Cocultivo , Imitación Molecular , Datos de Secuencia Molecular , Neuroglía/citología , Neuronas/citología , Ratas , Ratas Sprague-Dawley
3.
Anesthesiology ; 95(3): 756-65, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11575551

RESUMEN

BACKGROUND: Volatile anesthetics are known to ameliorate experimental ischemic brain injury. A possible mechanism is inhibition of excitotoxic cascades induced by excessive glutamatergic stimulation. This study examined interactions between volatile anesthetics and excitotoxic stress. METHODS: Primary cortical neuronal-glial cultures were exposed to N-methyl-D-aspartate (NMDA) or glutamate and isoflurane (0.1-3.3 mM), sevoflurane (0.1-2.9 mM), halothane (0.1-2.9 mM), or 10 microM (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]-cyclohepten-5,10-imine hydrogen maleate (MK-801). Lactate dehydrogenase release was measured 24 h later. In other cultures, effects of volatile anesthetics on Ca++ uptake and mitochondrial membrane potential were determined in the presence or absence of NMDA (0-200 microM). RESULTS: Volatile anesthetics reduced excitotoxin induced lactate dehydrogenase release by up to 52% in a dose-dependent manner. At higher concentrations, this protection was reversed. When corrected for olive oil solubility, the three anesthetics offered equivalent protection. MK-801 provided near-complete protection. Ca++ uptake was proportionally reduced with increasing concentrations of anesthetic but did not account for reversal of protection at higher anesthetic concentrations. Given equivalent NMDA-induced Ca++ loads, cells treated with volatile anesthetic had greater lactate dehydrogenase release than those left untreated. At protective concentrations, volatile anesthetics partially inhibited NMDA-induced mitochondrial membrane depolarization. At higher concentrations, volatile anesthetics alone were sufficient to induce mitochondrial depolarization. CONCLUSIONS: Volatile anesthetics offer similar protection against excitotoxicity, but this protection is substantially less than that provided by selective NMDA receptor antagonism. Peak effects of NMDA receptor antagonism were observed at volatile anesthetic concentrations substantially greater than those used clinically.


Asunto(s)
Anestésicos por Inhalación/farmacología , N-Metilaspartato/antagonistas & inhibidores , Neuroglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Maleato de Dizocilpina/farmacología , Relación Dosis-Respuesta a Droga , Ácido Glutámico/farmacología , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Neuroglía/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
4.
J Neurotrauma ; 18(6): 625-34, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11437085

RESUMEN

Oxidative stress is known to play an important role in the response of brain to traumatic insults. We tested the hypothesis that increased extracellular superoxide dismutase (EC-SOD) expression can reduce injury in a mouse model of closed head injury. Neurologic, cognitive, and histologic outcomes were compared between transgenic mice exhibiting a fivefold increase in EC-SOD activity and wild-type littermate controls. Severe or moderate transcranial impact was induced in anesthetized and physiologically controlled animals. After severe impact, transgenic mice had better neurological outcome at 24 hr postinjury (p = 0.038). Brain water content was increased, but there was no difference between groups. Moderate impact resulted in predominantly mild neurologic deficits in both groups at both 24 hr and 14 days postinjury. Morris water maze performance, testing cognitive function at 14-17 days after trauma, was better in EC-SOD overexpressors (p = 0.018). No differences were observed between groups for histologic damage in hippocampal CA1 and CA3. We conclude that EC-SOD has a beneficial effect on behavioral outcome after both severe and moderate closed head injury in mice. Because EC-SOD is believed to be predominantly located in the extracellular space, these data implicate an adverse effect of extracellular superoxide anion on outcome from closed head injury.


Asunto(s)
Conducta Animal/fisiología , Traumatismos Cerrados de la Cabeza/enzimología , Traumatismos Cerrados de la Cabeza/psicología , Superóxido Dismutasa/biosíntesis , Animales , Western Blotting , Recuento de Células , Cognición/fisiología , Espacio Extracelular/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Traumatismos Cerrados de la Cabeza/patología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Estrés Oxidativo/fisiología
5.
J Neurosci ; 21(13): 4582-92, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11425886

RESUMEN

Reactive oxygen species contribute to ischemic brain injury. This study examined whether the porphyrin catalytic antioxidant manganese (III) meso-tetrakis (N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+)) reduces oxidative stress and improves outcome from experimental cerebral ischemia. Rats that were subjected to 90 min focal ischemia and 7 d recovery were given MnTE-2-PyP(5+) (or vehicle) intracerebroventricularly 60 min before ischemia, or 5 or 90 min or 6 or 12 hr after reperfusion. Biomarkers of brain oxidative stress were measured at 4 hr after postischemic treatment (5 min or 6 hr). MnTE-2-PyP(5+), given 60 min before ischemia, improved neurologic scores and reduced total infarct size by 70%. MnTE-2-PyP(5+), given 5 or 90 min after reperfusion, reduced infarct size by 70-77% and had no effect on temperature. MnTE-2-PyP(5+) treatment 6 hr after ischemia reduced total infarct volume by 54% (vehicle, 131 +/- 60 mm(3); MnTE-2-PyP(5+), 300 ng, 60 +/- 68 mm(3)). Protection was observed in both cortex and caudoputamen, and neurologic scores were improved. No MnTE-2-PyP(5+) effect was observed if it was given 12 hr after ischemia. MnTE-2-PyP(5+) prevented mitochondrial aconitase inactivation and reduced 8-hydroxy-2'-deoxyguanosine formation when it was given 5 min or 6 hr after ischemia. In mice, MnTE-2-PyP(5+) reduced infarct size and improved neurologic scores when it was given intravenously 5 min after ischemia. There was no effect of 150 or 300 ng of MnTE-2-PyP(5+) pretreatment on selective neuronal necrosis resulting from 10 min forebrain ischemia and 5 d recovery in rats. Administration of a metalloporphyrin catalytic antioxidant had marked neuroprotective effects against focal ischemic insults when it was given up to 6 hr after ischemia. This was associated with decreased postischemic superoxide-mediated oxidative stress.


Asunto(s)
Antioxidantes/administración & dosificación , Isquemia Encefálica/tratamiento farmacológico , Infarto Cerebral/prevención & control , Metaloporfirinas/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Aconitato Hidratasa/metabolismo , Animales , Antioxidantes/química , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Isquemia Encefálica/etiología , Catálisis , Infarto Cerebral/etiología , ADN/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Fumarato Hidratasa/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Inyecciones Intravenosas , Inyecciones Intraventriculares , Masculino , Metaloporfirinas/química , Ratones , Ratones Endogámicos C57BL , Mitocondrias/enzimología , Necrosis , Fármacos Neuroprotectores/química , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Resultado del Tratamiento
6.
Anesthesiology ; 94(2): 303-212, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11176096

RESUMEN

BACKGROUND: Volatile anesthetics decrease ischemic brain injury. Mechanisms for this protection remain under investigation. The authors hypothesized that volatile anesthetics serve as antioxidants in a neuronal-glial cell culture system. METHODS: Primary cortical neuronal-glial cultures were prepared from fetal rat brain. Cultures were exposed to iron, H2O2, or xanthine-xanthine oxidase for 30 min in serum-free media containing dissolved isoflurane (0-3.2 mm), sevoflurane (0-3.6 mm), halothane (0-4.1 mm), n-hexanol, or known antioxidants. Cell damage was assessed by release of lactate dehydrogenase (LDH) and trypan blue exclusion 24 h later. Lipid peroxidation was measured by the production of thiobarbituric acid-reactive substances in a cell-free lipid system. Iron and calcium uptake and mitochondrial depolarization were measured after exposure to iron in the presence or absence of isoflurane. RESULTS: Deferoxamine reduced LDH release caused by H2O2 or xanthine-xanthine oxidase, but the volatile anesthetics had no effect. Iron-induced LDH release was prevented by the volatile anesthetics (maximum effect for halothane = 1.2 mm, isoflurane = 1.2 mm, and sevoflurane = 2.1 mm aqueous phase). When corrected for lipid solubility, the three volatile anesthetics were equipotent against iron-induced LDH release. In the cell-free system, there was no effect of the anesthetics on thiobarbituric acid-reactive substance formation in contrast to Trolox, which provided complete inhibition. Isoflurane (1.2 mm) reduced mean iron uptake by 46% and inhibited mitochondrial depolarization but had no effect on calcium uptake. CONCLUSIONS: Volatile anesthetics reduced cell death induced by oxidative stress only in the context of iron challenge. The likely reason for protection against iron toxicity is inhibition of iron uptake and therefore indirect reduction of subsequent intracellular oxidative stress caused by this challenge. These data argue against a primary antioxidant effect of volatile anesthetics.


Asunto(s)
Anestésicos por Inhalación/farmacología , Antioxidantes/farmacología , Neuroglía/efectos de los fármacos , Animales , Calcio/metabolismo , Células Cultivadas , Femenino , Peróxido de Hidrógeno/farmacología , Hierro/metabolismo , Hierro/farmacología , L-Lactato Deshidrogenasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Malondialdehído/farmacología , Neuroglía/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Anesthesiology ; 93(4): 1102-6, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11020767

RESUMEN

BACKGROUND: Isoflurane improves outcome from near-complete forebrain ischemia in rats compared with fentanyl-nitrous oxide (N2O). Sympathetic ganglionic blockade with trimethaphan abolishes this beneficial effect. To evaluate whether anesthesia-related differences in cerebral blood flow (CBF) may explain these findings, this study compared regional CBF before, during, and after near-complete forebrain ischemia in rats anesthetized with either isoflurane (with and without trimethaphan) or fentanyl-nitrous oxide. METHODS: Fasted, normothermic isoflurane anesthetized Sprague-Dawley rats were prepared for near-complete forebrain ischemia (10 min of bilateral carotid occlusion and mean arterial pressure = 30 mmHg). After surgery, rats were anesthetized with either 1.4% isoflurane (with or without 2.5 mg of trimethaphan intravenously at onset of ischemia) or fentanyl-nitrous oxide (25 microgram. kg-1. h-1. 70% N2O-1). Regional CBF was determined (14C-iodoantipyrine autoradiography) before ischemia, 8 min after onset of ischemia, and 30 min after onset of reperfusion. RESULTS: Regional CBF did not differ significantly among groups at any measurement interval. Ischemia caused a marked flow reduction to 5% or less of baseline (P < 0.001) in selectively vulnerable regions, such as the cortex, caudoputamen and hippocampus, whereas flow in the brain stem and cerebellum was preserved. Reperfusion at 30 min was associated with partial restoration of flow to 35-50% of baseline values in ischemic structures. CONCLUSIONS: The results indicate that improved histologic-behavioral outcome provided by isoflurane anesthesia cannot be explained by differential vasodilative effects of the anesthetic states before, during, or after severe forebrain ischemia. This study also shows severe postischemic delayed hypoperfusion that was not affected by choice of anesthetic or the presence of trimethaphan. Mechanisms other than effects on periischemic CBF must be responsible for beneficial effects of isoflurane in this model.


Asunto(s)
Anestésicos por Inhalación/farmacología , Isquemia Encefálica/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Isoflurano/farmacología , Prosencéfalo/irrigación sanguínea , Anestésicos Intravenosos/farmacología , Animales , Bloqueo Nervioso Autónomo , Isquemia Encefálica/tratamiento farmacológico , Circulación Cerebrovascular/fisiología , Interacciones Farmacológicas , Fentanilo/farmacología , Bloqueadores Ganglionares/farmacología , Masculino , Antagonistas Nicotínicos/farmacología , Óxido Nitroso/farmacología , Prosencéfalo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Trimetafan/farmacología
8.
Anesthesiology ; 93(2): 431-6, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10910493

RESUMEN

BACKGROUND: Although reduction of cerebral metabolic rate is thought to contribute to anesthetic neuroprotection, histologic evidence to support this concept has not been provided. In this study, histologic outcome was evaluated in rats subjected to different durations of severe forebrain ischemia while anesthetized with volatile anesthetics that have substantially different effects on cerebral metabolic rate. METHODS: Normothermic rats that underwent fasting were anesthetized with 0.75 minimum alveolar concentration (MAC) isoflurane-60% nitrous oxide (N2O) or 0.75 MAC halothane-60% N2O. Ischemia was induced with use of a combination of bilateral carotid occlusion and controlled hypotension. Rats in the isoflurane group were subjected to 6.5 min or 8.0 min ischemia, whereas the halothane group received 6.5 min ischemia. Histologic damage was assessed 4 days later. RESULTS: With 6.5 min ischemia, mean +/- SD, hippocampal CA1 percent of dead (% dead) neurons was reduced with isoflurane-N2O (45 +/- 18) versus halothane-N2O (60 +/- 23, P = 0. 023). Eight minutes of ischemia increased % dead neurons in the isoflurane-N2O group (60 +/- 17, P = 0.017). There was no difference between the isoflurane 8.0-min and halothane 6.5-min groups (P = 0. 935). A similar pattern was observed in hippocampal CA4 and the neocortex. Striatal damage was not affected by anesthetic or ischemic duration. CONCLUSIONS: At 6.5 min ischemia, isoflurane provided improved outcome versus halothane. Previous research has shown that 0.75 MAC isoflurane-N2O increases the time to onset of ischemic depolarization by 1.5 min and reduces cerebral metabolic rate by 42% versus 0.75 MAC halothane-N2O. In the current study, when the duration of ischemia was increased by 1.5 min in the isoflurane-N2O group, histologic outcome became similar to that in halothane-N2O-anesthetized rats. These results provide evidence that cerebral metabolic rate reduction has an advantageous effect on outcome from severe brain ischemia, but also suggest that such benefit is likely to be small.


Asunto(s)
Anestésicos por Inhalación/uso terapéutico , Isquemia Encefálica/prevención & control , Encéfalo/efectos de los fármacos , Halotano/uso terapéutico , Isoflurano/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/metabolismo , Isquemia Encefálica/patología , Hemodinámica/efectos de los fármacos , Masculino , Valor Predictivo de las Pruebas , Ratas , Ratas Sprague-Dawley
9.
Exp Neurol ; 163(2): 392-8, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10833313

RESUMEN

Transgenic mice, which exhibit a fivefold increase in brain parenchymal extracellular superoxide dismutase (EC-SOD) activity, were used to investigate the role of EC-SOD in global ischemic brain injury. Halothane-anesthetized normothermic wild-type (n = 22) and transgenic (n = 20) mice underwent 10 min of near-complete forebrain ischemia induced by bilateral carotid artery occlusion and systemic hypotension (mean arterial pressure = 30 mm Hg). After 3 days of recovery, the brains were histologically examined. Other mice underwent autoradiographic determination of regional CBF 10 min prior to, during, and 30 min after forebrain ischemia. Histologic injury in the cortex and caudoputamen was minimal in both groups. The percentage of dead hippocampal CA1 neurons was reduced in the EC-SOD transgenic group (wild type = 44 +/- 28%; EC-SOD transgenic = 23 +/- 21%, mean +/- SD, P = 0.015). CBF was similar between groups prior to ischemia. The intraischemic blood flow was severely reduced in forebrain structures and was similar between groups. Blood flow at 30 min postischemia had recovered to 50-60% of baseline values in both groups. These results indicate that EC-SOD can play an important role in defining the magnitude of selective neuronal necrosis resulting from near-complete forebrain ischemia. This implicates involvement of extracellular superoxide anions in the pathologic response to global cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Prosencéfalo/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Isquemia Encefálica/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Prosencéfalo/irrigación sanguínea , Prosencéfalo/patología
10.
Brain Res ; 868(1): 14-21, 2000 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-10841883

RESUMEN

Genetically engineered mice are increasingly important in stroke research. The strains on which these constructs are built are known to have inherent differential sensitivities to ischemic insults. This has been largely attributed to differences in vascular anatomy. This study compared the outcome from forebrain ischemia in two common murine background strains using two different types of ischemic insult. C57Bl/6 and SV129 mice were subjected to two vessel (bilateral carotid) occlusion (2VO) or 2VO plus systemic hypotension (2VO+Hypo; mean arterial pressure=30+/-2 mmHg) for 10-20 min. Ventilation and pericranial temperature were controlled. Cerebral blood flow (CBF) was determined by 14C-iodoantipyrine autoradiography. Histologic damage in forebrain structures was measured 3 days post-ischemia. During 2VO+Hypo, the EEG became isoelectric in all animals. During 2VO alone, EEG isoelectricity occurred in 73% of C57Bl/6 and 50% of SV129 mice. Forebrain CBF was reduced to a similar extent in both strains. Greater CBF variability was seen with 2VO alone versus 2VO+Hypo. CBF was less in the 2VO+Hypo model. SV129 mice had wider posterior communicating but smaller basilar artery diameters. With or without hypotension, SV129 mice had markedly less severe histologic damage than C57Bl/6 mice. A time-dependent increase in histologic damage was demonstrated in the 2VO+Hypo model but not with 2VO alone. The 2VO and 2VO+Hypo models produced similar magnitudes of histologic injury in C57Bl/6 mice subjected to 10-min ischemia. SV129 mice were resistant to ischemia in either model. The 2VO+Hypo model produced a more uniform severity of ischemia as defined by CBF and EEG examination. Despite this, the murine strain had a substantially greater impact on histologic outcome than did cerebrovascular anatomy or the type of model used to produce the ischemic insult.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatología , Animales , Circulación Cerebrovascular/genética , Electroencefalografía , Predisposición Genética a la Enfermedad , Hipocampo/irrigación sanguínea , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Recuperación de la Función/genética , Especificidad de la Especie
11.
Anesth Analg ; 91(1): 145-51, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10866902

RESUMEN

UNLABELLED: Rats exposed to forebrain ischemia have reduced injury when anesthetized with isoflurane versus fentanyl + N(2)O. The protection caused by isoflurane is reversed by trimethaphan. We hypothesized that these anesthetic-dependent effects on ischemic outcome can be associated with altered stress responses to ischemia. Rats were randomized to four treatments: isoflurane; fentanyl + N(2)O; isoflurane + trimethaphan; or isoflurane + metyrapone. Severe forebrain ischemia was then induced for 10 min. Plasma and brain corticosterone, tumor necrosis factor (TNF)-alpha, and interleukin (IL)-6 were assayed. Plasma corticosterone concentrations were similar in the isoflurane and isoflurane + trimethaphan groups, but greater than in the fentanyl + N(2)O and isoflurane + metyrapone groups. Brain corticosterone was similar among all groups except isoflurane + metyrapone, in which values were markedly reduced. The addition of metyrapone to isoflurane also reduced plasma TNF-alpha; however, values among other groups were similar. There were no differences among groups for brain TNF-alpha. Plasma IL-6 concentrations were below the limit of detection. Brain IL-6 concentrations were increased by ischemia; however, there was no difference among groups. In conclusion, there were no differences between the isoflurane and isoflurane + trimethaphan groups for any of the measured stress markers. Further, there was little difference between the isoflurane and fentanyl + N(2)O groups, except for plasma corticosterone concentration. Accordingly, isoflurane neuroprotection and its reversal by trimethaphan appear to be independent of effects on the stress responses measured in this study. IMPLICATIONS: Differential anesthetic effects on ischemic outcome are independent of effects on adrenergic/noradrenergic responses to ischemia. The absence of a consistent differential effect of anesthetics on either corticosterone or cytokine responses to ischemia serves to further refute the hypothesis that isoflurane neuroprotection can be attributed to dampening of adverse stress responses to ischemic insults.


Asunto(s)
Anestésicos/farmacología , Isquemia Encefálica/fisiopatología , Prosencéfalo/irrigación sanguínea , Reperfusión , Estrés Fisiológico/metabolismo , Animales , Isquemia Encefálica/metabolismo , Corticosterona/metabolismo , Fentanilo/farmacología , Interleucina-6/metabolismo , Isoflurano/farmacología , Metirapona/farmacología , Óxido Nitroso/farmacología , Ratas , Ratas Sprague-Dawley , Trimetafan/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
12.
Brain Res ; 847(2): 262-9, 1999 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-10575096

RESUMEN

Manipulations of plasma catecholamine concentrations influence outcome from ischemic brain insults. It has been suggested that these effects are mediated by influences on brain catecholamine concentrations. This study examined whether major changes in brain norepinephrine concentrations can alter outcome from severe forebrain ischemia. Sprague-Dawley rats were administered 50 mg/kg i. p. N-(chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4) or were left untreated (control). One week later, these rats were subjected to either 7 or 8 min of normothermic forebrain ischemia (bilateral carotid occlusion and MABP=30 mmHg) and allowed to recover for 4 days. Histologic damage was then evaluated. In other control and DSP-4-treated animals, hippocampal microdialysate norepinephrine concentrations were measured before, during and after 8 min of forebrain ischemia. Norepinephrine concentrations were also determined in brain homogenates from non-ischemic DSP-treated and control rats. A 95% depletion of norepinephrine was observed in brain homogenates from non-ischemic DSP-4-treated rats compared with control. During ischemia, microdialysate norepinephrine concentrations increased in control but not in DSP-4-treated rats (P=0.002). For plasma, intra-ischemic epinephrine concentrations increased 8-10-fold and returned to baseline values post-ischemia with no differences between groups. Plasma norepinephrine values remained unchanged in both groups. Histologic damage resulting from either 7 or 8 min of ischemia in hippocampal structures, caudoputamen, and neocortex was similar between DSP-4-treated and control groups. This study could not identify any effect of major changes in brain norepinephrine concentrations on ischemic brain damage. These data indicate that peripheral catecholamine effects on near-complete forebrain ischemic outcome are unlikely to be mediated by effects on central catecholamine concentrations.


Asunto(s)
Isquemia Encefálica/fisiopatología , Norepinefrina/sangre , Prosencéfalo/irrigación sanguínea , Animales , Bencilaminas/farmacología , Isquemia Encefálica/metabolismo , Epinefrina/sangre , Masculino , Microdiálisis , Inhibidores de la Captación de Neurotransmisores/farmacología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Ratas , Ratas Sprague-Dawley
13.
Neurosci Lett ; 267(1): 13-6, 1999 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-10400237

RESUMEN

The role of endogenous extracellular superoxide dismutase (EC-SOD) was examined in a murine model of transient focal cerebral ischemia. Homozygous EC-SOD deficient (EC-SOD-/-; n = 18) and wild type (EC-SOD+/+; n = 19) littermates were anesthetized with halothane and subjected to 50 min of intraluminal middle cerebral artery occlusion with pericranial temperature maintained at 37.0 degrees C. After 24 h of reperfusion, resultant hemiparesis and cerebral infarct size were measured. Total infarct volume was 81% greater (P = 0.03) and hemiparesis was more severe (P = 0.01) in EC-SOD-/- versus EC-SOD+/+ mice. The worsened ischemic outcome observed in EC-SOD-/- mice is consistent with prior work which found transgenic EC-SOD overexpressing mice to exhibit enhanced tolerance to focal ischemia. The results suggest that endogenous antioxidant activity in the extracellular compartment plays an important role in the histologic/neurologic response to focal cerebral ischemia.


Asunto(s)
Matriz Extracelular/enzimología , Ataque Isquémico Transitorio/enzimología , Superóxido Dismutasa/deficiencia , Superóxido Dismutasa/genética , Animales , Infarto Cerebral/enzimología , Infarto Cerebral/genética , Infarto Cerebral/fisiopatología , Modelos Animales de Enfermedad , Ataque Isquémico Transitorio/genética , Ataque Isquémico Transitorio/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
14.
J Neurosci Methods ; 88(1): 103-9, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10379584

RESUMEN

Transgenic/knockout murine variants allow roles of specific proteins to be studied in cerebral ischemia. Because of the size of mice, however, study of prolonged recovery from global ischemia has been limited. This project characterized an adaptation of the rat two-vessel occlusion model of global ischemia for use in the mouse. C57B1/6J mice (8 weeks old; 21 +/- 1 g) were overnight fasted, anesthetized with halothane, intubated and mechanically ventilated. The right internal jugular vein and femoral artery were cannulated. Pericranial temperature was held at 37.0 degrees C. The carotid arteries were occluded and mean arterial pressure was reduced to 35 mmHg with 0.3 mg intra-arterial trimethaphan and venous exsanguination. Electroencephalographic isoelectricity was confirmed in cohort mice. Ten minutes later ischemia was reversed. Mice were allowed 1, 3 or 5 days survival followed by histologic analysis. Regional cerebral blood flow (CBF) was determined autoradiographically. Outcome effects of intra-ischemic hyperglycemia (approximately 350 mg/dl) or hypothermia (34 degrees C) were also examined. The mortality rate was less than 10% in all recovery groups. Ischemia caused reduction of CBF to < 2% of sham values in cortex, hippocampus, and caudoputamen. CBF was unchanged in thalamus, brainstem and cerebellum. CA1 damage, greater after 3 days vs. 1 day reperfusion, was not further increased at 5 days. Histologic injury was increased by hyperglycemia although seizures did not occur. Hypothermia reduced CA1 damage. This study demonstrates feasibility of using the two-vessel occlusion + hypotension recovery model in the mouse. Recovery intervals of > or = 3 days are required to account for delayed CA1 neuronal necrosis. Histologic outcome can be modulated by known physiologic determinants of ischemic brain damage.


Asunto(s)
Modelos Animales de Enfermedad , Ataque Isquémico Transitorio/fisiopatología , Ratones Endogámicos C57BL , Daño por Reperfusión/fisiopatología , Animales , Análisis de los Gases de la Sangre , Presión Sanguínea , Muerte Celular , Circulación Cerebrovascular , Masculino , Ratones , Neuronas/citología , Prosencéfalo/irrigación sanguínea , Prosencéfalo/citología , Prosencéfalo/fisiología
15.
Stroke ; 30(5): 1118-24, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10229753

RESUMEN

BACKGROUND AND PURPOSE: Apolipoprotein E (apoE) has been found relevant in a variety of central nervous system disorders. This experiment examined the effect of endogenous murine apoE on selective neuronal necrosis resulting from a transient forebrain ischemia insult. METHODS: ApoE deficient (n=16) and wild type (n=17) halothane-anesthetized mice were subjected to severe forebrain ischemia (10 minutes of bilateral carotid occlusion and systemic hypotension). After 3 days' recovery, brain injury was determined histologically. In other apoE-deficient and wild-type mice, regional cerebral blood flow (CBF) was determined by 14C-iodoantipyrine autoradiography 10 minutes before, 5 minutes after onset of, and 30 minutes after reperfusion from 10 minutes of forebrain ischemia. RESULTS: The percentage of dead hippocampal CA1 neurons (mean+/-SD) was greater in the apoE-deficient group (apoE deficient=67+/-30%; wild type=37+/-33%; P=0.011). A similar pattern was observed in the caudoputamen (P=0.002) and neocortex (P=0.014). Cerebral blood flow was similar between groups at each measurement interval. Marked hypoperfusion persisted in both groups at 30 minutes after ischemia. CONCLUSIONS: ApoE deficiency worsens ischemic outcome. This is not attributable to effects on CBF. A role of apoE in the cerebral response to global ischemia is consistent with prior reports that murine apoE deficiency increases infarct size resulting from focal cerebral ischemia.


Asunto(s)
Apolipoproteínas E/genética , Arteriopatías Oclusivas/metabolismo , Isquemia Encefálica/metabolismo , Circulación Cerebrovascular/fisiología , Animales , Apolipoproteínas E/deficiencia , Análisis de los Gases de la Sangre , Presión Sanguínea , Cerebelo/irrigación sanguínea , Corteza Cerebral/irrigación sanguínea , Modelos Animales de Enfermedad , Femenino , Hipocampo/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neostriado/irrigación sanguínea , Puente/irrigación sanguínea , Tálamo/irrigación sanguínea
16.
Brain Res ; 828(1-2): 12-9, 1999 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10320720

RESUMEN

The pedunculopontine nucleus (PPN) located in the mesopontine tegmentum is innervated by descending projections from nuclei in the basal ganglia. The present study was performed to determine whether nigrostriatal dopaminergic neuron degeneration is associated with changes in PPN metabolic activity. Unilateral nigrostriatal lesioning was performed by injecting 6-hydroxydopamine (6-OHDA) into the substantia nigra pars compacta in 10 rats. Six of these animals exhibited apomorphine-induced rotations contralateral to the lesion and were included in the experimental group for determination of regional cerebral metabolic rate for glucose (rCMRglucose) along with five sham-lesioned and five normal controls. All studies were performed 13-15 days after lesioning using [14]C-2-deoxyglucose autoradiography. Significant hemispheric differences in metabolic activity were observed only in the 6-OHDA lesioned animals. Increased rCMRglucose was found in the globus pallidus (+63%) ipsilateral to the lesion as compared to the contralateral hemisphere, and reduced rCMRglucose in the primary motor, sensory, and auditory cortex (-7%, -12% and -7%, respectively), and in the subthalamic nucleus (-6%). Metabolic activity within the PPN ipsilateral to the lesion was significantly greater than the contralateral hemisphere (P<0.05; lesion 57+/-8, nonlesion 52+/-5), and significantly greater than the sham-lesioned side of the sham rat (P<0.05; sham lesion 47+/-5). No hemispheric differences were observed in the lateral dorsal tegmental nucleus. These observations offer further support for a role of the PPN in Parkinson's and for the utility of the rodent unilateral 6-OHDA model in defining the pathophysiologic significance of the mesopontine tegmental striatal-motor interfaces in basal ganglia disease.


Asunto(s)
Enfermedad de Parkinson Secundaria/metabolismo , Puente/metabolismo , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Antimetabolitos/farmacología , Enfermedades de los Ganglios Basales/inducido químicamente , Enfermedades de los Ganglios Basales/metabolismo , Corteza Cerebral/metabolismo , Desoxiglucosa/farmacología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Masculino , Oxidopamina , Enfermedad de Parkinson Secundaria/inducido químicamente , Ratas , Ratas Sprague-Dawley , Simpaticolíticos
17.
Brain Res ; 826(2): 172-80, 1999 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-10224294

RESUMEN

This study examined the effect of a pharmacologically induced rightward shift in the partial pressure of oxygen at which 50% of hemoglobin is saturated (P50) on outcome from transient focal cerebral ischemia in the rat. Halothane anesthetized rats (n=20 per group) were given saline or a single 15-min infusion of 150 mg/kg RSR13 (2-[4-[[3,5-dimethylanilino) carbonyl]methyl]phenoxy]-2-methylproprionic acid) intravenously before or 30 min after onset of 75 min of middle cerebral artery filament occlusion (MCAO). Seven days later, severity of hemiparesis and cerebral infarct size were examined. RSR13 alone did not significantly improve outcome. Conscious normothermic rats (n=12 per group) were also given RSR13 (150 mg/kg) or 0.9% NaCl intravenously and subjected to 75 min of MCAO with 7 days of recovery. Again, RSR13 alone did not significantly reduce infarct size or improve neurologic score. A dose-response curve for dizocilpine (MK-801) was then constructed in conscious normothermic rats subjected to 75 min of MCAO. Dizocilpine (0.5 mg/kg i.v.) caused a 90% reduction in mean infarct size while 0.25 mg/kg reduced infarct size by 48%. Other rats were then subjected to 75 min of MCAO after being given dizocilpine (0.25 mg/kg i.v.; n=18) or RSR13 (150 mg/kg i.v. )+dizocilpine (0.25 mg/kg i.v.; n=15). RSR13+dizocilpine resulted in smaller cortical infarct volume (8+/-14 mm3 vs. 34+/-37 mm3, p<0.02) and total cerebral infarct volume (46+/-28 mm3 vs. 81+/-60 mm3, p<0. 05) compared to dizocilpine alone, respectively. We conclude that a pre-ischemic peak increase in P50 of approximately 25 mmHg alone is insufficient to reduce focal ischemic injury, but may be advantageous when used in conjunction with other neuroprotective agents.


Asunto(s)
Compuestos de Anilina/farmacología , Antidrepanocíticos/farmacología , Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hemoglobinas/efectos de los fármacos , Ataque Isquémico Transitorio/tratamiento farmacológico , Propionatos/farmacología , Sitio Alostérico/efectos de los fármacos , Animales , Análisis de los Gases de la Sangre , Temperatura Corporal , Química Encefálica/efectos de los fármacos , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/patología , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/patología , Quimioterapia Combinada , Hemoglobinas/química , Ataque Isquémico Transitorio/patología , Masculino , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
18.
Anesth Analg ; 88(4): 787-92, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10195525

RESUMEN

UNLABELLED: We postulated that adrenergic responses to global cerebral ischemia are anesthetic-dependent and similar in both brain and arterial blood. Rats were anesthetized with isoflurane (1.4%), ketamine (1 mg x kg(-1) x min(-1)), or fentanyl (25 microg x kg(-1) x h(-1))/70% N2O. The carotid arteries were occluded for either 20 min with mean arterial pressure (MAP) 50 mm Hg (incomplete ischemia) or 10 min with MAP 30 mm Hg (near-complete ischemia). Norepinephrine was measured in hippocampal microdialysate. Norepinephrine and epinephrine were measured in arterial plasma. In both hippocampus and plasma, basal norepinephrine was similar among anesthetics. During incomplete ischemia, hippocampal norepinephrine was twofold greater with fentanyl/N2O than with isoflurane (P = 0.037), but plasma norepinephrine and epinephrine were similar and unchanged among all three anesthetics. During near-complete ischemia, hippocampal norepinephrine was threefold greater with ketamine than fentanyl/N2O (P = 0.005), whereas plasma norepinephrine and epinephrine were markedly greater with fentanyl/N2O than with ketamine (P < 0.0005) or isoflurane (P = 0.05). There was no correlation between norepinephrine concentrations in hippocampus and plasma for either incomplete or near-complete ischemia. This study demonstrates that adrenergic responses to global ischemia are anesthetic-dependent, particularly during more severe insults. The absence of a correlation between plasma and brain catecholamine concentrations indicates that adrenergic responses to ischemia are independent in brain and blood. IMPLICATIONS: It has been proposed that anesthetics modulate cerebral ischemic outcome by influencing peripheral adrenergic responses to ischemia. This experiment demonstrates that anesthetics differentially modulate adrenergic responses to ischemia but that effects in plasma and brain are independent. This suggests that events detected in the peripheral circulation do not implicate direct mechanisms of action of catecholamines at the neuronal/glial level.


Asunto(s)
Anestésicos por Inhalación/farmacología , Anestésicos Intravenosos/farmacología , Isquemia Encefálica/metabolismo , Catecolaminas/sangre , Fentanilo/farmacología , Hipocampo/efectos de los fármacos , Isoflurano/farmacología , Ketamina/farmacología , Óxido Nitroso/farmacología , Animales , Isquemia Encefálica/sangre , Isquemia Encefálica/etiología , Hipocampo/metabolismo , Masculino , Microdiálisis , Ratas , Ratas Sprague-Dawley
19.
Neuroscience ; 88(1): 185-91, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10051199

RESUMEN

Transgenic mice, which had been transfected with the human extracellular superoxide dismutase gene, causing an approximate five-fold increase in brain parenchymal extracellular superoxide dismutase activity, were used to investigate the role of extracellular superoxide dismutase in ischemic brain injury. Transgenic (n = 21) and wild-type (n = 19) mice underwent 90 min of intraluminal middle cerebral artery occlusion and 24 h of reperfusion. Severity of resultant hemiparesis and cerebral infarct size were measured. Wild-type mice had larger infarcts (cortex: wild type =37+/-14 mm3, transgenic = 27+/-13 mm3, P=0.03; subcortex: wild type = 33+/-14 mm3, transgenic = 23+/-10 mm3, P = 0.02). Neurological scores, however, were similar (P = 0.29). Other mice underwent autoradiographic determination of intra-ischemic cerebral blood flow. The volume of tissue at risk of infarction (defined as volume of tissue where blood flow was <25 ml/100g/min) was similar between groups (cortex: wild type = 51+/-15 mm3, transgenic = 47+/-9 mm3, P=0.65; subcortex: wild type = 39+/-16 mm3, transgenic= 37+/-17 mm3, P=0.81). These results indicate that antioxidant scavenging of free radicals by extracellular superoxide dismutase plays an important role in the histological response to a focal ischemic brain insult.


Asunto(s)
Infarto Cerebral/prevención & control , Hemiplejía/prevención & control , Ataque Isquémico Transitorio/fisiopatología , Superóxido Dismutasa/genética , Animales , Presión Sanguínea , Temperatura Corporal , Infarto Cerebral/patología , Circulación Cerebrovascular , Círculo Arterial Cerebral/anomalías , Círculo Arterial Cerebral/anatomía & histología , Lateralidad Funcional , Hematócrito , Hemiplejía/fisiopatología , Humanos , Inmunidad Innata/genética , Ataque Isquémico Transitorio/genética , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Ratones Transgénicos , Factores de Riesgo , Superóxido Dismutasa/metabolismo
20.
Anesthesiology ; 90(3): 873-81, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10078690

RESUMEN

BACKGROUND: Isoflurane-anesthetized rats have better outcome from global cerebral ischemia than rats anesthetized with fentanyl and nitrous oxide. The authors wanted to determine whether circulating catecholamine concentrations depend on the anesthetic agent and whether sympathetic ganglionic blockade affects anesthetic-mediated differences in outcome from near-complete forebrain ischemia. METHODS: For two different experiments, normothermic Sprague-Dawley rats that had fasted were assigned to one of four groups and subjected to 10 min of 30 mm Hg mean arterial pressure and bilateral carotid occlusion. Rats were anesthetized with 1.4% isoflurane or fentanyl (25 microg x kg(-1) x h(-1)) and 70% nitrous oxide, with or without preischemic trimethaphan (2.5 mg given intravenously). In experiment 1, arterial plasma catecholamine concentrations were measured before, at 2 and 8 min during, and after ischemia (n = 5-8). In experiment 2, animals (n = 15) underwent histologic analysis 5 days after ischemia. RESULTS: In experiment 1, intraischemic increases in plasma norepinephrine and epinephrine levels were 28 and 12 times greater in the fentanyl-nitrous oxide group than in the isoflurane group (P<0.01). Trimethaphan blocked all changes in plasma catecholamine concentrations (P<0.02). In experiment 2, isoflurane reduced the mean +/- SD percentage of dead hippocampal CA1 neurons compared with fentanyl-nitrous oxide (43+/-22% vs. 87+/-10%; P<0.001). Trimethaphan abolished the beneficial effects of isoflurane (91+/-6%; P<0.001). Similar observations were made in the cortex. CONCLUSIONS: Isoflurane attenuated the peripheral sympathetic response to ischemia and improved histologic outcome compared with fentanyl and nitrous oxide. This outcome benefit was reversed by sympathetic ganglionic blockade. The beneficial effects of isoflurane may result from a neuroprotective influence of an intermediate sympathetic response that is abolished by trimethaphan.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Isquemia Encefálica/fisiopatología , Ganglios Simpáticos/fisiopatología , Isoflurano/administración & dosificación , Animales , Isquemia Encefálica/patología , Ganglios Simpáticos/efectos de los fármacos , Bloqueadores Ganglionares/administración & dosificación , Inyecciones Intravenosas , Masculino , Prosencéfalo/irrigación sanguínea , Prosencéfalo/patología , Prosencéfalo/fisiopatología , Ratas , Ratas Sprague-Dawley , Trimetafan/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...