Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Neurobiol ; 36(1): 68-74, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17952651

RESUMO

In response to traumatic brain injury, there is local and transient accumulation of 2-AG at the site of injury, peaking at 4 h and sustained up to at least 24 h. Neuroprotection exerted by exogenous 2-AG suggests that the formation of 2-AG may serve as a molecular regulator of pathophysiological events, attenuating the brain damage. Inhibition of this protective effect by SR-141716A, a CB(1) cannabinoid receptor antagonist, and the lack of effect of 2-AG in CB(1) knockout mice suggest that 2-AG and the CB(1) receptor may be important in the pathophysiology of traumatic brain injury. 2-AG exerts its neuroprotective effect after traumatic brain injury, at least in part, by inhibition of NF-kappaB transactivation. 2-AG also inhibits, at an early stage (2-4 h), the expression of the main proinflammatory cytokines, TNF-alpha, IL-6, and IL-1beta, and is accompanied by reduction of BBB permeability. Moreover, the CB(1), CB(2), and TRVP1 receptors are expressed on microvascular endothelial cells, and their activation by 2-AG counteracts endothelin (ET-1)-induced cerebral microvascular responses (namely, Ca(2+) mobilization and cytoskeleton rearrangement). This suggests that the functional interaction between 2-AG and ET-1 may provide a potential alternative pathway for abrogating ET-1-inducible vasoconstriction after brain injury and play a role in the neuroprotective effects exerted by 2-AG, as a potent vasodilator.


Assuntos
Lesões Encefálicas/metabolismo , Moduladores de Receptores de Canabinoides/metabolismo , Endocanabinoides , Animais , Barreira Hematoencefálica , Edema Encefálico/metabolismo , Glicerídeos/metabolismo , Humanos , Oxirredução
2.
Cytokine Growth Factor Rev ; 10(2): 119-30, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10743503

RESUMO

Brain injury (ischemia, trauma) is among the leading cause of mortality and disability in the western world. It induces increased production of tumor necrosis factor (TNF alpha) by brain resident cells. There is conflicting evidence on the role of this response in the injured brain, showing its potential effect in both processes of repair and of damage. This review presents data from clinical and experimental studies on the stimulation of TNF alpha production in brain injury and on the deleterious consequence of this acute response. Its inhibition by pharmacologic agents, neutralizing antibodies or soluble receptors has protective effects. In contrast, there are reports (from in-vitro studies or knock-out mice) on the beneficial effects of TNF alpha. To reconcile these apparently conflicting reports, the exact timing and extent of TNF alpha activation must be taken into account, as well as the presence of other mediators such as reactive oxygen species. It is suggested that the appropriate context of mediators, at any given time after brain injury may well determine whether the effect of TNF alpha is protective or toxic.


Assuntos
Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Lesões Encefálicas/tratamento farmacológico , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Humanos , Camundongos , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Chamariz do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/farmacologia
3.
FASEB J ; 19(12): 1701-3, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16099948

RESUMO

Traumatic brain injury (TBI) is a leading cause of morbidity and mortality in young people in industrialized countries. Although various anti-inflammatory and antiapoptotic modalities have shown neuroprotective effects in experimental models of TBI, to date, no specific pharmacological agent aimed at blocking the progression of secondary brain damage has been approved for clinical use. Erythropoietin (Epo) belongs to the cytokine superfamily and has traditionally been viewed as a hematopoiesis-regulating hormone. The newly discovered neuroprotective properties of Epo lead us to investigate its effect in TBI in a mouse model of closed head injury. Recombinant human erythropoietin (rhEpo) was injected at 1 and 24 h after TBI, and the effect on recovery of motor and cognitive functions, tissue inflammation, axonal degeneration, and apoptosis was evaluated up to 14 days. Motor deficits were lower, cognitive function was restored faster, and less apoptotic neurons and caspase-3 expression were found in rhEpo-treated as compared with vehicle-treated animals (P<0.05). Axons at the trauma area in rhEpo-treated mice were relatively well preserved compared with controls (shown by their density; P<0.01). Immunohistochemical analysis revealed a reduced activation of glial cells by staining for GFAP and complement receptor type 3 (CD11b/CD18) in the injured hemisphere of Epo- vs. vehicle-treated animals. We propose that further studies on Epo in TBI should be conducted in order to consider it as a novel therapy for TBI.


Assuntos
Apoptose , Eritropoetina/farmacologia , Eritropoetina/fisiologia , Traumatismos Cranianos Fechados/patologia , Neurônios/patologia , Animais , Anti-Inflamatórios/farmacologia , Axônios/metabolismo , Encéfalo/patologia , Antígeno CD11b/biossíntese , Antígenos CD18/biossíntese , Caspase 3 , Caspases/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Eritropoetina/química , Eritropoetina/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Hematopoese , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Inflamação , Masculino , Camundongos , Neurônios/metabolismo , Ratos , Proteínas Recombinantes/química , Fatores de Tempo
4.
Biochim Biophys Acta ; 601(1): 206-19, 1980 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-6250608

RESUMO

The uptake of uridine by mammalian cells consists of transport of uridine across the plasma membrane followed by its metabolic conversion, mainly by phosphorylation. S-substituted aromatic derivatives of 6-mercaptopurine ribosides are potent inhibitors of the nucleoside uptake systems in human erythrocytes and in mammalian cells in culture and have been studied extensively. We present here a theoretical analysis which enables one to decide whether transport of metabolites, their metabolic trapping within the cell, or both, are susceptible to inhibition. This analysis was applied in the study of the effect of some inhibitors on uridine and cytosine-beta-D-arabinoside uptake by transformed Nil-8 cells. It was found that in Nil-SV cells, both transport and metabolic conversion are susceptible to inhibition by nitrobenzylmercaptoinosine and by dansylaminoethylmercaptoguanosine. Nitrobenzylmercaptoinosine displays inhibition constants of 20 and 7 nM for transport and phosphorylation, respectively, while for dansylaminoethylmercaptoguanosine the inhibition constants are 1.8 and 0.6 microM, respectively, for the same processes. Cytosine-beta-D-arabinoside is a synthetic nucleoside which is not metabolizable in Nil cells. Its uptake properties are determined by the transport mechanism alone. The transport of this nucleoside into Nil-SV cells in inhibited by nitrobenzylmercaptoinosine and the inhibition constant found is approx. 5 times greater than that for uridine.


Assuntos
Transformação Celular Viral , Inosina/análogos & derivados , Tioinosina/farmacologia , Uridina/metabolismo , Animais , Linhagem Celular , Cricetinae , Citarabina/metabolismo , Desoxiglucose/farmacologia , Cinética , Mesocricetus , Fosforilação , Vírus do Sarcoma Murino , Tioinosina/análogos & derivados
5.
Biochim Biophys Acta ; 1211(3): 283-8, 1994 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-8130261

RESUMO

Incubation of cultured ventricular cardiomyocytes with high oligomycin concentrations (100 micrograms/ml), either alone or combined with 2-deoxyglucose (20 mM), led to the rapid depletion of cellular ATP. Inositol (poly)phosphate production decreased, and 6-keto PGF1 alpha production was increased. In cells depleted of ATP, either by low oligomycin concentrations or by sodium azide, 6-keto PGF1 alpha was not appreciably increased. There was a 25% rise in the release of fatty acids from the sn-2 position in glycerophospholipids. We suggest that oligomycin at high concentrations causes the release of free arachidonic acid from phospholipids either by non-PIP2-specific PLC and DG lipase or by phospholipase D, phosphatidic acid phosphatase and DG lipase. The effect is unrelated to decreased cellular ATP content.


Assuntos
6-Cetoprostaglandina F1 alfa/biossíntese , Miocárdio/metabolismo , Oligomicinas/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Desoxiglucose/farmacologia , Fosfatos de Inositol/metabolismo , Ratos
6.
Biochim Biophys Acta ; 1453(3): 359-68, 1999 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-10101254

RESUMO

Apolipoprotein E deficient mice have distinct memory deficits and neurochemical derangements and their recovery from closed head injury is impaired. In the present study, we examined the possibility that the neuronal derangements of apolipoprotein E deficient mice are associated with oxidative stress, which in turn affects their ability to recover from close head injury. It was found that brain phospholipid levels in apolipoprotein E deficient mice are lower than those of the controls (55+/-15% of control, P<0. 01), that the cholesterol levels of the two mice groups are similar and that the levels of conjugated dienes of the apolipoprotein E deficient mice are higher than those of control mice (132+/-15% of P<0.01). Brains of apolipoprotein E deficient mice had higher Mn-superoxide dismutase (134+/-7%), catalase (122+/-8%) and glutathione reductase (167+/-7%) activities than control (P<0.01), whereas glutathione peroxidase activity and the levels of reduced glutathione and ascorbic acid were similar in the two mouse groups. Closed head injury increased catalase and glutathione peroxidase activities in both mouse groups, whereas glutathione reductase increased only in control mice. The superoxide dismutase activity was unaffected in both groups. These findings suggest that the antioxidative metabolism of apolipoprotein E deficient mice is altered both prior to and following head injury and that antioxidative mechanisms may play a role in mediating the neuronal maintenance and repair derangements of the apolipoprotein E deficient mice.


Assuntos
Apolipoproteínas E/deficiência , Química Encefálica , Traumatismos Cranianos Fechados/metabolismo , Estresse Oxidativo , Animais , Antioxidantes/análise , Ácido Ascórbico/análise , Catalase/análise , Glutationa/análise , Glutationa Peroxidase/análise , Glutationa Redutase/análise , Glutationa Transferase/análise , Masculino , Camundongos , Superóxido Dismutase/análise
7.
Sci STKE ; 2002(129): re5, 2002 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-11972360

RESUMO

Traumatic brain injury (TBI) releases harmful mediators that lead to secondary damage. On the other hand, neuroprotective mediators are also released, and the balance between these classes of mediators determines the final outcome after injury. Recently, it was shown that the endogenous brain cannabinoids anandamide and 2-Arachidonoyl glycerol (2-AG) are also formed after TBI in rat and mouse respectively, and when administered after TBI, they reduce brain damage. In the case of 2-AG, better results are seen when it is administered together with related fatty acid glycerol esters. Significant reduction of brain edema, better clinical recovery, and reduced infarct volume and hippocampal cell death are noted. This new neuroprotective mechanism may involve inhibition of transmitter release and of inflammatory response. 2-AG is also a potent modulator of vascular tone, and counteracts the endothelin (ET-1)-induced vasoconstriction that aggravates brain damage; it may thus help to restore blood supply to the injured brain.


Assuntos
Dano Encefálico Crônico/etiologia , Dano Encefálico Crônico/prevenção & controle , Lesões Encefálicas/complicações , Lesões Encefálicas/fisiopatologia , Canabinoides/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Química Encefálica/fisiologia , Dano Encefálico Crônico/metabolismo , Lesões Encefálicas/metabolismo , Moduladores de Receptores de Canabinoides , Canabinoides/metabolismo , Endocanabinoides , Humanos , Estrutura Molecular , Fármacos Neuroprotetores/metabolismo
8.
FASEB J ; 15(12): 2294-6, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11511530

RESUMO

The biogenic amine The biogenic amine N1-acetyl-N2-formyl-5-methoxykynuramine (AFMK) was investigated for its potential antioxidative capacity. AFMK is a metabolite generated through either an enzymatic or a chemical reaction pathway from melatonin. The physiological function of AFMK remains unknown. To our knowledge, this report is the first to document the potent antioxidant action of this biogenic amine. Cyclic voltammetry (CV) shows that AFMK donates two electrons at potentials of 456 mV and 668 mV, and therefore it functions as a reductive force. This function contrasts with all other physiological antioxidants that donate a single electron only when they neutralize free radicals. AFMK reduced 8-hydroxydeoxyguanosine formation induced by the incubation of DNA with oxidants significantly. Lipid peroxidation resulting from free radical damage to rat liver homogenates was also prevented by the addition of AFMK. The inhibitory effects of AFMK on both DNA and lipid damage appear to be dose-response related. In cell culture, AFMK efficiently reduced hippocampal neuronal death induced by either hydrogen peroxide, glutamate, or amyloid b25-35 peptide. AFMK is a naturally occurring molecule with potent free radical scavenging capacity (donating two electrons/molecule) and thus may be a valuable new antioxidant for preventing and treating free radical-related disorders.


Assuntos
Antioxidantes/farmacologia , Desoxiguanosina/análogos & derivados , Cinuramina/farmacologia , 8-Hidroxi-2'-Desoxiguanosina , Peptídeos beta-Amiloides/farmacologia , Animais , Morte Celular , Células Cultivadas , Quelantes , Desoxiguanosina/metabolismo , Compostos Ferrosos/farmacologia , Radicais Livres/metabolismo , Ácido Glutâmico/farmacologia , Peróxido de Hidrogênio/farmacologia , Cinuramina/análogos & derivados , Peroxidação de Lipídeos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Melatonina/metabolismo , Metais Pesados/metabolismo , Modelos Biológicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Ratos
9.
J Mol Med (Berl) ; 78(4): 228-36, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10933585

RESUMO

Closed head injury (CHI) is an important cause of death among young adults and a prominent risk factor for nonfamilial Alzheimer's disease. Emergency intervention following CHI should therefore strive to improve survival, promote recovery, and prevent delayed neuropathologies. We employed high-resolution nonradioactive in situ hybridization to determine whether a single intracerebro-ventricular injection of 500 ng 2'-O-methyl RNA-capped antisense oligonucleotide (AS-ODN) against acetylcholinesterase (AChE) mRNA blocks overexpression of the stress-related readthrough AChE (AChE-R) mRNA splicing variant in head-injured mice. Silver-based Golgi staining revealed pronounced dendrite outgrowth in somatosensory cortex of traumatized mice 14 days postinjury that was associated with sites of AChE-R mRNA overexpression and suppressed by anti-AChE AS-ODNs. Furthermore, antisense treatment reduced the number of dead CA3 hippocampal neurons in injured mice, and facilitated neurological recovery as determined by performance in tests of neuromotor coordination. In trauma-sensitive transgenic mice overproducing AChE, antisense treatment reduced mortality from 50% to 20%, similar to that displayed by head-injured control mice. These findings demonstrate the potential of antisense therapeutics in treating acute injury, and suggest antisense prevention of AChE-R overproduction to mitigate the detrimental consequences of various traumatic brain insults.


Assuntos
Acetilcolinesterase/metabolismo , Encéfalo/efeitos dos fármacos , Traumatismos Cranianos Fechados/prevenção & controle , Neurônios/efeitos dos fármacos , Oligonucleotídeos Antissenso/uso terapêutico , Acetilcolinesterase/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Traumatismos Cranianos Fechados/metabolismo , Traumatismos Cranianos Fechados/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hibridização In Situ/métodos , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/patologia , RNA Mensageiro/metabolismo , Receptores Colinérgicos/metabolismo
10.
Endocrinology ; 132(3): 941-5, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8440197

RESUMO

Glucocorticoids (GC) are known to inhibit eicosanoid production in various peripheral tissues; however, their role in the regulation of basal and induced prostaglandin (PG) biosynthesis in the brain is still not clear. In the present study we examined the effect of exogenous dexamethasone (dex) or endogenous GC on basal and on bacterial endotoxin (lipopolysaccharide, LPS) induced ex vivo production of PGE2 by the frontal cortex of rat brain. The experimental groups were: 1) intact rats; 2) rats in whom endogenous GC were removed either by surgical or by chemical (metopirone) adrenalectomy (adex); and 3) rats exposed to specific corticosteroid receptor antagonists. In intact rats, the basal rate of PGE2 ex vivo synthesis was about 120 pg/mg protein.hr; dex (0.05-0.5 mg/100 g body wt ip) did not affect this level. Exposure to LPS (50 micrograms, intracerebroventricular) induced a 2-fold increase in PGE2, whereas pretreatment with dex abolished this increase. Bilateral adex or metopirone alone did not change PGE2 synthesis, whereas LPS administration to surgical or chemical adex rats resulted in a 4-fold increase in PGE2 production. Administration (intracerebroventricular) of either one or both of the specific corticosteroid receptor antagonists, RU-28318 (type I) and RU-38486 (type II) did not affect basal PGE2 production. When LPS was given after either one of these antagonists, a slight but significant elevation of PGE2 occurred, as compared to LPS-treated controls. When both antagonists were coadministered, the LPS-induced production of PGE2 was much more pronounced, similar to levels of LPS-treated, adex rats. These results suggest that LPS-induced production of PGE2, but not the basal production, is regulated by either endogenous or exogenous GC, and the inhibitory effect of GC on brain PG synthesis is mediated via both type I and II corticosteroid receptors.


Assuntos
Córtex Cerebral/metabolismo , Ventrículos Cerebrais/fisiologia , Dexametasona/farmacologia , Dinoprostona/biossíntese , Lipopolissacarídeos/farmacologia , Adrenalectomia , Animais , Córtex Cerebral/efeitos dos fármacos , Ventrículos Cerebrais/efeitos dos fármacos , Técnicas In Vitro , Injeções Intraventriculares , Lipopolissacarídeos/administração & dosagem , Masculino , Mifepristona/farmacologia , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Ratos , Ratos Endogâmicos , Valores de Referência , Espironolactona/análogos & derivados , Espironolactona/farmacologia
11.
J Cereb Blood Flow Metab ; 4(1): 107-9, 1984 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-6363432

RESUMO

The effect of haemorrhagic hypotension on the levels of prostaglandin E2 (PGE2), thromboxane B2 (TXB2), and 6-keto prostaglandin F1 alpha (6-keto-PGF1 alpha) in cortical tissue of rats was studied. Lightly anesthetized rats were subjected to steady-state hypotension for 15 min, with a mean arterial blood pressure of 80, 60, and 40 mm Hg, and compared to a control group of normotensive rats. No significant change was found in the levels of PGE2 and TXB2. The level of 6-keto-PGF1 alpha increased from 7.8 +/- 0.9 to 14.1 +/- 1.9 pg/mg protein (p less than 0.02) at 80 mm Hg. Our findings suggest that prostacyclin, which is a potent vasodilator, might play a role in setting the lower limit of the autoregulation range.


Assuntos
Encéfalo/metabolismo , Epoprostenol/metabolismo , Epoprostenol/fisiologia , Hemorragia/fisiopatologia , Hipotensão/fisiopatologia , 6-Cetoprostaglandina F1 alfa/metabolismo , Animais , Gasometria , Dinoprostona , Homeostase , Masculino , Prostaglandinas E/metabolismo , Ratos , Tromboxano B2/metabolismo
12.
J Cereb Blood Flow Metab ; 7(1): 58-63, 1987 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-3468118

RESUMO

Head injury was induced in the left hemisphere of rats. The rats were killed at various time intervals after trauma (immediately, 15 min, 1 and 18 h, and 4 and 10 days), and the rates of synthesis and release of prostaglandin PGE2, 6-keto-PGF1 alpha, and thromboxane TXB2 from cortical slices of both hemispheres were studied. The rate of synthesis of PGE2 after 18 h was six and four times higher than control in the contused and contralateral hemispheres, respectively. By 10 days post-trauma, both hemispheres had normal rate of PGE2 release. TXB2 and 6-keto-PGF1 alpha synthetases were affected already 15 min after the injury, and a similarly elevated rate of synthesis was found in both hemispheres. The maximal effect was detected after 1 or 18 h with return to normal after 4 or 10 days for TXB2 and 6-keto-PGF1 alpha, respectively. Tissue specific gravity was determined for both hemispheres using linear gradient columns. The results of these determinations indicate that development of edema occurs in the contused hemisphere as early as 15 min post trauma; it reaches its maximal level at 18 h and returns to normal at 10 days. Arterial pressure was monitored, and a transient increase was found at 10 min post trauma. We suggest that the production of edema after brain injury may be related to the increased rate of PGE2 and PGI2 synthesis, which occurs at similar time intervals after injury.


Assuntos
Encéfalo/metabolismo , Traumatismos Craniocerebrais/metabolismo , Prostaglandinas/biossíntese , 6-Cetoprostaglandina F1 alfa/biossíntese , Animais , Pressão Sanguínea , Edema Encefálico/etiologia , Dinoprostona , Masculino , Prostaglandinas E/biossíntese , Ratos , Tromboxano B2/biossíntese
13.
J Cereb Blood Flow Metab ; 14(4): 615-9, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8014208

RESUMO

In a model of closed head injury (CHI) in the rat we have shown the activation of phospholipase A2 and the production of eicosanoids after injury: at 15 min, mainly 5-hydroxyeicosatetraenoic acid (5-HETE), and at 24 h, mainly prostaglandin E2. The present study was designed to test whether CHI can also trigger the production of cytokines in the brain. CHI was induced in ether-anesthesized rats by a weight-drop device falling over the exposed skull covering the left hemisphere, 1-2 mm lateral to the midline in the midcoronal plane. In the posttraumatic period (1-24 h), the rats were decapitated, cortical tissue from the injured zone of the contused and contralateral hemispheres was removed and sonicated, and cytokine activity was assessed. Whereas no tumor necrosis factor alpha (TNF alpha) activity was found in normal brain tissue, it was detectable in the contused hemisphere (approximately of 72 +/- 50 pg/mg protein) as early as 1 h post-CHI. TNF alpha levels increased at 2 h, peaked at 4 h, (approximately of 609 +/- 540 pg/mg protein), and declined thereafter. At parallel intervals, only low levels of TNF alpha were detected in the contralateral hemisphere. In normal brain, interleukin-6 (IL-6) was nondetectable. Following CHI, high levels of IL-6 were present, although their accumulation lagged behind that of TNF alpha by 2-4 h, peaking at 8 h (62 +/- 31 ng/mg protein). We suggest that the rapid production of TNF alpha and IL-6 following CHI is a local inflammatory response of brain tissue to primary insult.


Assuntos
Encéfalo/metabolismo , Traumatismos Craniocerebrais/metabolismo , Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Masculino , Concentração Osmolar , Ratos , Ratos Endogâmicos , Fatores de Tempo , Distribuição Tecidual
14.
J Cereb Blood Flow Metab ; 17(3): 273-9, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9119900

RESUMO

Reactive oxygen species (ROS) are normally generated in the brain during metabolism, and their production is enhanced by various insults. Low molecular weight antioxidants (LMWA) are one of the defense mechanisms of the living cell against ROS. The reducing capacity of brain tissue (total LMWA) was measured by cyclic voltammetry (CV), which records biological oxidation potential specific to the type of scavenger(s) present and anodic current intensity (Ia), which depends on scavenger concentration. In the present study, the reducing capacity of rat brain following closed head injury (CHI) was measured. In addition, CV of heat-acclimated traumatized rats was used to correlate endogenous cerebroprotection after CHI with LMWA activity. Sham-injured rat brains displayed two anodic potentials: at 350 +/- 50 mV (Ia = 0.75 +/- 0.06 microA/mg protein) and at 750 +/- 50 mV (Ia = 1.00 +/- 0.05 microA/mg protein). Following CHI, the anodic waves appeared at the same potentials as in the sham animals. However, within 5 min of CHI, the total reducing capacity was transiently decreased by 40% (p < 0.01). A second dip was detected at 24 h (60%, p < 0.005). By 48 h and at 7 days, the Ia levels normalized. The acclimated rats displayed anodic potentials identical to those of normothermic rats. However, the Ia of both potentials was lower (60% of control, p < 0.001). The Ia profile after CHI was the direct opposite of the normothermic Ia profile: no immediate decrease of Ia and an increase from 4 h and up to 7 days (40-50%, p < 0.001). We suggest that the lowered levels of LMWA in the post-CHI period reflect their consumption due to overproduction of free radicals. The augmented concentration of LMWA found in the brain of the heat-acclimated rats suggests that these rats are better able to cope with these harmful radicals, resulting in a more favorable outcome following CHI.


Assuntos
Aclimatação/fisiologia , Antioxidantes/metabolismo , Lesões Encefálicas/metabolismo , Temperatura Alta , Espécies Reativas de Oxigênio/metabolismo , Animais , Ácido Ascórbico/farmacologia , Química Encefálica/efeitos dos fármacos , Lesões Encefálicas/fisiopatologia , Traumatismos Craniocerebrais/metabolismo , Suscetibilidade a Doenças , Condutividade Elétrica , Sequestradores de Radicais Livres , Coração/fisiopatologia , Masculino , Miocárdio/metabolismo , Oxirredução , Estresse Oxidativo , Ratos , Triptofano/farmacologia
15.
J Cereb Blood Flow Metab ; 16(3): 378-84, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8621742

RESUMO

We recently demonstrated that closed head injury (CHI) in the rat triggers the production of tumor necrosis factor alpha (TNFalpha) in the contused hemisphere. Other investigations have shown that this cytokine plays a role in the inflammatory response following trauma. The present study was designed to determine whether inhibition of TNFalpha production or activity affects the development of cerebral edema as well as neurological dysfunction and hippocampal cell loss after CHI. To this end, we used two pharmacological agents, each acting via a different mechanism: pentoxifylline (PTX), which attenuates the production of TNFalpha, and tumor necrosis factor binding protein (TBP), a physiological inhibitor of TNFalpha activity. Both agents significantly lessened peak edema formation at 24 h and facilitated the recovery of motor function for < or = 4 days postinjury. In addition, TBP attenuated disruption of the blood-brain barrier and protected hippocampal cells. PTX significantly lowered the brain TNFalpha level (by approximately 80%), and TBP completely abolished the activity of recombinant human TNF when they were added at the same time in the in vitro bioassay. We suggest, therefore, that a decrease in TNFalpha level or the inhibition of its activity is accompanied by reduced brain damage.


Assuntos
Encéfalo/metabolismo , Proteínas de Transporte/farmacologia , Traumatismos Cranianos Fechados/metabolismo , Fármacos Neuroprotetores/farmacologia , Pentoxifilina/farmacologia , Receptores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Edema Encefálico/prevenção & controle , Sobrevivência Celular/efeitos dos fármacos , Traumatismos Cranianos Fechados/patologia , Humanos , Masculino , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Receptores Tipo I de Fatores de Necrose Tumoral , Proteínas Recombinantes , Receptores Chamariz do Fator de Necrose Tumoral
16.
J Cereb Blood Flow Metab ; 17(10): 1007-19, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9346425

RESUMO

It has been suggested that reactive oxygen species (ROS) play a role in the pathophysiology of brain damage. A number of therapeutic approaches, based on scavenging these radicals, have been attempted both in experimental models and in the clinical setting. In an experimental rat and mouse model of closed-head injury (CHI), we have studied the total tissue nonenzymatic antioxidant capacity to combat ROS. A major mechanism for neutralizing ROS uses endogenous low-molecular weight antioxidants (LMWA). This review deals with the source and nature of ROS in the brain, along with the endogenous defense mechanisms that fight ROS. Special emphasis is placed on LMWA such as ascorbate, urate, tocopherol, lipoic acid, and histidine-related compounds. A novel electrochemical method, using cyclic voltammetry for the determination of total tissue LMWA, is described. The temporal changes in brain LMWA after CHI, as part of the response of the tissue to high ROS levels, and the correlation between the ability of the brain to elevate LMWA and clinical outcome are addressed. We relate to the beneficial effects observed in heat-acclimated rats and the detrimental effects of injury found in apolipoprotein E-deficient mice. Finally, we summarize the effects of cerebroprotective pharmacological agents including the iron chelator desferal, superoxide dismutase, a stable radical from the nitroxide family, and HU-211, a nonpsychotoropic cannabinoid with antioxidant properties. We conclude that ROS play a key role in the pathophysiology of brain injury, and that their neutralization by endogenous or exogenous antioxidants has a protective effect. It is suggested, therefore, that the brain responds to ROS by increasing LMWA, and that the degree of this response is correlated with clinical recovery. The greater the response, the more favorable the outcome.


Assuntos
Antioxidantes/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Estresse Oxidativo , Animais , Humanos , Peso Molecular , Prognóstico , Espécies Reativas de Oxigênio/metabolismo
17.
J Cereb Blood Flow Metab ; 8(3): 395-402, 1988 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3366801

RESUMO

Head trauma was induced in rats by a weight-drop device, falling over the exposed skull over the left hemisphere. The neurological state of the rats was evaluated by a neurological severity score at 1 h and 18 h post head trauma. At 18 h post head trauma, rats were decapitated and tissue from the vicinity of the injury and from a corresponding area in the contralateral hemisphere was taken for specific gravity (SG) determination using linear gradient columns. Slices were taken from the same sites for incubation in Krebs-Ringer solution, and the concentrations of prostaglandin (PG)E2, 6-keto-PGF1 alpha, and thromboxane B2 accumulated in the medium during 1 h were measured by radioimmunoassay. In one experimental group, rats were pretreated with intraperitoneal dexamethasone sodium phosphate (4 mg/kg) 18 and 2 h before head trauma, and a third dose was given 8 h post head trauma. Another group was treated with intraperitoneal indomethacin (10 mg/kg) 1 h before and 7 h after head trauma. Other groups were treated immediately and 8 h after head trauma with 4, 8, 15, or 30 mg/kg of dexamethasone sodium phosphate. Another group of rats was treated with free dexamethasone (10 mg/kg) right after head trauma and 8 h later. Head trauma induced edema, as expressed by decreased SG, in the left hemisphere of all traumatized rats. Neither treatment protocol affected the neurological severity score of the injured rats or the SG of the contused hemisphere. PG synthesis, on the other hand, was significantly reduced following indomethacin or free dexamethasone, both in sham and traumatized rats, but not in dexamethasone sodium phosphate-treated rats. We conclude that pretreatment with indomethacin, dexamethasone sodium phosphate, or dexamethasone, used in the present protocols, does not affect posttraumatic cerebral edema. Thus, the role of PGs as mediators of edema formation remains unclear.


Assuntos
Edema Encefálico/etiologia , Traumatismos Craniocerebrais/complicações , Dexametasona/farmacologia , Indometacina/farmacologia , Animais , Pressão Sanguínea , Encéfalo/patologia , Edema Encefálico/patologia , Traumatismos Craniocerebrais/fisiopatologia , Dexametasona/análogos & derivados , Masculino , Sistema Nervoso/fisiopatologia , Ratos , Ratos Endogâmicos , Índice de Gravidade de Doença , Gravidade Específica
18.
J Cereb Blood Flow Metab ; 2(4): 429-38, 1982 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7142307

RESUMO

The effects of hypoxaemia on regional cerebral blood flow (CBF) and brain cortical metabolite concentrations were investigated at different blood glucose concentrations in rats under nitrous oxide anaesthesia. Tissue hypoxia of 15-min duration was induced by a combination of arterial hypoxaemia, hypotension, and clamping of the right carotid artery. Blood glucose concentrations were manipulated by varying the food intake in the 24 h before the experiment, and by glucose administration. Cortical CBF doubled during hypoxia on the intact side, but did not differ significantly from control values on the clamped side. In the clamped hemisphere there was a substantial decrease in adenylate energy charge. At brain tissue glucose concentration of 1 mumol g-1 and above, there was an inverse correlation between adenylate energy charge and brain lactate concentration. In starved animals with mean brain glucose of 0.32 +/- 0.00 mumol g-1, lactate concentration was significantly lower, in spite of equally severe disruption of energy state. Recovery of brain adenylate energy charge was worse in fed and glucose-infused groups than in the fasted group. These results demonstrate that limitation of substrate supply during severe hypoxia in the rat allows enhanced recovery of brain energy metabolism following the hypoxic episode.


Assuntos
Glicemia/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Hipóxia Encefálica/metabolismo , Lactatos/metabolismo , Animais , Circulação Cerebrovascular , Ácido Láctico , Masculino , Ratos , Ratos Endogâmicos , Fluxo Sanguíneo Regional
19.
J Cereb Blood Flow Metab ; 20(2): 369-80, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10698075

RESUMO

Cytokines are important mediators of intracranial inflammation following traumatic brain injury (TBI). In the present study, the neurological impairment and mortality, blood-brain barrier (BBB) function, intracranial polymorphonuclear leukocyte (PMN) accumulation, and posttraumatic neuronal cell death were monitored in mice lacking the genes for tumor necrosis factor (TNF)/lymphotoxin-alpha (LT-alpha) (TNF/LT-alpha-/-) and interleukin-6 (IL-6) and in wild-type (WT) littermates subjected to experimental closed head injury (total n = 107). The posttraumatic mortality was significantly increased in TNF/LT-alpha-/- mice (40%; P < 0.02) compared with WT animals (10%). The IL-6-/- mice also showed a higher mortality (17%) than their WT littermates (5.6%), but the difference was not statistically significant (P > 0.05). The neurological severity score was similar among all groups from 1 to 72 hours after trauma, whereas at 7 days, the TNF/LT-alpha-/- mice showed a tendency toward better neurological recovery than their WT littermates. Interestingly, neither the degree of BBB dysfunction nor the number of infiltrating PMNs in the injured hemisphere was different between WT and cytokine-deficient mice. Furthermore, the analysis of brain sections by in situ DNA nick end labeling (TUNEL histochemistry) at 24 hours and 7 days after head injury revealed a similar extent of posttraumatic intracranial cell death in all animals. These results show that the pathophysiological sequelae of TBI are not significantly altered in mice lacking the genes for the proinflammatory cytokines TNF, LT-alpha, and IL-6. Nevertheless, the increased posttraumatic mortality in TNF/LT-alpha-deficient mice suggests a protective effect of these cytokines by mechanisms that have not been elucidated yet.


Assuntos
Barreira Hematoencefálica/fisiologia , Citocinas/genética , Traumatismos Cranianos Fechados/imunologia , Traumatismos Cranianos Fechados/fisiopatologia , Neutrófilos/imunologia , Animais , Morte Celular , Modelos Animais de Doenças , Traumatismos Cranianos Fechados/mortalidade , Marcação In Situ das Extremidades Cortadas , Interleucina-6/genética , Linfotoxina-alfa/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Exame Neurológico , Neurônios/citologia , Fator de Necrose Tumoral alfa/genética
20.
Free Radic Biol Med ; 24(2): 332-40, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9433909

RESUMO

Reactive oxygen-derived species were previously implicated in mediation of post-traumatic brain damage; however, the efficacy of traditional antioxidants in preventing/reversing the damage is sometimes limited. The present work focused on the mechanisms underlying the neuroprotective activity of cell permeable, nontoxic, antioxidants, namely stable nitroxide radicals in an experimental model of rat closed-head injury. Brain damage was induced by the weight-drop method and the clinical status was evaluated according to a neurological severity score at 1 h and 24 h, where the difference between these scores reflects the extent of recovery. The metal chelator deferoxamine as well as three nitroxide derivatives, differing in hydrophilicity and charge, and one hydroxylamine (a reduced nitroxide) facilitated the clinical recovery and decreased the brain edema. The nitroxides, but neither the hydroxylamine nor deferoxamine, protected the integrity of the blood-brain barrier. Superoxide dismutase also improved the clinical recovery but did not affect brain edema or the blood-brain barrier. The results suggest that by switching back and forth between themselves, the nitroxide and hydroxylamine act catalytically as self-replenishing antioxidants, and protect brain tissue by terminating radical-chain reactions, oxidizing deleterious metal ions, and by removal of intracellular superoxide.


Assuntos
Antioxidantes/uso terapêutico , Encefalopatias/prevenção & controle , Traumatismos Cranianos Fechados/complicações , Fármacos Neuroprotetores/uso terapêutico , Óxidos de Nitrogênio/uso terapêutico , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Encefalopatias/etiologia , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Desferroxamina/uso terapêutico , Hidroxilamina/uso terapêutico , Masculino , Piperidinas/farmacologia , Ratos , Superóxido Dismutase/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA